Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Appl Pharmacol ; 213(2): 87-97, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16257430

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that stimulates transcription directed by xenobiotic response elements upstream of target genes. Recently, AhR ligands were reported to induce formation of an AhR-estrogen receptor (ER) complex, which can bind to estrogen response elements (EREs) and stimulate transcription of ER target genes. Presently, we investigate the effect of the AhR ligands 3-methylcholanthrene (3MC), 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 3,3',4,4',5-pentachlorobiphenyl (BZ126) on ERE-regulated luciferase reporter activity and endogenous ER target gene expression. In MCF-7 human breast cancer cells, 3MC induced transcription of ER reporter genes containing native promoter sequences of the ER-responsive genes complement 3 and pS2 and heterologous promoters regulated by isolated EREs. Dose-response studies revealed that the concentration of 3MC required to half-maximally activate transcription (EC(50)) was >100-fold higher for an ER reporter (27-57 muM) than for an AhR reporter (86-250 nM) in both MCF-7 cells and in human endometrial cancer Ishikawa cells. 3MC also stimulated expression of the endogenous ER target genes amphiregulin, cathepsin D and progesterone receptor, albeit to a much lower extent than was achieved following stimulation with 17beta-estradiol. In Ishikawa cells, 3MC, but not BZ126 or TCDD, stimulated ERalpha-dependent reporter activity but did not induce expression of endogenous ER target genes. Finally, studies carried out in the AhR-positive rat hepatoma cell line 5L and the AhR-deficient variant BP8 demonstrated that ER reporter activity could be induced by 3MC in a manner that was independent of AhR and thus distinct from the AhR-ER 'hijacking' mechanism described recently. 3MC may thus elicit estrogenic activity by multiple mechanisms.


Subject(s)
Estrogen Receptor alpha/metabolism , Gene Expression Regulation/drug effects , Methylcholanthrene/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Selective Estrogen Receptor Modulators/pharmacology , Transcriptional Activation/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Endometrial Neoplasms/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Female , Genes, Reporter/drug effects , Genes, Reporter/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Ligands , Liver Neoplasms/metabolism , Polychlorinated Biphenyls/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Rats , Receptors, Aryl Hydrocarbon/metabolism , Transcriptional Activation/physiology
2.
Toxicol Appl Pharmacol ; 199(3): 275-84, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15364543

ABSTRACT

The transcription factors peroxisome proliferator-activated receptor (PPAR) and signal transducer and activator of transcription 5 (STAT5) activate genes involved in fatty acid metabolism (PPARalpha) and adipogenesis (PPARgamma) and mediate hormonal responses important for body growth, liver gene expression, and mammary gland development (STAT5a and STAT5b). These seemingly disparate pathways are subject to mutually inhibitory crosstalk, with growth hormone (GH)-activated STAT5 able to inhibit PPAR-regulated gene transcription by approximately 80%, and conversely, ligand-activated PPAR able to inhibit STAT5-regulated transcription to a similar degree. Given the co-expression of PPAR and STAT5 in multiple tissues, we investigated whether one of the factors dominates the inhibitory crosstalk. A PPAR-responsive Renilla luciferase reporter was constructed and used to monitor PPAR transcriptional activity in COS-1 cells co-transfected with a STAT5 firefly luciferase reporter. In cells co-stimulated with GH and a PPAR agonist, STAT5b inhibited expression of the PPAR-regulated Renilla luciferase reporter, whereas PPARalpha and PPARgamma inhibited transcription of the STAT5b-regulated firefly luciferase reporter. The extent of the inhibitory crosstalk was dependent on the relative levels of expression of each transcription factor and on the relative concentrations of GH and PPAR agonist. Dose-response studies revealed that STAT5b was inhibited at an approximately 7-fold lower concentration of the PPARgamma ligand troglitazone than was required for activation of PPARgamma, indicating that only a portion of cellular PPARgamma is needed for STAT5b inhibition. Similarly, mono-(2-ethylhexyl)phthalate (MEHP), a reproductive toxicant and primary metabolite of the environmental chemical di-(2-ethylhexyl)phthalate (DEHP), inhibited STAT5b transcriptional activity with an EC50 value of 1.1 microM, corresponding to an approximately 10-fold lower concentration than required for activation of PPARgamma-dependent transcription. We conclude that the cross-inhibition between PPAR and STAT5 proceeds in a simultaneous, bidirectional manner. Exposure to phthalates and other environmental chemical activators of PPARs may thus lead to alteration of hormone-induced, STAT5-regulated gene expression in tissues such as liver, fat and breast, where both transcription factors are expressed. Conversely, STAT5-activating hormones and cytokines may modulate the responsiveness of PPARs to their foreign chemical ligands.


