101
|
Wang H, Li J, Gao Y, Xu Y, Pan Y, Tsuji I, Sun ZJ, Li XM. Xeno-oestrogens and phyto-oestrogens are alternative ligands for the androgen receptor. Asian J Androl 2010; 12:535-47. [PMID: 20436506 DOI: 10.1038/aja.2010.14] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The androgen receptor (AR) plays a critical role in prostate cancer development and progression. This study aimed to use a computerized docking approach to examine the interactions between the human AR and phyto-oestrogens (genistein, daidzein, and flavone) and xeno-oestrogens (bisphenol A, 4-nonylphenol, dichlorodiphenyl trichloroethane [DDT], diethylstilbestrol [DES]). The predicted three-dimensional structure of AR and androgens was established using X-ray diffraction. The binding of four xeno-oestrogens and three phyto-oestrogens to AR was analysed. The steroids estradiol and dihydrotestosterone (DHT) were used as positive controls and thyroxine as negative control. All the ligands shared the same binding site except for thyroxine. The endogenous hormones DHT and 17beta-oestradiol showed the strongest binding with the lowest affinity energy (< -10 kcal mol(-1)). All three phyto-oestrogens and two xeno-oestrogens (bisphenol A and DES) showed strong binding to AR. The affinities of flavone, genistein, and daidzein were between -8.8 and -8.5 kcal mol(-1), while that of bisphenol A was -8.1 kcal mol(-1) and DES -8.3 kcal mol(-1). Another two xeno-oestrogens, 4-nonylphenol and DDT, although they fit within the binding domain of AR, showed weak affinity (-6.4 and -6.7 kcal mol(-1), respectively). The phyto-oestrogens genistein, daidzein and flavone, and the xeno-oestrogens bisphenol A and DES can be regarded as androgenic effectors. The xeno-oestrogens DDT and 4-nonylphenol bind only weakly to AR.
Collapse
Affiliation(s)
- Hao Wang
- School of Life Sciences, Northeast Normal University, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Donner N, Handa RJ. Estrogen receptor beta regulates the expression of tryptophan-hydroxylase 2 mRNA within serotonergic neurons of the rat dorsal raphe nuclei. Neuroscience 2009; 163:705-18. [PMID: 19559077 PMCID: PMC2740745 DOI: 10.1016/j.neuroscience.2009.06.046] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/18/2009] [Accepted: 06/18/2009] [Indexed: 10/20/2022]
Abstract
Dysfunctions of the brain 5-HT system are often associated with affective disorders, such as depression. The raphe nuclei target the limbic system and most forebrain areas and constitute the main source of 5-HT in the brain. All 5-HT neurons express tryptophan hydroxylase-2 (TPH2), the brain specific, rate-limiting enzyme for 5-HT synthesis. Estrogen receptor (ER) beta agonists have been shown to attenuate anxiety- and despair-like behaviors in rodent models. Therefore, we tested the hypothesis that ER beta may contribute to the regulation of gene expression in 5-HT neurons of the dorsal raphe nuclei (DRN) by examining the effects of systemic and local application of the selective ER beta agonist diarylpropionitrile (DPN) on tph2 mRNA expression. Ovariectomized (OVX) female rats were injected s.c. with DPN or vehicle once daily for 8 days. In situ hybridization revealed that systemic DPN-treatment elevated basal tph2 mRNA expression in the caudal and mid-dorsal DRN. Behavioral testing of all animals in the open field (OF) and on the elevated plus maze (EPM) on days 6 and 7 of treatment confirmed the anxiolytic nature of ER beta activation. Another cohort of female OVX rats was stereotaxically implanted bilaterally with hormone-containing wax pellets flanking the DRN. Pellets contained 17-beta-estradiol (E), DPN, or no hormone. Both DPN and E significantly enhanced tph2 mRNA expression in the mid-dorsal DRN. DPN also increased tph2 mRNA in the caudal DRN. DPN- and E-treated rats displayed a more active stress-coping behavior in the forced-swim test (FST). No behavioral differences were found in the OF or on the EPM. These data indicate that ER beta acts at the level of the rat DRN to modulate tph2 mRNA expression and thereby influence 5-HT synthesis in DRN subregions. Our results also suggest that local activation of ER beta neurons in the DRN may be sufficient to decrease despair-like behavior, but not anxiolytic behaviors.
Collapse
Affiliation(s)
- N Donner
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80526, USA
| | | |
Collapse
|
103
|
Jeng YJ, Watson CS. Proliferative and anti-proliferative effects of dietary levels of phytoestrogens in rat pituitary GH3/B6/F10 cells - the involvement of rapidly activated kinases and caspases. BMC Cancer 2009; 9:334. [PMID: 19765307 PMCID: PMC2755011 DOI: 10.1186/1471-2407-9-334] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 09/18/2009] [Indexed: 12/18/2022] Open
Abstract
Background Phytoestogens are a group of lipophillic plant compounds that can have estrogenic effects in animals; both tumorigenic and anti-tumorigenic effects have been reported. Prolactin-secreting adenomas are the most prevalent form of pituitary tumors in humans and have been linked to estrogen exposures. We examined the proliferative effects of phytoestrogens on a rat pituitary tumor cell line, GH3/B6/F10, originally subcloned from GH3 cells based on its ability to express high levels of the membrane estrogen receptor-α. Methods We measured the proliferative effects of these phytoestrogens using crystal violet staining, the activation of several mitogen-activated protein kinases (MAPKs) and their downstream targets via a quantitative plate immunoassay, and caspase enzymatic activities. Results Four phytoestrogens (coumestrol, daidzein, genistein, and trans-resveratrol) were studied over wide concentration ranges. Except trans-resveratrol, all phytoestrogens increased GH3/B6/F10 cell proliferation at some concentration relevant to dietary levels. All four phytoestrogens attenuated the proliferative effects of estradiol when administered simultaneously. All phytoestrogens elicited MAPK and downstream target activations, but with time course patterns that often differed from that of estradiol and each other. Using selective antagonists, we determined that MAPKs play a role in the ability of these phytoestrogens to elicit these responses. In addition, except for trans-resveratrol, a serum removal-induced extrinsic apoptotic pathway was blocked by these phytoestrogens. Conclusion Phytoestrogens can block physiological estrogen-induced tumor cell growth in vitro and can also stimulate growth at high dietary concentrations in the absence of endogenous estrogens; these actions are correlated with slightly different signaling response patterns. Consumption of these compounds should be considered in strategies to control endocrine tumor cell growth, such as in the pituitary.
Collapse
Affiliation(s)
- Yow-Jiun Jeng
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| | | |
Collapse
|
104
|
Nguyen TVV, Yao M, Pike CJ. Dihydrotestosterone activates CREB signaling in cultured hippocampal neurons. Brain Res 2009; 1298:1-12. [PMID: 19729001 DOI: 10.1016/j.brainres.2009.08.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 08/20/2009] [Accepted: 08/24/2009] [Indexed: 12/17/2022]
Abstract
Although androgens induce numerous actions in brain, relatively little is known about which cell signaling pathways androgens activate in neurons. Recent work in our laboratory showed that the androgens testosterone and dihydrotestosterone (DHT) activate androgen receptor (AR)-dependent mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling. Since the transcription factor cyclic AMP response element binding protein (CREB) is a downstream effector of MAPK/ERK and androgens activate CREB in non-neuronal cells, we investigated whether androgens activate CREB signaling in neurons. First, we observed that DHT rapidly activates CREB in cultured hippocampal neurons, as evidenced by CREB phosphorylation. Further, we observed that DHT-induced CREB phosphorylation is AR-dependent, as it occurs in PC12 cells stably transfected with AR but in neither wild-type nor empty vector-transfected cells. Next, we sought to identify the signal transduction pathways upstream of CREB phosphorylation using pharmacological inhibitors. DHT-induced CREB phosphorylation in neurons was found to be dependent upon protein kinase C (PKC) signaling but independent of MAPK/ERK, phosphatidylinositol 3-kinase, protein kinase A, and Ca(2+)/calmodulin-dependent protein kinase IV. These results demonstrate that DHT induces PKC-dependent CREB signaling, which may contribute to androgen-mediated neural functions.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | | | | |
Collapse
|
105
|
Xu C, He J, Jiang H, Zu L, Zhai W, Pu S, Xu G. Direct effect of glucocorticoids on lipolysis in adipocytes. Mol Endocrinol 2009; 23:1161-70. [PMID: 19443609 PMCID: PMC5419195 DOI: 10.1210/me.2008-0464] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 05/05/2009] [Indexed: 12/27/2022] Open
Abstract
Hypercortisolemia and glucocorticoid treatment cause elevated level of circulating free fatty acids (FFAs). The basis of this phenomenon has long been linked to the effect of glucocorticoids permitting and enhancing the adipose lipolysis response to various hormones. In this study, we demonstrate that glucocorticoids directly stimulate lipolysis in rat primary adipocytes in a dose- and time-responsive manner; this lipolytic action was attenuated by treatment with the glucocorticoid antagonist RU486. Dexamethasone down-regulates mRNA and protein levels of cyclic-nucleotide phosphodiesterase 3B, thereby elevating cellular cAMP production and activating protein kinase A (PKA). On inhibition of PKA but not other kinases, the lipolysis response ceases. Furthermore, dexamethasone induces phosphorylation and down-regulation of perilipin, a lipid droplet-associating protein that modulates lipolysis; this effect is restored by RU486 or PKA inhibitor H89. Dexamethasone up-regulates mRNA and protein levels of hormone-sensitive lipase (HSL) and adipose triglyceride lipase; these effects, parallel to increased lipolysis, are attenuated by RU486 or actinomycin D. Phosphorylation at Ser-563 and Ser-660 residues of HSL and activity of cellular lipases are elevated on dexamethasone stimulation but abrogated by the coaddition of H89. However, dexamethasone does not induce HSL translocation to the lipid droplet surface in differentiated adipocytes. We show that elevated FFA concentration in plasma is associated with increased lipase activity and lipolysis in vivo in adipose tissues of dexamethasone-treated rats. Therefore, the lipolytic action of glucocorticoids liberates FFA efflux from adipocytes to the bloodstream, which could be a cellular basis of systemic FFA elevation in response to glucocorticoid challenge.
