Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Steroids ; 78(1): 15-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123738

ABSTRACT

Dehydroepiandrosterone (DHEA) levels were reported to associate with increased breast cancer risk in postmenopausal women, but some carcinogen-induced rat mammary tumor studies question this claim. The purpose of this study was to determine how DHEA and its metabolites affect estrogen receptors α or ß (ERα or ERß)-regulated gene transcription and cell proliferation. In transiently transfected HEK-293 cells, androstenediol, DHEA, and DHEA-S activated ERα. In ERß transfected HepG2 cells, androstenedione, DHEA, androstenediol, and 7-oxo DHEA stimulated reporter activity. ER antagonists ICI 182,780 (fulvestrant) and 4-hydroxytamoxifen, general P450 inhibitor miconazole, and aromatase inhibitor exemestane inhibited activation by DHEA or metabolites in transfected cells. ERß-selective antagonist R,R-THC (R,R-cis-diethyl tetrahydrochrysene) inhibited DHEA and DHEA metabolite transcriptional activity in ERß-transfected cells. Expression of endogenous estrogen-regulated genes: pS2, progesterone receptor, cathepsin D1, and nuclear respiratory factor-1 was increased by DHEA and its metabolites in an ER-subtype, gene, and cell-specific manner. DHEA metabolites, but not DHEA, competed with 17ß-estradiol for ERα and ERß binding and stimulated MCF-7 cell proliferation, demonstrating that DHEA metabolites interact directly with ERα and ERßin vitro, modulating estrogen target genes in vivo.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/physiology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Androstenediol/pharmacology , Androstenedione/pharmacology , Animals , Cell Line , Cell Proliferation , Cricetinae , Dehydroepiandrosterone/pharmacology , Estradiol/pharmacology , Estradiol/physiology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Female , Genes, Reporter , Humans , Inhibitory Concentration 50 , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Miconazole/pharmacology , Response Elements , Transcriptional Activation , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
2.
Arch Toxicol ; 86(4): 633-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22105178

ABSTRACT

Endothelial dysfunction precedes cardiovascular disease and is accompanied by mitochondrial dysfunction. Here we tested the hypothesis that diesel exhaust particulate extracts (DEPEs), prepared from a truck run at different speeds and engine loads, would inhibit genomic estrogen receptor activation of nuclear respiratory factor-1 (NRF-1) transcription in human umbilical vein endothelial cells (HUVECs). Additionally, we examined how DEPEs affect NRF-1-regulated TFAM expression and, in turn, Tfam-regulated mtDNA-encoded cytochrome c oxidase subunit I (COI, MTCO1) and NADH dehydrogenase subunit I (NDI) expression as well as cell proliferation and viability. We report that 17ß-estradiol (E(2)), 4-hydroxytamoxifen (4-OHT), and raloxifene increased NRF-1 transcription in HUVECs in an ER-dependent manner. DEPEs inhibited NRF-1 transcription, and this suppression was not ablated by concomitant treatment with E(2), 4-OHT, or raloxifene, indicating that the effect was not due to inhibition of ER activity. While E(2) increased HUVEC proliferation and viability, DEPEs inhibited viability but not proliferation. Resveratrol increased NRF-1 transcription in an ER-dependent manner in HUVECs, and ablated DEPE inhibition of basal NRF-1 expression. Given that NRF-1 is a key nuclear transcription factor regulating genes involved in mitochondrial activity and biogenesis, these data suggest that DEPEs may adversely affect mitochondrial function leading to endothelial dysfunction and resveratrol may block these effects.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Nuclear Respiratory Factor 1/genetics , Particulate Matter/toxicity , Transcription, Genetic/drug effects , Vehicle Emissions/toxicity , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Antagonism , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/physiology , Estradiol/toxicity , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mitochondria/drug effects , Mitochondria/physiology , Nuclear Respiratory Factor 1/metabolism , Raloxifene Hydrochloride/toxicity , Resveratrol , Stilbenes/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/toxicity
3.
FASEB J ; 25(4): 1402-16, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21233487