Subject(s)
DNA-Binding Proteins/physiology , Receptor Cross-Talk/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Chromans/pharmacology , Coleoptera/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , DNA-Binding Proteins/agonists , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genes, Reporter/genetics , Ligands , Luciferases/genetics , Mice , Milk Proteins/agonists , Milk Proteins/biosynthesis , Phthalic Acids/pharmacology , Plasmids/genetics , Rats , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/biosynthesis , STAT5 Transcription Factor , Thiazolidinediones/pharmacology , Trans-Activators/agonists , Trans-Activators/biosynthesis , Transcription Factors/agonists , Transcription Factors/biosynthesis , Transfection , Troglitazone
3.
Toxicol Sci ; 80(1): 151-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15071170

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors that activate target genes involved in lipid metabolism, energy homeostasis, and cell differentiation in response to diverse compounds, including environmental chemicals. The liver-expressed receptor PPARalpha mediates peroxisome proliferative responses associated with rodent hepatocarcinogenesis. Previous studies have established that certain perfluorooctanesulfonamide-based chemicals (PFOSAs) alter lipid metabolism, are hepatic peroxisome proliferators, and induce hepatocellular adenoma formation in rodents, suggesting that they activate PPARalpha. The present study investigates this question and characterizes the activation of mouse and human PPARalpha by PFOSAs. Perfluorooctanesulfonate (PFOS), an end-stage metabolite common to several PFOSAs, was found to activate both mouse and human PPARalpha in a COS-1 cell-based luciferase reporter trans-activation assay. Half-maximal activation (EC50) occurred at 13-15 microM PFOS, with no significant difference in the responsiveness of mouse and human PPARalpha. Mouse and human PPARalpha were activated by perfluorooctanesulfonamide (FOSA) over a similar concentration range; however, cellular toxicity precluded an accurate determination of EC50 values. Studies of 2-N-ethylperfluorooctanesulfonamido ethanol were less informative due to its insolubility. These findings were verified in an FAO rat hepatoma cell line that stably expresses PPARalpha, where the endogenous PPARalpha target genes peroxisomal bifunctional enzyme and peroxisomal 3-ketoacyl-CoA thiolase were activated up to approximately 10-20-fold by PFOS and FOSA. The interactions of PPARalpha with PFOS and FOSA, and the potential of these chemicals for activation of unique sets of downstream target genes, may help explain the diverse biological effects exhibited by PFOSAs and may aid in the evaluation of human and environmental risks associated with exposure to this important class of fluorochemicals.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , PPAR alpha/biosynthesis , Peroxisome Proliferators/pharmacology , Sulfonamides/toxicity , Alkanesulfonic Acids/chemistry , Animals , Blotting, Western , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Fluorocarbons/chemistry , Gene Expression Regulation , Genes, Reporter , Humans , Hydrocarbons, Fluorinated , Luciferases/genetics , Mice , PPAR alpha/genetics , Peroxisome Proliferators/chemistry , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Solubility , Sulfonamides/chemistry , Transcriptional Activation
4.
Mol Pharmacol ; 64(2): 355-64, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12869640

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor (PPAR) is activated by a diverse group of acidic ligands, including many peroxisome proliferator chemicals present in the environment. Janus tyrosine kinase-signal transducer and activator of transcription (JAK-STAT) signaling is activated by multiple cytokines and hormones and leads to the translocation of dimerized STAT proteins to the nucleus where they activate transcription of target genes. Previous studies have shown that growth hormone (GH)-activated STAT5b can inhibit PPAR-regulated transcription. Here, we show that this inhibitory cross-talk is mutual, and that GH-induced, STAT5b-dependent beta-casein promoter-luciferase reporter gene transcription can be inhibited up to approximately 80% by ligand-activated PPARalpha or PPARgamma. Dose-response experiments showed a direct relationship between the extent of PPAR activation and the degree of inhibition of STAT5-regulated transcription. PPAR did not block STAT5b tyrosine phosphorylation or inhibit DNA-binding activity. Both PPARs inhibited the transcriptional activity of a constitutively active STAT5b mutant, indicating that inhibition occurs downstream of the GH-stimulated STAT5 activation step. Transcriptionally inactive, dominant-negative PPAR mutants did not block STAT5b inhibition by wild-type PPAR, indicating that PPAR target gene transcription is not required. PPARalpha retained its STAT5b inhibitory activity in the presence of the histone deacetylase inhibitor trichostatin, indicating that enhanced histone deacetylase recruitment does not contribute to STAT5b inhibition. PPARalpha lacking the ligand-independent AF-1 trans-activation domain failed to inhibit STAT5b, highlighting the importance of the AF-1 region in STAT5-PPAR inhibitory cross-talk. These findings demonstrate the bidirectionality of cross-talk between the PPAR and STAT pathways and provide a mechanism whereby exposure to environmental chemical activators of PPAR can suppress expression of GH target genes.


Subject(s)
DNA-Binding Proteins/genetics , Milk Proteins , Receptors, Cytoplasmic and Nuclear/physiology , Trans-Activators/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Down-Regulation , Histone Deacetylases/metabolism , Ligands , Phosphorylation , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Interferon/metabolism , STAT5 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/genetics , Transcriptional Activation , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...