Collapse
Affiliation(s)
- Chong Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, the Ministry of Education of China, Beijing 100191, China.
| | | | | | | | | | | | | |
Collapse
|
106
|
Nierth-Simpson EN, Martin MM, Chiang TC, Melnik LI, Rhodes LV, Muir SE, Burow ME, McLachlan JA. Human uterine smooth muscle and leiomyoma cells differ in their rapid 17beta-estradiol signaling: implications for proliferation. Endocrinology 2009; 150:2436-45. [PMID: 19179429 PMCID: PMC2671893 DOI: 10.1210/en.2008-0224] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Uterine leiomyomas, benign uterine smooth muscle tumors that affect 30% of reproductive-aged women, are a significant health concern. The initiation event for these tumors is unclear, but 17beta-estradiol (E2) is an established promoter of leiomyoma growth. E2 not only alters transcription of E2-regulated genes but also can rapidly activate signaling pathways. The aim of our study is to investigate the role of rapid E2-activated cytoplasmic signaling events in the promotion of leiomyomas. Western blot analysis revealed that E2 rapidly increases levels of phosphorylated protein kinase C alpha (PKC alpha) in both immortalized uterine smooth muscle (UtSM) and leiomyoma (UtLM) cell lines, but increases levels of phosphorylated ERK1/2 only in UtLM cells. Our studies demonstrate a paradoxical effect of molecular and pharmacological inhibition of PKC alpha on ERK1/2 activation and cellular proliferation in UtLM and UtSM cells. PKC alpha inhibition decreases levels of phosphorylated ERK1/2 and proliferation in UtLM cells but raises these levels in UtSM cells. cAMP-PKA signaling is rapidly activated only in UtSM cells with E2 and inhibits ERK1/2 activation and proliferation. We therefore propose a model whereby E2's rapid activation of PKC alpha and cAMP-PKA signaling plays a central role in the maintenance of a low proliferative index in normal uterine smooth muscle via its inhibition of the MAPK cascade and these pathways are altered in leiomyomas to promote MAPK activation and proliferation. These studies demonstrate that rapid E2-signaling pathways contribute to the promotion of leiomyomas.
Collapse
|
107
|
Jeng YJ, Kochukov MY, Watson CS. Membrane estrogen receptor-alpha-mediated nongenomic actions of phytoestrogens in GH3/B6/F10 pituitary tumor cells. J Mol Signal 2009; 4:2. [PMID: 19400946 PMCID: PMC2679742 DOI: 10.1186/1750-2187-4-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 04/28/2009] [Indexed: 01/08/2023] Open
Abstract
Background Estradiol (E2) mediates various intracellular signaling cascades from the plasma membrane via several estrogen receptors (ERs). The pituitary is an estrogen-responsive tissue, and we have previously reported that E2 can activate mitogen-activated protein kinases (MAPKs) such as ERK1/2 and JNK1/2/3 in the membrane ERα (mERα)-enriched GH3/B6/F10 rat pituitary tumor cell line. Phytoestrogens are compounds found in plants and foods such as soybeans, alfalfa sprouts, and red grapes. They are structurally similar to E2 and share a similar mechanism of action through their binding to ERs. Phytoestrogens bind to nuclear ERs with a much lower affinity and therefore are less potent in mediating genomic responses. However, little is known about their ability to act via mERs to mediate nongenomic effects. Methods To investigate the activation of different nongenomic pathways, and determine the involvement of mERα, we measured prolactin (PRL) release by radio-immunoassay, MAPK activations (ERK1/2 and JNK1/2/3) via a quantitative plate immunoassay, and intracellular [Ca2+] by Fura-2 fluorescence imaging in cells treated with E2 or four different phytoestrogens (coumestrol, daidzein, genistein, and trans-resveratrol). Results Coumesterol and daidzein increased PRL release similar to E2 in GH3/B6/F10 cells, while genistein and trans-resveratrol had no effect. All of these compounds except genistein activated ERK1/2 signaling at 1–10 picomolar concentrations; JNK 1/2/3 was activated by all compounds at a 100 nanomolar concentration. All compounds also caused rapid Ca2+ uptake, though in unique dose-dependent Ca2+ response patterns for several aspects of this response. A subclone of GH3 cells expressing low levels of mERα (GH3/B6/D9) did not respond to any phytoestrogen treatments for any of these responses, suggesting that these nongenomic effects were mediated via mERα. Conclusion Phytoestrogens were much more potent in mediating these nongenomic responses (activation of MAPKs, PRL release, and increased intracellular [Ca2+]) via mERα than was previously reported for genomic responses. The unique non-monotonic dose responses and variant signaling patterns caused by E2 and all tested phytoestrogens suggest that complex and multiple signaling pathways or binding partners could be involved. By activating these different nongenomic signaling pathways, phytoestrogens could have significant physiological consequences for pituitary cell functions.
Collapse
Affiliation(s)
- Yow-Jiun Jeng
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| | | | | |
Collapse
|
108
|
Keay J, Thornton JW. Hormone-activated estrogen receptors in annelid invertebrates: implications for evolution and endocrine disruption. Endocrinology 2009; 150:1731-8. [PMID: 19036877 PMCID: PMC2659264 DOI: 10.1210/en.2008-1338] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 11/18/2008] [Indexed: 12/30/2022]
Abstract
As the primary mediators of estrogen signaling in vertebrates, estrogen receptors (ERs) play crucial roles in reproduction, development, and behavior. They are also the major mediators of endocrine disruption by xenobiotic pollutants that mimic or block estrogen action. ERs that are sensitive to estrogen and endocrine disrupters have long been thought to be restricted to vertebrates: although there is evidence for estrogen signaling in invertebrates, the only ERs studied to date, from mollusks and cephalochordates, have been insensitive to estrogen and therefore incapable of mediating estrogen signaling or disruption. To determine whether estrogen sensitivity is ancestral or a unique characteristic of vertebrate ERs, we isolated and characterized ERs from two annelids, Platynereis dumerilii and Capitella capitata, because annelids are the sister phylum to mollusks and have been shown to produce and respond to estrogens. Functional assays show that annelid ERs specifically activate transcription in response to low estrogen concentrations and bind estrogen with high affinity. Furthermore, numerous known endocrine-disrupting chemicals activate or antagonize the annelid ER. This is the first report of a hormone-activated invertebrate ER. Our results indicate that estrogen signaling via the ER is as ancient as the ancestral bilaterian animal and corroborate the estrogen sensitivity of the ancestral steroid receptor. They suggest that the taxonomic scope of endocrine disruption by xenoestrogens may be very broad and reveal how functional diversity evolved in a gene family central to animal endocrinology.
Collapse
Affiliation(s)
- June Keay
- Center for Ecology and Evolutionary Biology, University of Oregon, Eugene, Oregon 97403, USA
| | | |
Collapse
|
109
|
Roepke TA, Qiu J, Bosch MA, Rønnekleiv OK, Kelly MJ. Cross-talk between membrane-initiated and nuclear-initiated oestrogen signalling in the hypothalamus. J Neuroendocrinol 2009; 21:263-70. [PMID: 19187465 PMCID: PMC2796511 DOI: 10.1111/j.1365-2826.2009.01846.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It is increasingly evident that 17beta-oestradiol (E(2)), via a distinct membrane oestrogen receptor (Gq-mER), can rapidly activate kinase pathways to have multiple downstream actions in central nervous system (CNS) neurones. We have found that E(2) can rapidly reduce the potency of the GABA(B) receptor agonist baclofen and mu-opioid receptor agonist DAMGO to activate G-protein-coupled, inwardly rectifying K(+) (GIRK) channels in hypothalamic neurones, thereby increasing the excitability (firing activity) of pro-opiomelanocortin (POMC) and dopamine neurones. These effects are mimicked by the membrane impermeant E(2)-BSA and a new ligand (STX) that is selective for the Gq-mER that does not bind to ERalpha or ERbeta. Both E(2) and STX are fully efficacious in attenuating the GABA(B) response in ERalpha, ERbeta and GPR 30 knockout mice in an ICI 182 780 reversible manner. These findings are further proof that E(2) signals through a unique plasma membrane ER. We have characterised the coupling of this Gq-mER to a Gq-mediated activation of phospholipase C leading to the up-regulation of protein kinase Cdelta and protein kinase A activity in these neurones, which ultimately alters gene transcription. Finally, as proof of principle, we have found that STX, similar to E(2), reduces food intake and body weight gain in ovariectomised females. STX, presumably via the Gq-mER, also regulates gene expression of a number of relevant targets including cation channels and signalling molecules that are critical for regulating (as a prime example) POMC neuronal excitability. Therefore, E(2) can activate multiple receptor-mediated pathways to modulate excitability and gene transcription in CNS neurones that are critical for controlling homeostasis and motivated behaviors.
Collapse
Affiliation(s)
- Troy A. Roepke
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
| | - Jian Qiu
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
| | - Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
- Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
110
|
Li M, Schatten H, Sun QY. Androgen receptor's destiny in mammalian oocytes: a new hypothesis. Mol Hum Reprod 2009; 15:149-54. [PMID: 19193657 DOI: 10.1093/molehr/gap006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Unlike the well-established roles of androgen and androgen receptor (AR) in males, the functions of this steroid and its receptor in the ovary are still unclear. For decades, androgen and AR have long been considered to play a negative (at least not a positive) role in mammalian oocyte maturation. However, recent studies by us and others showed their positive influence in promoting meiotic maturation. On the other hand, rapid non-genomic effects of androgens have been observed and are now generally accepted as contributing to the physiological effects of the steroids and their related receptors in somatic cells, and this has stimulated us to explore the complex roles of AR in the ovary. Based on the classic dogma and new findings, we collected evidence to propose that the expression of AR shifts from the oocytes to the theca cells and finally disappears in the oocytes during evolution. It is suggested that the non-genomic pathway involving androgen and AR in the mammalian oocytes, unlike somatic cells, cells will undergo elimination. The function of androgen and AR in promoting meiotic maturation may have been replaced gradually by gonadotrophins. Moreover, a possible relationship between AR and polycystic ovary syndrome is also discussed, which might provide a clue for the pathology of the disease.
Collapse
Affiliation(s)
- Mo Li
- Institute of Zoology, Chinese Academy of Sciences, Chaoyang, Beijing, People's Republic of China
| | | | | |
Collapse
|
111
|
Kayahara M, Ohanian J, Ohanian V, Berry A, Vadlamudi R, Ray DW. MNAR functionally interacts with both NH2- and COOH-terminal GR domains to modulate transactivation. Am J Physiol Endocrinol Metab 2008; 295:E1047-55. [PMID: 18682536 PMCID: PMC2584814 DOI: 10.1152/ajpendo.90429.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucocorticoids are potent anti-inflammatory agents, acting through the glucocorticoid receptor (GR) to regulate target gene transcription. However, GR may also exert acute effects, including activation of signaling kinases such as c-Src and protein kinase B, possibly via the scaffold protein, modulator of nongenomic action of the estrogen receptor (MNAR). MNAR inhibited GR transactivation in A549 cells, but in HEK293 cells there was a ligand concentration-dependent biphasic effect. Transactivation driven by low ligand concentrations was inhibited by MNAR expression, whereas higher ligand concentrations were potentiating. Further analysis revealed that MNAR inhibited transactivation by the ligand-independent activation function (AF)1 but potentiated the COOH-terminal AF2 domain. The effect of MNAR was independent of c-Src activity, demonstrated by inhibitors and c-Src knockdown studies. In support of the role of MNAR in modulating GR transactivation, coimmunoprecipitation studies showed interaction between MNAR and GR in the nucleus but not the cytoplasm. Furthermore, MNAR and c-Src were also found to physically interact in the nucleus. Immunofluorescence studies showed MNAR to be predominantly a nuclear protein, with significant colocalization with GR. Deletion studies revealed that MNAR 884-1130 was coimmunoprecipitated with GR, and furthermore this fragment inhibited GR transactivation function when overexpressed. In addition, MNAR 1-400, which contains multiple LxxLL motifs, also inhibited GR transactivation. Taken together, MNAR interacts with GR in the nucleus but not cytoplasm and regulates GR transactivation in a complex manner depending on cell type. MNAR is capable of regulating both AF1 and AF2 functions of the GR independently. MNAR expression is likely to mediate important cell variation in glucocorticoid responsiveness, in a c-Src-independent mechanism.