ABSTRACT

Little is known about endogenous estrogen receptor ß (ERß) gene targets in human breast cancer. We reported that estradiol (E(2)) induces nuclear respiratory factor-1 (NRF-1) transcription through ERα in MCF-7 breast cancer cells. Here we report that 4-hydroxytamoxifen (4-OHT), with an EC(50) of ~1.7 nM, increases NRF-1 expression by recruiting ERß, cJun, cFos, CBP, and RNA polymerase II to and dismissing NCoR from the NRF1 promoter. Promoter deletion and transient transfection studies showed that the estrogen response element (ERE) is essential and that an adjacent AP-1 site contributes to maximal 4-OHT-induced NRF-1 transcription. siRNA knockdown of ERß revealed that ERß inhibits basal NRF-1 expression and is required for 4-OHT-induced NRF-1 transcription. An AP-1 inhibitor blocked 4-OHT-induced NRF-1 expression. The 4-OHT-induced increase in NRF-1 resulted in increased transcription of NRF-1 target CAPNS1 but not CYC1, CYC2, or TFAM despite increased NRF-1 coactivator PGC-1α protein. The absence of TFAM induction corresponds to a lack of Akt-dependent phosphorylation of NRF-1 with 4-OHT treatment. Overexpression of NRF-1 inhibited 4-OHT-induced apoptosis and siRNA knockdown of NRF-1 increased apoptosis, indicating an antiapoptotic role for NRF-1. Overall, NRF-1 expression and activity is regulated by 4-OHT via endogenous ERß in MCF-7 cells.


Subject(s)
Estrogen Receptor beta/metabolism , Nuclear Respiratory Factor 1/genetics , Tamoxifen/analogs & derivatives , Transcription Factor AP-1/metabolism , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Estrogen Receptor alpha/physiology , Humans , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Retinoids/pharmacology , Signal Transduction , Tamoxifen/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Transcription, Genetic/drug effects
4.
Mol Endocrinol ; 22(3): 609-22, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18048642

ABSTRACT

Estrogen has direct and indirect effects on mitochondrial activity, but the mechanisms mediating these effects remain unclear. Others reported that long-term estradiol (E(2)) treatment increased nuclear respiratory factor-1 (NRF-1) protein in cerebral blood vessels of ovariectomized rats. NRF-1 is a transcription factor that regulates the expression of nuclear-encoded mitochondrial genes, e.g. mitochondrial transcription factor A (TFAM), that control transcription of the mitochondrial genome. Here we tested the hypothesis that E(2) increases NRF-1 transcription resulting in a coordinate increase in the expression of nuclear- and mitochondrial- encoded genes and mitochondrial respiratory activity. We show that E(2) increased NRF-1 mRNA and protein in MCF-7 breast and H1793 lung adenocarcinoma cells in a time-dependent manner. E(2)-induced NRF-1 expression was inhibited by the estrogen receptor (ER) antagonist ICI 182,780 and actinomycin D but not by phosphoinositide-3 kinase and MAPK inhibitors, indicating a genomic mechanism of E(2) regulation of NRF-1 transcription. An estrogen response element (ERE) in the NRF-1 promoter bound ER alpha and ER beta in vitro, and E(2) induced ER alpha and ER beta recruitment to this ERE in chromatin immunoprecipitation assays in MCF-7 cells. The NRF-1 ERE activated reporter gene expression in transfected cells. Small interfering RNA to ER alpha and ER beta revealed that ER alpha mediates E(2)-induced NRF-1 transcription. The E(2)-induced increase in NRF-1 was followed by increased TFAM and the transcription of Tfam-regulated mitochondrial DNA-encoded COI and NDI genes and increased mitochondrial biogenesis. Knockdown of NRF-1 blocked E(2) stimulation of mitochondrial biogenesis and activity, indicating a mechanism by which estrogens regulate mitochondrial function by increasing NRF-1 expression.