Collapse
Affiliation(s)
- Midori Kayahara
- Faculty of Medical and Human Sciences, Centre for Molecular Medicine, School of Clinical and Laboratory Sciences, University of Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
112
|
Park YS, Ha Choi Y, Park CH, Kim KT. Nongenomic glucocorticoid effects on activity-dependent potentiation of catecholamine release in chromaffin cells. Endocrinology 2008; 149:4921-7. [PMID: 18583423 PMCID: PMC2734489 DOI: 10.1210/en.2007-1798] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adrenal medulla chromaffin cells are neuroendocrine and modified sympathetic ganglion cells. Catecholamines released from chromaffin cells mediate the fight-or-flight response or alert reaction against dangerous conditions. Here we report that short-term treatment with glucocorticoids, released from adrenal cortex cells in response to chronic stress, inhibits activity-dependent potentiation (ADP) of catecholamine release. First, short-term treatment with dexamethasone (DEX), a synthetic glucocorticoid, reduces ADP in a concentration-dependent manner (IC50 324.2+/-54.5 nM). The inhibitory effect of DEX is not reversed by RU-486 treatment, suggesting that the rapid inhibitory effect of DEX on ADP of catecholamine release is independent of glucocorticoid receptors. Second, DEX treatment reduces the frequency of fusion between vesicles and plasma membrane without affecting calcium influx. DEX disrupts activity-induced vesicle translocation and F-actin disassembly, thereby leading to inhibition of the vesicle fusion frequency. Third, we provide evidence that DEX reduces F-actin disassembly via inhibiting phosphorylation and translocation of myristoylated alanine-rich C kinase substrate and its upstream kinase protein kinase Cepsilon. Altogether, we suggest that glucocorticoids inhibit ADP of catecholamine release by decreasing myristoylated alanine-rich C kinase substrate phosphorylation, which inhibits F-actin disassembly and vesicle translocation.
Collapse
Affiliation(s)
- Yong-Soo Park
- Department of Life Science, Pohang University of Science and Technology, San 31, Hyoja Dong, Pohang 790-784, Republic of Korea
| | | | | | | |
Collapse
|
113
|
Pegram M. Can we circumvent resistance to ErbB2-targeted agents by targeting novel pathways? Clin Breast Cancer 2008; 8 Suppl 3:S121-30. [PMID: 18777951 DOI: 10.3816/cbc.2008.s.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The recent development of targeted therapies using monoclonal antibodies has added new dimensions to the rapidly evolving field of breast cancer treatment. In particular, the incorporation of trastuzumab into regimens containing existing chemotherapeutic agents has significantly improved clinical outcomes for patients with breast cancer in the adjuvant and metastatic settings. De novo and acquired resistance to this treatment, however, is widespread. A substantial amount of research has therefore been dedicated to the elucidation of molecular mechanisms that could explain resistance to this otherwise effective therapy. Potential mechanisms for resistance to trastuzumab include steric inhibition imposed by other extracellular factors, molecular changes in the target receptor itself (ErbB2), alterations in the regulation of downstream signaling components, and crosstalk with other pathways that could compensate for attenuated ErbB2 signaling. In addition, preclinical and clinical studies have been performed to identify potential methods for overcoming trastuzumab resistance, including targeting alternate ErbB2 epitopes and the combined inhibition of multiple signaling components and/or pathways (vertical or horizontal inhibition). Studies continue to evaluate the most promising approaches for overcoming mechanisms of resistance to trastuzumab and other ErbB2-targeted therapies. This review will summarize the most recent research designed to address this substantial clinical problem and provide clinicians with relevant background for understanding some of the potential molecular mechanisms for resistance to targeted therapies in the treatment of patients with breast cancer.
Collapse
Affiliation(s)
- Mark Pegram
- Division of Hematology/Oncology, Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
114
|
Regitz‐Zagrosek V, Becher E, Mahmoodzadeh S, Schubert C. Sex Steroid Hormones. CARDIOVASCULAR HORMONE SYSTEMS 2008:39-64. [DOI: 10.1002/9783527626236.ch2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
115
|
Kelly MJ, Rønnekleiv OK. Membrane-initiated estrogen signaling in hypothalamic neurons. Mol Cell Endocrinol 2008; 290:14-23. [PMID: 18538919 PMCID: PMC2601664 DOI: 10.1016/j.mce.2008.04.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 10/24/2022]
Abstract
It is well known that many of the actions of 17beta-estradiol (E2) in the central nervous system are mediated via intracellular receptor/transcription factors that interact with steroid response elements on target genes. However, there is compelling evidence for membrane steroid receptors for estrogen in hypothalamic and other brain neurons. But it is not well understood how estrogen signals via membrane receptors, and how these signals impact not only membrane excitability but also gene transcription in neurons. Indeed, it has been known for sometime that E2 can rapidly alter neuronal activity within seconds, indicating that some cellular effects can occur via membrane delimited events. In addition, E2 can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell signaling. Therefore, this review will consider our current knowledge of rapid membrane-initiated and intracellular signaling by E2 in the hypothalamus, the nature of receptors involved and how they contribute to homeostatic functions.
Collapse
Affiliation(s)
- Martin J Kelly
- Department of Physiology and Pharmacology, L334, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| | | |
Collapse
|
116
|
Abstract
Athletes and bodybuilders have recognized for several decades that the use of anabolic steroids can promote muscle growth and strength but it is only relatively recently that these agents are being revisited for clinical purposes. Anabolic steroids are being considered for the treatment of cachexia associated with chronic disease states, and to address loss of muscle mass in the elderly, but nevertheless their efficacy still needs to be demonstrated in terms of improved physical function and quality of life. In sport, these agents are performance enhancers, this being particularly apparent in women, although there is a high risk of virilization despite the favourable myotrophic-androgenic dissociation that many xenobiotic steroids confer. Modulation of androgen receptor expression appears to be key to partial dissociation, with consideration of both intracellular steroid metabolism and the topology of the bound androgen receptor interacting with co-activators. An anticatabolic effect, by interfering with glucocorticoid receptor expression, remains an attractive hypothesis. Behavioural changes by non-genomic and genomic pathways probably help motivate training. Anabolic steroids continue to be the most common adverse finding in sport and, although apparently rare, designer steroids have been synthesized in an attempt to circumvent the dope test. Doping with anabolic steroids can result in damage to health, as recorded meticulously in the former German Democratic Republic. Even so, it is important not to exaggerate the medical risks associated with their administration for sporting or bodybuilding purposes but to emphasize to users that an attitude of personal invulnerability to their adverse effects is certainly misguided.
Collapse
Affiliation(s)
- A T Kicman
- King's College London, Drug Control Centre, Department of Forensic Science and Drug Monitoring, London, UK.
| |
Collapse
|
117
|
Arruvito L, Giulianelli S, Flores AC, Paladino N, Barboza M, Lanari C, Fainboim L. NK cells expressing a progesterone receptor are susceptible to progesterone-induced apoptosis. THE JOURNAL OF IMMUNOLOGY 2008; 180:5746-53. [PMID: 18390760 DOI: 10.4049/jimmunol.180.8.5746] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been proposed that progesterone (P4) induces the suppression of immune responses, particularly during pregnancy. However, knowledge about the mechanisms involved has remained largely elusive. We demonstrate herein that peripheral blood NK (PBNK) cells express both classical progesterone receptor (PR) isoforms and are specifically affected by the actions of P4 through two apparently independent mechanisms. Progesterone induces caspase-dependent PBNK cell death, which is reversed by two different anti-progestins, ZK 98.299 and RU 486, supporting the involvement of classical PR isoforms. It was suggested that CD56(bright)CD16(-) killer Ig-like receptor (KIR)(-) NK cells might represent precursor cells, which, upon activation, acquire the features of a more mature NK subset expressing KIR receptors. The present study demonstrates that PR expression seems to be restricted to more mature KIR(+) PBNK cells. The expression of PR had a functional counterpart in the suppressive effect of P4 on IL-12-induced IFN-gamma secretion. This cytokine suppression was mainly observed in KIR(+) PBNK cells, without affecting the high secretion of IFN-gamma by CD56(bright) PBNK cells. The lack of PR expression on CD56(bright)KIR(-) PBNK cells provides an additional phenotypic marker to test the idea that they might represent the PBNK precursors selectively recruited into the endometrium where they differentiate to become the uterine NK cells. Additionally, these findings may be relevant to NK cell function in viral immunity, human reproduction, and tumor immunity.
Collapse
Affiliation(s)
- Lourdes Arruvito
- Immunogenetics Laboratory, José de San Martín Clinical Hospital, School of Medicine, University of Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
118
|
Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol 2008; 29:238-57. [PMID: 18083219 DOI: 10.1016/j.yfrne.2007.08.003] [Citation(s) in RCA: 263] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 08/14/2007] [Indexed: 12/16/2022]
Abstract
Ligands for the nuclear receptor superfamily have at least two mechanisms of action: (a) classical transcriptional regulation of target genes (genomic mechanisms); and (b) non-genomic actions, which are initiated at the cell membrane, which could also impact transcription. Though transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. This has led to considerable debate over the physiological relevance of membrane-initiated actions of hormones versus genomic actions of hormones, with genomic actions predominating in the endocrine field. There is good evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium and that these are linked to physiologically relevant scenarios in the brain. We show evidence in this review, that membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription in both the central nervous system and in non-neuronal cell lines. We present a theoretical scenario which can be used to understand this phenomenon. These signaling cascades may occur in parallel or in series but subsequently, converge at the modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other non-cognate hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription, though the relevance of this is less clear. The idea that coupling between membrane-initiated and genomic actions of hormones is a novel idea in neuroendocrinology and provides us with a unified view of hormone action in the central nervous system.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Cell and Molecular Biology Department, Tulane University, LA 70118, USA.
| | | |
Collapse
|
119
|
Piccart M. Circumventing De Novo and Acquired Resistance to Trastuzumab: New Hope for the Care of ErbB2-Positive Breast Cancer. Clin Breast Cancer 2008; 8 Suppl 3:S100-13. [DOI: 10.3816/cbc.2008.s.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
120
|
Kapoor R, Sheng JJ. Transfection of human prostate cancer CA-HPV-10 cells with cytosolic sulfotransferase SULT1E1 affects estrogen signaling and gene transcription. Drug Metab Dispos 2008; 36:316-21. [PMID: 17984287 DOI: 10.1124/dmd.107.017632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Human cytosolic sulfotransferase SULT1E1 catalyzes the sulfation of estrogens and estrogenic drugs in human reproductive tissues. Logically, this estrogen-preferring sulfotransferase isoform could play a regulatory role in estrogen signaling activities in human reproductive cells, including the prostate cells. This hypothesis was tested using DNA microarray and real-time reverse transcription-polymerase chain reaction methods in the present work. Potential changes in the transcriptional expression of selected signal transduction-related genes in human prostate cancer CA-HPV-10 cell line after SULT1E1 transfection were examined by DNA microarray methods. Notable changes were observed in the mRNA expression levels of TFRC, a cell membrane transferrin receptor gene, and TMEPAI, a gene encoding a steroid-dependent mRNA product. Expression of TFRC was down-regulated, whereas expression of TMEPAI was up-regulated by SULT1E1 transfection in CA-HPV-10 cells. Data from the current studies also showed that the estrogen-induced estrogen response element activation in CA-HPV-10 cells was repressed after the cells were transfected with SULT1E1. These results indicate that SULT1E1 may function as a transcriptional mediator in human prostate cancer CA-HPV-10 cells.
Collapse
Affiliation(s)
- Ruchita Kapoor
- Department of Pharmaceutical Sciences, College of Pharmacy, North Dakota State University, Fargo, ND 58105, USA
| | | |
Collapse
|
121
|
Morrill GA, Kostellow AB, Askari A. Progesterone binding to the alpha1-subunit of the Na/K-ATPase on the cell surface: insights from computational modeling. Steroids 2008; 73:27-40. [PMID: 17936318 PMCID: PMC2275170 DOI: 10.1016/j.steroids.2007.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 08/22/2007] [Accepted: 08/23/2007] [Indexed: 11/26/2022]
Abstract
Progesterone triggers the resumption of meiosis in the amphibian oocyte through a signaling system at the plasma membrane. Analysis of [(3)H]ouabain and [(3)H]progesterone binding to the plasma membrane of the Rana pipiens oocyte indicates that progesterone competes with ouabain for a low affinity ouabain binding site on a 112kDa alpha1-subunit of the membrane Na/K-ATPase. Published amino acid sequences from both low and high affinity ouabain binding alpha1-subunits are compared, together with published site-directed mutagenesis studies of ouabain binding. We propose that the progesterone binding site is located in the external loop (23 amino acids) between the M1-M2 transmembrane helices. Analysis of loop topology and the countercurrent hydrophobicity/polarity gradients within the M1-M2 loop further suggest that the polar beta and hydrophobic alpha surfaces of the planar progesterone molecule interact with opposite sides of the amino acid loop. The 19-angular methyl group of progesterone is essential for activity; it could bind to the C-terminal region of the M1-M2 loop. Maximum biological activity requires formation of hydrogen-bond networks between the 3-keto group of progesterone and Arg(118), Asp(129) and possibly Glu(122-124) in the C-terminal region of the loop. The 20-keto group hydrogen may in turn hydrogen bond to Cys(111) near the M1 helix. Peptide flexibility undergoes a maximal transition near the midway point in the M1-M2 loop, suggesting that folding occurs within the loop, which further stabilizes progesterone binding.
Collapse
Affiliation(s)
- Gene A Morrill
- Department of Physiology & Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
122
|
Stahn C, Löwenberg M, Hommes DW, Buttgereit F. Molecular mechanisms of glucocorticoid action and selective glucocorticoid receptor agonists. Mol Cell Endocrinol 2007; 275:71-8. [PMID: 17630118 DOI: 10.1016/j.mce.2007.05.019] [Citation(s) in RCA: 276] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 05/15/2007] [Accepted: 05/17/2007] [Indexed: 11/20/2022]
Abstract
Glucocorticoids (GC) are the most common used anti-inflammatory and immunosuppressive drugs in the treatment of rheumatic and other inflammatory diseases. Their therapeutic effects are considered to be mediated by four different mechanisms of action: the classical genomic mechanism of action caused by the cytosolic glucocorticoid receptor (cGCR); secondary non-genomic effects which are also initiated by the cGCR; membrane-bound glucocorticoid receptor (mGCR)-mediated non-genomic effects; non-specific, non-genomic effects caused by interactions with cellular membranes. The classical, genomic mechanism of GC-action can be divided into two processes: "transrepression", which is responsible for a large number of desirable anti-inflammatory and immunomodulating effects, and "transactivation" which is associated with frequently occurring side effects as well as with some immunosuppressive activities [Ehrchen, J., Steinmuller, L., Barczyk, K., Tenbrock, K., Nacken, W., Eisenacher, M., Nordhues, U., Sorg, C., Sunderkotter, C., Roth, J., 2007. Glucocorticoids induce differentiation of a specifically activated, anti-inflammatory subtype of human monocytes. Blood 109, 1265-1274]. Great efforts have been made to diminish glucocorticoid-induced adverse effects, but the improvement of conventional glucocorticoids has almost reached its limits. As a consequence, new variations of the conventional "good old drugs" are being tested and nitro-steroids and long circulating liposomal glucocorticoids indeed show promising results. Nevertheless, crux of the matter should be the design of qualitatively new drugs, such as selective glucocorticoid receptor agonists (SEGRAs). These innovative steroidal or non-steroidal molecules induce transrepression, while transactivation processes are less affected. First reports on two different GCR ligands, A276575 and ZK216348, show promising results. Here, we review the above-mentioned mechanisms of glucocorticoid action and give particular attention to the development of optimized glucocorticoids and SEGRAs.
Collapse
Affiliation(s)
- Cindy Stahn
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Schumannstrasse 20/21, 10117 Berlin, Germany.
| | | | | | | |
Collapse
|
123
|
Yue W, Fan P, Wang J, Li Y, Santen RJ. Mechanisms of acquired resistance to endocrine therapy in hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 2007; 106:102-10. [PMID: 17616457 PMCID: PMC2147683 DOI: 10.1016/j.jsbmb.2007.05.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Acquired resistance is a major problem limiting the clinical benefit of endocrine therapy. To investigate the mechanisms involved, two in vitro models were developed from MCF-7 cells. Long-term culture of MCF-7 cells in estrogen deprived medium (LTED) mimics aromatase inhibition in patients. Continued exposure of MCF-7 to tamoxifen represents a model of acquired resistance to antiestrogens (TAM-R). Long-term estrogen deprivation results in sustained activation of the ERK MAP kinase and the PI3 kinase/mTOR pathways. Using a novel Ras inhibitor, farnesylthiosalicylic acid (FTS), to achieve dual inhibition of the pathways, we found that the mTOR pathway plays the primary role in mediation of proliferation of LTED cells. In contrast to the LTED model, there is no sustained activation of ERK MAPK but enhanced responsiveness to rapid stimulation induced by E(2) and TAM in TAM-R cells. An increased amount of ERalpha formed complexes with EGFR and c-Src in TAM-R cells, which apparently resulted from extra-nuclear redistribution of ERalpha. Blockade of c-Src activity drove ERalpha back to the nucleus and reduced ERalpha-EGFR interaction. Prolonged blockade of c-Src activity restored sensitivity of TAM-R cells to tamoxifen. Our results suggest that different mechanisms are involved in acquired endocrine resistance and the necessity for individualized treatment of recurrent diseases.
Collapse
Affiliation(s)
- Wei Yue
- Department of Internal Medicine, University of Virginia Health System, Charlottesville, VA 22903, USA.
| | | | | | | | | |
Collapse
|
124
|
Bezdickova M, Molikova R, Bebarova L, Kolar Z. DISTRIBUTION OF NUCLEAR RECEPTORS FOR STEROID HORMONES IN THE HUMAN BRAIN: A PRELIMINARY STUDY. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2007; 151:69-71. [PMID: 17690743 DOI: 10.5507/bp.2007.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Expression of the nuclear steroid hormone receptors (SHR) within certain parts of the human brain has been described by many authors. However, a comprehensive analysis of SHR expression in the human brain still has not been performed. AIM To investigate the expression of SHR in different anatomical areas of the brain, especially within the neocortex. METHOD Immunohistochemical expression of estrogen receptors (ER), progesterone receptors (PR) and androgen receptors (AR) in different regions of the human brain was examined. RESULTS Nuclear expression of the AR was found in the mamillary body, praecentral gyrus and hippocampus of males. The same expression in analysed structures of female was not found. The expression of ER and PR was not observed. CONCLUSIONS The analysis revealed unexpected localization of SHR within the brain cortex, which could be the first step to the explanation of SHR action in brain as an interrelationship to function and behaviour. These results indicate on the possibility of SHR detection in post-mortal brain.
Collapse
Affiliation(s)
- Marcela Bezdickova
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic.
| | | | | | | |
Collapse
|
125
|
Li S, Li X, Li J, Deng X, Li Y, Cong Y. Experimental arterial thrombosis regulated by androgen and its receptor via modulation of platelet activation. Thromb Res 2007; 121:127-34. [PMID: 17451792 DOI: 10.1016/j.thromres.2007.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 02/26/2007] [Accepted: 03/06/2007] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The aim of our study is to elucidate whether experimental arterial thrombosis is regulated by physiological doses of androgen and its receptor via modulation of platelet activation. METHODS Surgical castration was performed in male rats and ferric chloride (FeCl(3)), as a stimulator, induced the experimental arterial thrombosis. Testosterone was measured directly by chemiluminescent immunoassay on the Bayer ADVIA Centaur analyzer. Dihydrotestosterone (DHT) was determined by ELISA using a commercially available kit. A platelet aggregometer was used to assess aggregation, and a platelet adherometer was used to measure adhesion. The contents of TXB(2) and 6-Keto-PGF(1alpha) were assayed by radio-immunoassay using commercially available kits. RESULTS Our data showed that DHT replaced restored circulating DHT of castrated rats to physiological levels, without being altered by treatment with flutamide. Castration caused significant increases in the thrombus area and weight in castrated rats as compared with control group. In PRP diluted with autologous PPP, ADP-induced platelet aggregation rate was only 9.10%. However, in PRP diluted with Tyrode's buffer, 1 microM ADP-induced platelet aggregation rate rose to 63.65%. In PRP diluted with Tyrode's buffer, and pretreated with DHT (1 nM, 2 nM), ADP-induced platelet aggregation was significantly lowered again. Platelet aggregation in PRP diluted with autologous PPP was enhanced in castrated rats as compared with sham-operated rats, and DHT (2 nM) replacement suppressed platelet aggregation in castrated PRP to the level similar to that of sham-operated rats. However, presence of flutamide (3 microM) significantly increased platelet aggregation in PRP diluted with autologous PPP or Tyrode's buffer. DHT (2 nM) replacement significantly inhibited the ADP-induced platelet adhesion. However, presence of flutamide (3 microM) increased ADP-induced platelet adhesion again. DHT replacement obviously reduced the ratio of TXB(2) to 6-keto-PGF(1alpha) in castrated rats. However, administration of flutamide and DHT to castrated rats caused an increase in the ratio of TxB(2) to 6-keto-PGF1alpha. CONCLUSION Inhibition of experimental arterial thrombosis by androgen at physiological doses and its receptor is mediated via modulation of platelet activation.