Subject(s)
Estradiol/pharmacology , Mitochondria/drug effects , Nuclear Respiratory Factor 1/biosynthesis , Transcription, Genetic/physiology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA, Mitochondrial/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Fulvestrant , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Nitriles/pharmacology , Nuclear Respiratory Factor 1/genetics , Phenols , Promoter Regions, Genetic/drug effects , Propionates/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics
5.
Am J Physiol Cell Physiol ; 293(2): C566-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17360813

ABSTRACT

Postflight orthostatic intolerance (POI) was reported to be higher in female than male astronauts and may result from sex-dependent differences in endothelial cell (EC) barrier permeability. Here the effect of 17beta-estradiol (E(2)) and dihydrotestosterone (DHT) on the expression of the tight junction protein occludin, EC barrier function, and MAPK activation over time was tested after subjecting human umbilical vein EC (HUVEC) to brief hypergravity identical to that experienced by astronauts during liftoff (LO) into space. After LO hypergravity, HUVEC showed a time-dependent decrease in occludin correlating with an increase in paracellular permeability and a decrease in transendothelial electrical resistance, indicating a decrease in EC barrier function. LO hypergravity inhibited MAPK activation, which remained suppressed 4 h after LO. Inhibition of MAPK activation correlated with decreased phosphotyrosine occludin, decreased cytochrome-c oxidase activity, and increased paracellular permeability, suggesting a mechanism by which LO hypergravity decreased EC barrier function. Time-dependent differences in MAPK activation, decreased occludin, and EC barrier function between HUVEC treated with E(2) vs. DHT were observed. HUVEC showed delayed activation of MAPK with DHT, i.e., 4 h rather than 2 h for E(2), which correlated with decreased paracellular permeability and the observed sex differences in POI in astronauts. These data temporally separate E(2) and DHT effects in HUVEC and provide evidence for the possible protective roles of sex steroids on EC function after brief exposure to low hypergravity.


Subject(s)
Capillary Permeability , Dihydrotestosterone/metabolism , Endothelial Cells/metabolism , Estradiol/metabolism , Hypergravity , Mitogen-Activated Protein Kinases/metabolism , Tight Junctions/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Dihydrotestosterone/pharmacology , Electric Impedance , Electron Transport Complex IV/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation , Estradiol/pharmacology , Female , Humans , Hypotension, Orthostatic/metabolism , Male , Membrane Proteins/metabolism , Occludin , Phosphorylation , Prostaglandin-Endoperoxide Synthases/metabolism , Sex Factors , Space Flight , Tight Junctions/drug effects , Time Factors , Tyrosine/metabolism
6.
Appl Microbiol Biotechnol ; 74(6): 1256-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17318543

ABSTRACT

The production of estrogen receptors (ER) in cultured insect cells is advantageous because these cells are relatively easy to culture and they perform post-translation modifications necessary for protein stability and function. There are three options for protein expression in insect cells: transient transfection, lytic baculovirus infection, or transfection followed by selection to create stable cell lines. Stable transfection has been promoted to be advantageous for the production of recombinant proteins because no re-infection is required, which might provide better lot-to-lot reproducibility in protein production. In this paper, we demonstrate that lytic baculovirus infection of Sf21 cells yields approximately tenfold more bioactive ERbeta than cells stably transformed with pIZ/V5-His plasmid under OpIE2 promoter. We provide the first evidence that stable expression of recombinant human ERbeta decreases the proliferation of Sf21 cells by inhibition of cell replication in a ligand-independent manner. These results mirror findings in breast cancer cells showing that an increase in ERbeta expression decreases cell proliferation. We conclude that baculovirus infection of Sf21 cells is better for human ERbeta production than stable-transformation of Sf21 cells.