Collapse
Affiliation(s)
- ShiJun Li
- Division of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
126
|
Bains M, Cousins JC, Roberts JL. Neuroprotection by estrogen against MPP+-induced dopamine neuron death is mediated by ERalpha in primary cultures of mouse mesencephalon. Exp Neurol 2007; 204:767-76. [PMID: 17320868 PMCID: PMC3841287 DOI: 10.1016/j.expneurol.2007.01.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 11/13/2006] [Accepted: 01/08/2007] [Indexed: 11/16/2022]
Abstract
Estrogen involvement in neuroprotection is now widely accepted, although the specific molecular and cellular mechanisms of estrogen action in neuroprotection remain unclear. This study examines estrogenic effects in a mixed population of cells in attempts to identify the contributing cells that result in estrogen-mediated neuroprotection. Utilizing primary mesencephalic neurons, we found expression of both estrogen receptor alpha (ERalpha) and estrogen receptor beta (ERbeta) with a predominance of ERalpha on both dopamine neurons and astrocytes. We also found that 17beta-estradiol protects dopamine neurons from injury induced by the complex I inhibitor, 1-methyl-4-phenyl pyridinium (MPP(+)) in a time- and ER-dependent manner. At least 4 h of estrogen pre-treatment was required to elicit protection, an effect that was blocked by the ER antagonist, ICI 182,780. Moreover, ERalpha mediated the protection afforded by estrogen since only the ERalpha agonist, HPTE, but not the ERbeta agonist, DPN, protected against dopamine cell loss. Since glial cells were shown to express significant levels of ERalpha, we investigated a possible indirect mechanism of estrogen-mediated neuroprotection through glial cell interaction. Removal of glial cells from the cultures by application of the mitotic inhibitor, 5-fluoro-2'-deoxyuridine, significantly reduced the neuroprotective effects of estrogen. These data indicate that neuroprotection provided by estrogen against MPP(+) toxicity is mediated by ERalpha and involves an interplay among at least two cell types.
Collapse
Affiliation(s)
- Mona Bains
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Joanne C. Cousins
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - James L. Roberts
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| |
Collapse
|
127
|
Wierman ME. Sex steroid effects at target tissues: mechanisms of action. ADVANCES IN PHYSIOLOGY EDUCATION 2007; 31:26-33. [PMID: 17327579 DOI: 10.1152/advan.00086.2006] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Our understanding of the mechanisms of sex hormone action has changed dramatically over the last 10 years. Estrogens, progestins, and androgens are the steroid hormones that modulate reproductive function. Recent data have shown that many other tissues are targets of sex hormones in addition to classical reproductive organs. This review outlines new advances in our understanding of the spectrum of steroid hormone ligands, newly recognized target tissues, structure-function relationships of steroid receptors, and, finally, their genomic and nongenomic actions. Sex-based specific effects are often related to the different steroid hormone mileu in men compared with women. Understanding the mechanisms of sex steroid action gives insight into the differences in normal physiology and disease states.
Collapse
Affiliation(s)
- Margaret E Wierman
- University of Colorado at Denver and Health Sciences Center, Aurora, Colorado, USA.
| |
Collapse
|
128
|
Gill A, Hammes SR. G beta gamma signaling reduces intracellular cAMP to promote meiotic progression in mouse oocytes. Steroids 2007; 72:117-23. [PMID: 17178138 PMCID: PMC1853321 DOI: 10.1016/j.steroids.2006.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 11/12/2006] [Indexed: 10/23/2022]
Abstract
In nearly every vertebrate species, elevated intracellular cAMP maintains oocytes in prophase I of meiosis. Prior to ovulation, gonadotropins trigger various intra-ovarian processes, including the breakdown of gap junctions, the activation of EGF receptors, and the secretion of steroids. These events in turn decrease intracellular cAMP levels in select oocytes to allow meiotic progression, or maturation, to resume. Studies suggest that cAMP levels are kept elevated in resting oocytes by constitutive G protein signaling, and that the drop in intracellular cAMP that accompanies maturation may be due in part to attenuation of this inhibitory G protein-mediated signaling. Interestingly, one of these G protein regulators of meiotic arrest is the Galpha(s) protein, which stimulates adenylyl cyclase to raise intracellular cAMP in two important animal models of oocyte development: Xenopus leavis frogs and mice. In addition to G(alpha)(s), constitutive Gbetagamma activity similarly stimulates adenylyl cyclase to raise cAMP and prevent maturation in Xenopus oocytes; however, the role of Gbetagamma in regulating meiosis in mouse oocytes has not been examined. Here we show that Gbetagamma does not contribute to the maintenance of murine oocyte meiotic arrest. In fact, contrary to observations in frog oocytes, Gbetagamma signaling in mouse oocytes reduces cAMP and promotes oocyte maturation, suggesting that Gbetagamma might in fact play a positive role in promoting oocyte maturation. These observations emphasize that, while many general concepts and components of meiotic regulation are conserved from frogs to mice, specific differences exist that may lead to important insights regarding ovarian development in vertebrates.
Collapse
Affiliation(s)
- Arvind Gill
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, Texas 75390-8857, USA
| | - Stephen R Hammes
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, Texas 75390-8857, USA
- Corresponding author: Phone: 214-648-3749, FAX: 214-648-7934,
| |
Collapse
|
129
|
Bhattacharyya S, Brown DE, Brewer JA, Vogt SK, Muglia LJ. Macrophage glucocorticoid receptors regulate Toll-like receptor 4-mediated inflammatory responses by selective inhibition of p38 MAP kinase. Blood 2007; 109:4313-9. [PMID: 17255352 PMCID: PMC1885507 DOI: 10.1182/blood-2006-10-048215] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To explore the role of glucocorticoids in regulation of kinase pathways during innate immune responses, we generated mice with conditional deletion of glucocorticoid receptor (GR) in macrophages (MGRKO). Activation of toll-like receptor 4 (TLR4) by lipopolysaccharide (LPS) caused greater mortality and cytokine production in MGRKO mice than in controls. Ex vivo, treatment with dexamethasone (Dex) markedly inhibited LPS-mediated induction of inflammatory genes in control but not GR-deficient macrophages. We show that Dex inhibits p38 MAPK, but not PI3K/Akt, ERK, or JNK, in control macrophages. Associated with p38 inhibition, Dex induced MAP kinase phosphatase-1 (MKP-1) in control, but not MGRKO, macrophages. Consistent with the ex vivo studies, treatment with a p38 MAPK-specific inhibitor resulted in rescue of MGRKO mice from LPS-induced lethality. Taken together, we identify p38 MAPK and its downstream targets as essential for GR-mediated immunosuppression in macrophages.
Collapse
Affiliation(s)
- Sandip Bhattacharyya
- Departments of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
130
|
Greger JG, Fursov N, Cooch N, McLarney S, Freedman LP, Edwards DP, Cheskis BJ. Phosphorylation of MNAR promotes estrogen activation of phosphatidylinositol 3-kinase. Mol Cell Biol 2006; 27:1904-13. [PMID: 17194752 PMCID: PMC1820473 DOI: 10.1128/mcb.01732-06] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Estrogen actions are mediated by a complex interface of direct control of gene expression (the so-called "genomic action") and by regulation of cell signaling/phosphorylation cascades, referred to as the "nongenomic," or extranuclear, action. We have previously described the identification of MNAR (modulator of nongenomic action of estrogen receptor) as a novel scaffold protein that regulates estrogen receptor alpha (ERalpha) activation of cSrc. In this study, we have investigated the role of MNAR in 17beta-estradiol (E2)-induced activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Consistent with our previous results, a direct correlation was established between MNAR expression levels and E2-induced activation of PI3 and Akt kinases. Endogenous MNAR, ERalpha, cSrc, and p85, the regulatory subunit of PI3 kinase, interacted in MCF7 cells treated with E2. The interaction between p85 and MNAR required activation of cSrc and MNAR phosphorylation on Tyr 920. Consequently, the mutation of this tyrosine to alanine (Y920A) abrogated the interaction between MNAR and p85 and the E2-induced activation of the PI3K/Akt pathway, which was required for the E2-induced protection of MCF7 cells from apoptosis. Nonetheless, the Y920A mutant potentiated the E2-induced activation of the Src/MAPK pathway and MCF7 cell proliferation, as observed with the wild-type MNAR. These results provide new and important insights into the molecular mechanisms of E2-induced regulation of cell proliferation and apoptosis.
Collapse
Affiliation(s)
- James G Greger
- Women's Health and Musculoskeletal Biology, Wyeth Research, Nuclear Receptors, 500 Arcola Road, Collegeville, PA 19426, USA
| | | | | | | | | | | | | |
Collapse
|
131
|
Kousteni S, Almeida M, Han L, Bellido T, Jilka RL, Manolagas SC. Induction of osteoblast differentiation by selective activation of kinase-mediated actions of the estrogen receptor. Mol Cell Biol 2006; 27:1516-30. [PMID: 17158928 PMCID: PMC1800724 DOI: 10.1128/mcb.01550-06] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Estrogens control gene transcription by cis or trans interactions of the estrogen receptor (ER) with target DNA or via the activation of cytoplasmic kinases. We report that selective activation of kinase-mediated actions of the ER with 4-estren-3alpha,17beta-diol (estren) or an estradiol-dendrimer conjugate, each a synthetic compound that stimulates kinase-mediated ER actions 1,000 to 10,000 times more potently than direct DNA interactions, induced osteoblastic differentiation in established cell lines of uncommitted osteoblast precursors and primary cultures of osteoblast progenitors by stimulating Wnt and BMP-2 signaling in a kinase-dependent manner. In sharp contrast, 17beta-estradiol (E(2)) suppressed BMP-2-induced osteoblast progenitor commitment and differentiation. Consistent with the in vitro findings, estren, but not E(2), stimulated Wnt/beta-catenin-mediated transcription in T-cell factor-lacZ transgenic mice. Moreover, E(2) stimulated BMP signaling in mice in which ERalpha lacks DNA binding activity and classical estrogen response element-mediated transcription (ERalpha(NERKI/-)) but not in wild-type controls. This evidence reveals for the first time the existence of a large signalosome in which inputs from the ER, kinases, bone morphogenetic proteins, and Wnt signaling converge to induce differentiation of osteoblast precursors. ER can either induce it or repress it, depending on whether the activating ligand (and presumably the resulting conformation of the receptor protein) precludes or accommodates ERE-mediated transcription.
Collapse
Affiliation(s)
- Stavroula Kousteni
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA
| | | | | | | | | | | |
Collapse
|
132
|
Germain P, Staels B, Dacquet C, Spedding M, Laudet V. Overview of nomenclature of nuclear receptors. Pharmacol Rev 2006; 58:685-704. [PMID: 17132848 DOI: 10.1124/pr.58.4.2] [Citation(s) in RCA: 473] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear receptor pharmacology has, to a certain extent, led the way, compared with other receptor systems, in the appreciation that ligands may exert very diverse pharmacology, based on their individual chemical structure and the allosteric changes induced in the receptor/accessory protein complex. This can lead to very selective pharmacological effects, which may not necessarily be predicted from the experience with other agonists/partial agonists/antagonists. If this is the case, then drug discovery may be back to drug-specific pharmacology (where each drug may have an original profile), rather than specific-drug pharmacology (where agents specific for a receptor have a distinct profile). As functional selectivity is indeed a crucial mechanism to be considered when going through the drug discovery development process, then initial screens using reconstituted systems may not show the appropriate pharmacology, simply because the required stoichiometry of corepressors and coactivators may not be present to select the best compounds; therefore, multiple effector systems are necessary to screen for differential activation, and, even then, screening with in vivo pathophysiological models may ultimately be required for the selection process-a massive but necessary task for pharmacologists. Thus, the characterization of nuclear receptors and their associated proteins and the ligands that interact with them will remain a challenge to pharmacologists.
Collapse
Affiliation(s)
- Pierre Germain
- Department of Cell Biology and Signal Transduction, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France.
| | | | | | | | | |
Collapse
|
133
|
Clark AS, Costine BA, Jones BL, Kelton-Rehkopf MC, Meerts SH, Nutbrown-Greene LL, Penatti CAA, Porter DM, Yang P, Henderson LP. Sex- and age-specific effects of anabolic androgenic steroids on reproductive behaviors and on GABAergic transmission in neuroendocrine control regions. Brain Res 2006; 1126:122-38. [PMID: 17010954 DOI: 10.1016/j.brainres.2006.08.081] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 08/16/2006] [Accepted: 08/17/2006] [Indexed: 12/21/2022]
Abstract
Illicit use of anabolic androgenic steroids (AAS) has become a prevalent health concern not only among male professional athletes, but, disturbingly, among a growing number of women and adolescent girls. Despite the increasing use of AAS among women and adolescents, few studies have focused on the effects of these steroids in females, and female adolescent subjects are particularly underrepresented. Among the hallmarks of AAS abuse are changes in reproductive behaviors. Here, we discuss work from our laboratories on the actions of AAS on the onset of puberty and sexual behaviors in female rodents, AAS interactions and sex- and age-specific effects of these steroids on neural transmission mediated by gamma-aminobutyric acid receptors within forebrain neuroendocrine control regions that may underlie AAS-induced changes in these behaviors.
Collapse
Affiliation(s)
- Ann S Clark
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH 03755, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Spies CM, Schaumann DHS, Berki T, Mayer K, Jakstadt M, Huscher D, Wunder C, Burmester GR, Radbruch A, Lauster R, Scheffold A, Buttgereit F. Membrane glucocorticoid receptors are down regulated by glucocorticoids in patients with systemic lupus erythematosus and use a caveolin-1-independent expression pathway. Ann Rheum Dis 2006; 65:1139-46. [PMID: 16449313 PMCID: PMC1798283 DOI: 10.1136/ard.2005.048272] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2006] [Indexed: 11/04/2022]
Abstract
BACKGROUND Membrane-bound glucocorticoid receptors (mGCR) are up regulated on monocytes after in vitro stimulation and in patients with rheumatoid arthritis. Caveolin-1 is critical for the transport of plasma membrane oestrogen receptors to the cell surface. OBJECTIVES To investigate the expression of mGCR in patients with systemic lupus erythematosus (SLE)-a disease with different aetiopathogenesis and treatment regimens-and to examine whether caveolin-1 is critical for the transport of mGCR to the cell surface. METHODS Frequencies of mGCR+ peripheral blood mononuclear cells were measured using high-sensitivity immunofluorescent staining and tested for correlation with SLE disease activity and glucocorticoid treatment. Semiquantitative polymerase chain reaction, immunofluorescence, recombinant expression and confocal laser-scanning microscopy were used to search for an association of mGCR with caveolin-1. RESULTS The frequencies of mGCR+ monocytes (CD14+) were considerably higher in patients with SLE (n = 33) than in healthy controls (n = 58), whereas B cells (CD19+) were not different in this regard. T cells (CD3+) were always mGCR-. The frequency of mGCR+ monocytes in patients with SLE did not correlate with disease activity, but did inversely correlate with glucocorticoid dosages; this inverse correlation was confirmed by corresponding in vitro experiments with stimulated monocytes. The induced up regulation of mGCR was not accompanied by an up regulation of caveolin-1, and mGCR are not colocalised with caveolin-1 in plasma membrane caveolae. CONCLUSION mGCR are (a) up regulated in patients with SLE and by inflammatory stimuli and (b) down regulated by glucocorticoids, suggesting a negative feedback loop to control glucocorticoid action. Drugs binding selectively to mGCR may in future prove to be of therapeutic value.
Collapse
Affiliation(s)
- C M Spies
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Campus Mitte, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Subbarayan V, Krieg P, Hsi LC, Kim J, Yang P, Sabichi AL, Llansa N, Mendoza G, Logothetis CJ, Newman RA, Lippman SM, Menter DG. 15-Lipoxygenase-2 gene regulation by its product 15-(S)-hydroxyeicosatetraenoic acid through a negative feedback mechanism that involves peroxisome proliferator-activated receptor gamma. Oncogene 2006; 25:6015-25. [PMID: 16682954 DOI: 10.1038/sj.onc.1209617] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An inverse relationship exists between the expression of 15-lipoxygenase-2 (15-LOX-2) and peroxisome proliferator-activated receptor gamma (PPARgamma) in normal prostate epithelial cells (PrECs) compared with their expression in prostate carcinoma cells (PC-3). The reason for this difference, however, is unknown. We hypothesized that this inverse expression partly involves the 15-LOX-2 promoter and 15-S-hydroxyeicosatetraenoic acid (15-(S)-HETE), a product of 15-LOX-2 that binds to PPARgamma. We identified an active steroid nuclear receptor half-site present in the 15-LOX-2 promoter fragment F-5 (-618/+177) that can interact with PPARgamma. After forced expression of wild-type PPARgamma, 15-(S)-HETE (1 microM) decreased F-5 reporter activity in PrECs whereas forced expression of 15-LOX-2 resulted in 15-(S)-HETE production which enhanced F-5 activity in PC-3. In contrast, the expression of dominant-negative PPARgamma reversed the transcriptional activation of F-5 by enhancing it 202-fold in PrEC or suppressing it in PC-3; the effect in PC-3 was positively increased 150-fold in the presence of 15-(S)-HETE (1 microM). Peroxisome proliferator-activated receptor gamma interacted with 15-LOX-2 promoter sequences in pulldown experiments using biotinylated 15-LOX-2 (-560/-596 bp) oligonucleotides. In gelshift analyses PPARgamma and orphan receptor RORalpha were shown to interact with the F-5 fragment in PC-3 cells. These data suggest that crosstalk mechanisms exist between the 15-LOX-2 gene and PPARgamma to counterbalance expression and help explain the inverse relationship of these genes in normal versus cancer cells.
Collapse
Affiliation(s)
- V Subbarayan
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Cazzola M. Single inhaler budesonide/formoterol in exacerbations of chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2006; 19:79-89. [PMID: 15964228 DOI: 10.1016/j.pupt.2005.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 02/24/2005] [Accepted: 03/21/2005] [Indexed: 11/25/2022]
Abstract
Inhaled bronchodilators, particularly short-acting inhaled beta(2)-agonists, and systemic glucocorticosteroids are effective treatments for acute exacerbations of chronic obstructive pulmonary disease (COPD). However, in the treatment of these episodes there may be some advantages to the longer-acting agents in that there will be prolonged bronchodilation. Moreover, high doses of systemic glucocorticosteroids are associated with a significant risk of side effects. In the last few years, evidence is mounting that nebulized budesonide and inhaled formoterol might be an alternative to oral prednisolone and short-acting beta(2)-agonists, respectively, in the treatment of acute exacerbations of COPD. Interestingly, some new data suggest that a combination therapy with single inhaler containing budesonide and formoterol may be an alternative to traditional therapy in the treatment of acute exacerbations of this disorder. However, since individual studies are typically statistically underpowered and are remarkably heterogeneous with regard to their conclusions, larger studies are needed to confirm these preliminary findings and determine conclusively any impact of budesonide/formoterol combination in acutely ill COPD patients.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Pneumology and Allergology, Department of Respiratory Medicine, Antonio Cardarelli High Specialty Hospital, Naples, Italy. mcazzola@qubisoftit
| |
Collapse
|
137
|
Dambaki C, Kogia C, Kampa M, Darivianaki K, Nomikos M, Anezinis P, Theodoropoulos PA, Castanas E, Stathopoulos EN. Membrane testosterone binding sites in prostate carcinoma as a potential new marker and therapeutic target: study in paraffin tissue sections. BMC Cancer 2005; 5:148. [PMID: 16293185 PMCID: PMC1318463 DOI: 10.1186/1471-2407-5-148] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2005] [Accepted: 11/17/2005] [Indexed: 11/28/2022] Open
Abstract
Background Steroid action is mediated, in addition to classical intracellular receptors, by recently identified membrane sites, that generate rapid non-genomic effects. We have recently identified a membrane androgen receptor site on prostate carcinoma cells, mediating testosterone rapid effects on the cytoskeleton and secretion within minutes. Methods The aim of this study was to investigate whether membrane androgen receptors are differentially expressed in prostate carcinomas, and their relationship to the tumor grade. We examined the expression of membrane androgen receptors in archival material of 109 prostate carcinomas and 103 benign prostate hyperplasias, using fluorescein-labeled BSA-coupled testosterone. Results We report that membrane androgen receptors are preferentially expressed in prostate carcinomas, and they correlate to their grade using the Gleason's microscopic grading score system. Conclusion We conclude that membrane androgen receptors may represent an index of tumor aggressiveness and possibly specific targets for new therapeutic regimens.