Subject(s)
Baculoviridae/growth & development , Estrogen Receptor beta/metabolism , Animals , Baculoviridae/genetics , Cell Line , Cell Proliferation , Cell Survival/genetics , Estrogen Receptor beta/genetics , Female , Humans , Immunoblotting , Microscopy, Fluorescence , Plasmids/genetics , Promoter Regions, Genetic/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spodoptera , Transfection
7.
Endocr Relat Cancer ; 13(1): 113-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16601283

ABSTRACT

The higher frequency of lung adenocarcinoma in women smokers than in men smokers suggests a role for gender-dependent factors in the etiology of lung cancer. We evaluated estrogen receptor (ER) alpha and beta expression and activity in human lung adenocarcinoma cell lines and normal lung fibroblasts. Full-length ERalpha and ERbeta proteins were expressed in all cell lines with higher ERbeta than ERalpha. Although estradiol (E(2)) binding was similar, E(2) stimulated proliferation only in cells from females, and this response was inhibited by anti-estrogens 4-hydroxytamoxifen (4-OHT) and ICI 182,780. In contrast, E(2) did not stimulate replication of lung adenocarcinoma cells from males and 4-OHT or ICI did not block cell proliferation. Similarly, transcription of an estrogen response element-driven reporter gene was stimulated by E(2) in lung adenocarcinoma cells from females, but not males. Progesterone receptor (PR) expression was increased by E(2) in two out of five adenocarcinoma cell lines from females, but none from males. E(2) decreased E-cadherin protein expression in some of the cell lines from females, as it did in MCF-7 breast cancer cells, but not in the cell lines from males. Thus, ERalpha and ERbeta expression does not correlate with the effect of ER ligands on cellular activities in lung adenocarcinoma cells. On the other hand, coactivator DRIP205 expression was higher in lung adenocarcinoma cells from females versus males and higher in adenocarcinoma cells than in normal human bronchial epithelial cells. DRIP205 and other ER coregulators may contribute to differences in estrogen responsiveness between lung adenocarcinoma cells in females and males.


Subject(s)
Adenocarcinoma/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cadherins/metabolism , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/pathology , Mediator Complex Subunit 1 , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , Response Elements , Sex , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured
8.
Endocrinology ; 145(1): 149-60, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14500565

ABSTRACT

Estrogen receptor beta (ERbeta) has been reported to have lower estradiol (E2)-induced transcriptional activity than human (h)ERalpha from estrogen response element (ERE)-driven reporters in transiently transfected cells. Conflicting data for activities of full-length and short hERbeta [hERbeta1, 530 amino acids (aa); and hERbeta1s, 477aa] have been reported. To test the hypothesis that hERbeta1 has higher transcriptional activity than hERbeta1s, we compared E2, 2,3-bis(4-hydroxyphenyl)propionitrile (a selective ERbeta agonist), and resveratrol-induced transcription by hERbeta1, hERbeta1s, and rat (r) ERbeta with hERalpha on different EREs in transiently transfected CHO-K1 and HEC-1A cells. Our results demonstrate for the first time that hERbeta1 has similar E2-induced activity to hERalpha and greater activity than rERbeta or hERbeta1s on a consensus palindromic ERE, either as a single or double copy; a minimal ERE; and the nonpalindromic pS2 ERE. 2,3-Bis(4-hydroxyphenyl)propionitrile showed greater efficacy with hERbeta1 and hERbeta1s than for rERbeta or hERalpha. We found that transcriptional differences between the ERbeta isoforms and ERalpha depend on the ERE sequence, confirming that the DNA sequence bound by ER is an allosteric effector of ER action. For the minimal 13-bp ERE and the pS2 ERE, the increase in transcriptional activity with hERbeta1 correlated with increased binding affinity. Coactivators steroid receptor coactivator-1 and cAMP response element binding protein-binding protein synergistically activated hERalpha and ERbeta transcription and showed reduced efficacy with rERbeta and hERbeta1s, suggesting a role for the N terminus of ERbeta1 in coactivator interaction. Collectively, these data indicate that the cellular expression of ERbeta isoforms may differentially impact ERE-regulated target gene expression in a ligand-dependent manner.


Subject(s)
Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transcriptional Activation/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CHO Cells , Cricetinae , Estrogen Receptor alpha , Estrogen Receptor beta , Estrogens/pharmacology , Humans , Isomerism , Ligands , Rats , Receptors, Estrogen/chemistry , Response Elements/genetics , Resveratrol , Stilbenes/pharmacology , Transcriptional Activation/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...