Collapse
Affiliation(s)
- Constantina Dambaki
- Department of Pathology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Christina Kogia
- Department of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Marilena Kampa
- Department of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Katherine Darivianaki
- Department of Pathology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Michael Nomikos
- Department of Urology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Ploutarchos Anezinis
- Department of Urology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Panayiotis A Theodoropoulos
- Department of Biochemistry, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Elias Castanas
- Department of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| | - Efstathios N Stathopoulos
- Department of Pathology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion, GR-71003, Crete, Greece
| |
Collapse
|
138
|
Schaefer TM, Wright JA, Pioli PA, Wira CR. IL-1β-Mediated Proinflammatory Responses Are Inhibited by Estradiol via Down-Regulation of IL-1 Receptor Type I in Uterine Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:6509-16. [PMID: 16272305 DOI: 10.4049/jimmunol.175.10.6509] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objective of this study was to examine the effects of sex hormones on IL-1beta-mediated responses by uterine epithelial cells. The mRNA expression and secretion of human beta-defensin-2 and CXCL8 by uterine epithelial cells was examined following stimulation with IL-1beta in the presence of estradiol or progesterone. Estradiol inhibited the IL-1beta-mediated mRNA expression and secretion of human beta-defensin-2 and CXCL8 by uterine epithelial cells while progesterone had no effect. Inhibition of the IL-1beta-mediated response by estradiol was dose dependent, with maximal inhibition observed using 10(-7) to 10(-10) M, and was shown to be mediated through the estrogen receptor because addition of a pure estrogen receptor antagonist abrogated this effect. The mechanism by which estradiol inhibits IL-1beta-mediated responses by uterine epithelial cells appears to be the down-modulation of the IL-1R type I, thereby reducing the uterine epithelial cell's ability to respond to IL-1beta. These results suggest that the inhibitory effect of estradiol on IL-1beta-mediated inflammatory responses by uterine epithelial cells indicates a link between the endocrine and immune systems and may be crucial for dampening proinflammatory responses during the time of ovulation or pregnancy.
Collapse
MESH Headings
- Cells, Cultured
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Estradiol/administration & dosage
- Estradiol/pharmacology
- Female
- Humans
- Inflammation/etiology
- Inflammation/immunology
- Inflammation/prevention & control
- Interleukin-1/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/drug effects
- Receptors, Estrogen/metabolism
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1 Type I
- Uterus/cytology
- Uterus/drug effects
- Uterus/immunology
- Uterus/metabolism
- beta-Defensins/biosynthesis
- beta-Defensins/genetics
Collapse
Affiliation(s)
- Todd M Schaefer
- Department of Physiology, Dartmouth Medical School, Lebanon, NH 03756, USA.
| | | | | | | |
Collapse
|
139
|
Affiliation(s)
- Turk Rhen
- Department of Biology, University of North Dakota, Grand Forks, USA
| | | |
Collapse
|
140
|
Devidze N, Mong JA, Jasnow AM, Kow LM, Pfaff DW. Sex and estrogenic effects on coexpression of mRNAs in single ventromedial hypothalamic neurons. Proc Natl Acad Sci U S A 2005; 102:14446-51. [PMID: 16186484 PMCID: PMC1242333 DOI: 10.1073/pnas.0507144102] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulated gene expression in single neurons can be linked to biophysical events and behavior in the case of estrogen-regulated gene expression in neurons in the ventrolateral portion of the ventromedial nucleus (VMN) of the hypothalamus. These cells are essential for lordosis behavior. What genes are coexpressed in neurons that have high levels of mRNAs for estrogen receptors (ERs)? We have been able to isolate and measure certain mRNAs from individual VMN neurons collected from rat hypothalamus. Large numbers of neurons express mRNA for ERalpha, but these neurons are not identical with the population of VMN neurons expressing the likely gene duplication product, ERbeta. An extremely high proportion of neurons expressing either ER also coexpress mRNA for the oxytocin receptor (OTR). This fact matches the known participation of oxytocin binding and signaling in sexual and affiliative behaviors. In view of data that ER and OTR can signal through PKCs, we looked at coexpression of selected PKCs in the same individual neurons. The most discriminating analysis was for triple coexpression of ERs, OTR, and each selected PKC isoform. These patterns of triple coexpression were significantly different for male vs. female VMN neurons. Further, individual neurons expressing ERalpha could distribute their signaling across the various PKC isoforms differently in different cells, whereas the reverse was not true. These findings and this methodology establish the basis for systematic linkage of the brain's hormone-sensitive signaling pathways to biophysical and behavioral mechanisms in a well studied mammalian system.
Collapse
Affiliation(s)
- N Devidze
- Laboratory of Neurobiology and Behavior, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
141
|
Heberden C, Reine F, Grosse B, Henry C, Zagar Y, Chaumaz G, Lieberherr M. Detection of a raft-located estrogen receptor-like protein distinct from ER alpha. Int J Biochem Cell Biol 2005; 38:376-91. [PMID: 16263324 DOI: 10.1016/j.biocel.2005.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 08/30/2005] [Accepted: 09/08/2005] [Indexed: 11/20/2022]
Abstract
17Beta-estradiol (17beta-E2) elicits at the cell membrane rapid actions that remain insensitive to the inhibitory effect of ICI 182,780, a pure estrogen antagonist, and therefore cannot be attributed to the classic nuclear receptors. We addressed the question of the identity of the protein involved in these rapid actions. We first examined the responses of several cell lines for intracellular calcium mobilization, an effect not inhibited by ICI 182,780, tamoxifen and raloxifen. We then demonstrated the presence of binding sites in the membranes, by incubating them with antibodies directed against different domains of ER alpha, and by flow cytometry analysis. The membrane proteins were eluted by affinity chromatography using E2 conjugated to bovine serum albumin as a ligand. Western blots of the elution fractions using an antibody directed against the ligand binding site of ER alpha showed the existence of a protein of approximately 50 kDa. The protein was concentrated in the lipid rafts, together with another heavier form of approximately 66 kDa. The 50 kDa protein was immunoprecipitable, and co-immunoprecipitation experiments showed that it was associated with the Gbeta(1-4) protein, but not with caveolin-1. The protein was expressed in ER alpha-null cells, like HO-23 and Cos-7 cells. Therefore, in the lipid rafts, there exists a protein, similar to, but molecularly distinct from ER alpha.
Collapse
Affiliation(s)
- Christine Heberden
- Laboratoire de Nutrition et Sécurité Alimentaire, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | | | | | | | | | | | | |
Collapse
|
142
|
Freeman MR, Cinar B, Lu ML. Membrane rafts as potential sites of nongenomic hormonal signaling in prostate cancer. Trends Endocrinol Metab 2005; 16:273-9. [PMID: 16002302 DOI: 10.1016/j.tem.2005.06.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 03/02/2005] [Accepted: 06/23/2005] [Indexed: 01/03/2023]
Abstract
Recent evidence indicates that nuclear receptors for steroid hormones can signal by nongenomic mechanisms that operate independently of their transcription function. These signal-transduction processes occur within seconds to minutes after initiation with agonist and involve interactions between nuclear receptors and other signaling proteins in the cytoplasm and at membrane surfaces. This review provides an overview of published information on possible nongenomic activities of the androgen receptor (AR) and other nuclear receptors, focusing on the potential involvement of these processes in prostate cancer. We discuss the hypothesis that the cholesterol-rich lipid-raft compartment(s) of cancer-cell membranes might provide privileged sites for nongenomic signals mediated by the AR.
Collapse
Affiliation(s)
- Michael R Freeman
- The Urological Diseases Research Center, Department of Urology, Children's Hospital Boston, Boston, MA 02115, USA.
| | | | | |
Collapse
|
143
|
Löwenberg M, Tuynman J, Bilderbeek J, Gaber T, Buttgereit F, van Deventer S, Peppelenbosch M, Hommes D. Rapid immunosuppressive effects of glucocorticoids mediated through Lck and Fyn. Blood 2005; 106:1703-10. [PMID: 15899916 DOI: 10.1182/blood-2004-12-4790] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Glucocorticoids (GCs) are effective immunosuppressive agents and mediate well-defined transcriptional effects via GC receptors. There is increasing evidence that GCs also initiate rapid nongenomic signaling events. Using activated human CD4(+) lymphocytes and a peptide array containing 1176 different kinase consensus substrates, we generated a comprehensive profile of GC-induced rapid effects on signal transduction. The results show marked early differences in phosphorylation between GC-pretreated cells and control cells, including impaired phosphorylation of p56lck/p59fyn (Lck/Fyn) consensus substrates. Immunoprecipitation and in vitro kinase assays reveal rapid GC-induced down-modulation of Lck and Fyn kinases using SAM68 (Src [pp60c-src]-associated in mitosis 68 kDa) as a substrate. Additionally, immunoprecipitation experiments revealed reduced Lck-CD4 and Fyn-CD3 associations, suggesting GC inhibited recruitment of these kinases to the T-cell receptor complex. Western blot analysis revealed reduced phosphorylation of a series of downstream signaling intermediates following GC treatment, including protein kinase B (PKB), protein kinase C (PKC), and mitogen-activated protein kinases (MAPKs). Experiments with GC receptor-negative Jurkat cells and a pharmacologic GC receptor ligand (RU486) indicated that rapid inhibition of Lck and Fyn kinases is GC receptor dependent. Parallel experiments conducted following the application of GCs in healthy individuals confirmed suppression of Lck/Fyn in T cells within 1 hour in vivo. These results identify the inhibition of Lck and Fyn kinases as rapid targets of GCs, mediated via a GC receptor-dependent pathway.
Collapse
Affiliation(s)
- Mark Löwenberg
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Meibergdreef 9, NL-1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Abstract
Tremendous progress has been made in elucidating numerous critical aspects of estrogen signaling. New tools and techniques have enabled detailed molecular analysis of components that direct estrogen responses. At the other end of the spectrum, generation of a multiplicity of transgenic animals has allowed analysis of the physiological roles of the estrogen-signaling components in biologically relevant models. Here, we review the ever-increasing body of knowledge in the field of estrogen biology, especially as applied to the female reproductive processes.
Collapse
Affiliation(s)
- Sylvia C Hewitt
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 277009, USA.
| | | | | |
Collapse
|
145
|
Haas D, White SN, Lutz LB, Rasar M, Hammes SR. The modulator of nongenomic actions of the estrogen receptor (MNAR) regulates transcription-independent androgen receptor-mediated signaling: evidence that MNAR participates in G protein-regulated meiosis in Xenopus laevis oocytes. Mol Endocrinol 2005; 19:2035-46. [PMID: 15831520 PMCID: PMC1482432 DOI: 10.1210/me.2004-0531] [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: 01/21/2023] Open
Abstract
Classical steroid receptors mediate many transcription-independent (nongenomic) steroid responses in vitro, including activation of Src and G proteins. Estrogen-triggered activation of Src can be regulated by the modulator of nongenomic actions of the estrogen receptor (MNAR), which binds to estrogen receptors and Src to create a signaling complex. In contrast, the mechanisms regulating steroid-induced G protein activation are not known, nor are the physiologic responses mediated by MNAR. These studies demonstrate that MNAR regulates the biologically relevant process of meiosis in Xenopus laevis oocytes. MNAR was located throughout oocytes, and reduction of its expression by RNA interference markedly enhanced testosterone-triggered maturation and activation of MAPK. Additionally, Xenopus MNAR augmented androgen receptor (AR)-mediated transcription in CV1 cells through activation of Src. MNAR and AR coimmunoprecipitated as a complex involving the LXXLL-rich segment of MNAR and the ligand binding domain of AR. MNAR and Gbeta also precipitated together, with the same region of MNAR being important for this interaction. Finally, reduction of MNAR expression decreased Gbetagamma-mediated signaling in oocytes. MNAR therefore appears to participate in maintaining meiotic arrest, perhaps by directly enhancing Gbetagamma-mediated inhibition of meiosis. Androgen binding to AR might then release this inhibition, allowing maturation to occur. Thus, MNAR may augment multiple nongenomic signals, depending upon the context and cell type in which it is expressed.
Collapse
Affiliation(s)
| | | | | | | | - Stephen R. Hammes
- Address all correspondence and requests for reprints to: Stephen R. Hammes, Department of Internal Medicine, Division of Endocrinology and Metabolism, Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390–8857. E-mail:
| |
Collapse
|
146
|
Moro L, Reineri S, Piranda D, Pietrapiana D, Lova P, Bertoni A, Graziani A, Defilippi P, Canobbio I, Torti M, Sinigaglia F. Nongenomic effects of 17β-estradiol in human platelets: potentiation of thrombin-induced aggregation through estrogen receptor β and Src kinase. Blood 2005; 105:115-21. [PMID: 15198955 DOI: 10.1182/blood-2003-11-3840] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AbstractThe impact of estrogens on the cardiovascular system and their ability to regulate platelet function are matters of controversy. The recent finding that estrogen receptors are expressed in human platelets renders these cells an excellent model for studying the nongenomic effects of these hormones. In this work, we investigated 17β-estradiol–dependent signaling in platelets from adult healthy men. 17β-estradiol caused the rapid phosphorylation of the tyrosine kinases Src and Pyk2 and the formation of a signaling complex, which included Src, Pyk2, and the phosphatidylinositol 3-kinase. Both these events were dependent on estrogen receptor β engagement. We found that estrogen receptor β was membrane-associated in platelets. On treatment with 17β-estradiol, Src and Pyk2 activation occurred in the membrane fraction but not in the cytosol. In contrast, no significant activation of phosphatidylinositol 3-kinase was detected in estrogen-treated platelets. 17β-estradiol did not induce any platelet response directly, but it strongly potentiated the activation of integrin αIIbβ3 and the platelet aggregation induced by subthreshold concentrations of thrombin. These effects were dependent on estrogen receptor β recruitment and were associated with a strong synergistic effect with thrombin on Src activation. Taken together, these results indicate that 17β-estradiol can modulate platelet function by exercising a proaggregating role.
Collapse
Affiliation(s)
- Laura Moro
- Department of Medical Sciences, DISCAFF, University A. Avogadro, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Zivadinovic D, Gametchu B, Watson CS. Membrane estrogen receptor-alpha levels in MCF-7 breast cancer cells predict cAMP and proliferation responses. Breast Cancer Res 2004; 7:R101-12. [PMID: 15642158 PMCID: PMC1064104 DOI: 10.1186/bcr958] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 08/18/2004] [Accepted: 10/07/2004] [Indexed: 05/01/2023] Open
Abstract
Introduction 17β-estradiol (E2) can rapidly induce cAMP production, but the conditions under which these cAMP levels are best measured and the signaling pathways responsible for the consequent proliferative effects on breast cancer cells are not fully understood. To help resolve these issues, we compared cAMP mechanistic responses in MCF-7 cell lines selected for low (mERlow) and high (mERhigh) expression of the membrane form of estrogen receptor (mER)-α, and thus addressed the receptor subform involved in cAMP signaling. Methods MCF-7 cells were immunopanned and subsequently separated by fluorescence activated cell sorting into mERhigh (mER-α-enriched) and mERlow (mER-α-depleted) populations. Unique (compared with previously reported) incubation conditions at 4°C were found to be optimal for demonstrating E2-induced cAMP production. Time-dependent and dose-dependent effects of E2 on cAMP production were determined for both cell subpopulations. The effects of forskolin, 8-CPT cAMP, protein kinase A inhibitor (H-89), and adenylyl cyclase inhibitor (SQ 22,536) on E2-induced cell proliferation were assessed using the crystal violet assay. Results We demonstrated a rapid and transient cAMP increase after 1 pmol/l E2 stimulation in mERhigh cells; at 4°C these responses were much more reliable and robust than at 37°C (the condition most often used). The loss of cAMP at 37°C was not due to export. 3-Isobutyl-1-methylxanthine (IBMX; 1 mmol/l) only partially preserved cAMP, suggesting that multiple phosphodiesterases modulate its level. The accumulated cAMP was consistently much higher in mERhigh cells than in mERlow cells, implicating mER-α levels in the process. ICI172,780 blocked the E2-induced response and 17α-estradiol did not elicit the response, also suggesting activity through an estrogen receptor. E2 dose-dependent cAMP production, although biphasic in both cell types, was responsive to 50-fold higher E2 concentrations in mERhigh cells. Proliferation of mERlow cells was stimulated over the whole range of E2concentrations, whereas the number of mERhigh cells was greatly decreased at concentrations above 1 nmol/l, suggesting that estrogen over-stimulation can lead to cell death, as has previously been reported, and that mER-α participates. E2-mediated activation of adenylyl cyclase and downstream participation of protein kinase A were shown to be involved in these responses. Conclusion Rapid mER-α-mediated nongenomic signaling cascades generate cAMP and downstream signaling events, which contribute to the regulation of breast cancer cell number.
Collapse
Affiliation(s)
- Dragoslava Zivadinovic
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bahiru Gametchu
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cheryl S Watson
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
148
|
Hewitt SC, Collins J, Grissom S, Deroo B, Korach KS. Global uterine genomics in vivo: microarray evaluation of the estrogen receptor alpha-growth factor cross-talk mechanism. Mol Endocrinol 2004; 19:657-68. [PMID: 15528273 DOI: 10.1210/me.2004-0142] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cross-talk between growth factor receptors and the estrogen receptor (ER) has been proposed as a signaling mechanism in estrogen target tissues, with ER(alpha) as a direct target of growth factor receptor-activated signals, leading to regulation of estrogen target genes and estrogen-like biological responses to growth factors. We evaluated whether global genomic changes in the mouse uterus in response to epidermal growth factor or IGF-I mimic those of estradiol (E2), reflecting the cross-talk mechanism. Overlapping responses to growth factors and E2 were expected in the wild type (WT) whereas no response was expected in mice lacking ER(alpha) (ER(alpha) knockout). Surprisingly, although most of the E2 response in the WT also occurred after growth factor treatment, some genes were induced only by E2. Second, although E2 did not induce gene changes in the ER(alpha) knockout, the growth factor response was almost indistinguishable from that of the WT. Differences in response of some genes to IGF-I or epidermal growth factor indicated selective regulation mechanisms, such as phosphatidylinositol 3-kinase or MAPK-dependent responses. The robust ER(alpha)-independent genomic response to growth factor observed here is surprising considering that the biological growth response is ER(alpha) dependent. We propose two mechanisms as alternatives to the cross-talk mechanism for uterine gene regulation. First, E2 increases uterine growth factors, which activate downstream signaling cascades, resulting in gene regulation. Second, growth factors and estrogen regulate similar genes. Our results suggest that the estrogen response in the uterus involves E2-specific ER(alpha)-mediated responses as well as responses resulting from convergence of growth factor and ER-initiated activities.
Collapse
Affiliation(s)
- Sylvia Curtis Hewitt
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, PO Box 12233, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | |
Collapse
|
149
|
Kow LM, Pfaff DW. The membrane actions of estrogens can potentiate their lordosis behavior-facilitating genomic actions. Proc Natl Acad Sci U S A 2004; 101:12354-7. [PMID: 15302933 PMCID: PMC514479 DOI: 10.1073/pnas.0404889101] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The membrane actions of estrogens can facilitate their genomic actions. To determine whether this facilitation bears on CNS mechanisms for estrogen-dependent behaviors, ovariectomized rats were subjected to a two-pulse treatment of estrogen directly in the hypothalamic ventromedial nucleus. Two days later, each rat was given progesterone and then tested for lordosis behavior, the induction of which requires the genomic actions of estrogen. When estrogen was given in both pulses (15 min to 2 h duration, and 5 h apart) lordosis was induced. Based on results from studies on neuroblastoma cells, we hypothesized that the membrane actions of estrogen in the first pulse would potentiate the genomic actions of estrogen in the second. This hypothesis was confirmed with the use of a membrane-impermeable estrogen. However, surprisingly, the order of the pulses could be reversed and still achieve lordosis behavior induction. Finally, activators of protein kinase A or PKC were effective substitutes for the membrane-limited pulse of estrogen. Thus, estrogen-induced membrane actions in the hypothalamus can potentiate its lordosis-inducing genomic actions on behavior and may be mediated by signaling pathways involving the activation of protein kinase A and PKC.
Collapse
Affiliation(s)
- Lee-Ming Kow
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA.
| | | |
Collapse
|
150
|
Malyala A, Pattee P, Nagalla SR, Kelly MJ, Rønnekleiv OK. Suppression subtractive hybridization and microarray identification of estrogen-regulated hypothalamic genes. Neurochem Res 2004; 29:1189-200. [PMID: 15176476 DOI: 10.1023/b:nere.0000023606.13670.1d] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The gonadal steroid estrogen is a pleiotropic hormone that has multiple effects on numerous cellular functions. One of estrogen's major targets is the brain, where the steroid not only affects growth, differentiation, and survival of neurons, but also regulates cell excitability. Because estrogen modulates multiple, overlapping signaling pathways, it has been difficult to scrutinize the transcriptional activity of the steroid. Therefore, we still lack a global picture of how different genes interact and are regulated by estrogen. Herein we report the use of suppression subtractive hybridization followed by custom microarray analysis of thousands of genes that are differentially expressed during the negative feedback phase of the female reproductive cycle. We have found a number of key transcripts that are regulated by estrogen and contribute to the alteration in synaptic transmission and hence excitability of hypothalamic neurons (e.g., GABA neurons). These include gec-1, GABA(B)R2, PI3 kinase subunit p55gamma, and a number of proteins containing pleckstrin homology domains that are critical for plasma membrane targeting. Studies are underway to refine our analysis to individual nuclei and individual cells. However, what has emerged from this highly sensitive microarray analysis is that estrogen affects neuronal plasticity in hypothalamic neurons not only by transcription of new membrane proteins (e.g., receptors and channels), but also by altering expression of downstream signaling molecules and proteins involved in neurosecretory pathways.
Collapse
Affiliation(s)
- Anna Malyala
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | | | | | |
Collapse
|