Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Materials (Basel) ; 17(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38998228

ABSTRACT

The 1:1 resveratrol-piperazine cocrystal was successfully synthesized and scaled-up to 300 g scale with the mechanochemical method, as a result of investigating key process parameters, namely the solvent and the grinding time. The use of water, ethanol or ethanol-water mixtures and reaction times up to 50 min were evaluated relative to the dry grinding process. Cocrystal formation and purity were monitored through X-ray diffraction and calorimetry measurements. The dry grinding resulted in an incomplete cocrystal formation, while the use of water or water-ethanol mixture yielded a monohydrate solid phase. Pure ethanol was found to be the optimal solvent for large-scale cocrystallization, as it delivered cocrystals with high crystallinity and purity after 10-30 min grinding time at the laboratory scale. Notably, a relatively fast reaction time (30-60 min) was sufficient for the completion of cocrystallization at larger scales, using a planetary ball mill and a plant reactor. Also, the obtained cocrystal increases the aqueous solubility of resveratrol by 6%-16% at pH = 6.8. Overall, this study highlights the potential of solvent-assisted mechanochemical synthesis as a promising new approach for the efficient production of pure resveratrol-piperazine cocrystals.

2.
Bioorg Med Chem ; 21(14): 4120-31, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23735829

ABSTRACT

A collection of compounds, structurally related to the anticancer drug tamoxifen, used in breast cancer therapy, were designed and synthesized as potential anticancer agents. McMurry coupling reaction was used as the key synthetic step in the preparation of these analogues and the structural assignment of E, Z isomers was determined on the basis of 2D-NOESY experiments. The compounds were evaluated for their antiproliferative activity on breast cancer (MCF-7), cervix adenocarcinoma (HeLa) and biphasic mesothelioma (MSTO-211H) human tumor cell lines. The estrogen like properties of the novel compounds were compared with those of the untreated controls using an estrogen responsive element-based (ERE) luciferase reporter assay and compared to 17ß-estradiol (E2). Finally, with the aim to correlate the antiproliferative activity with an intracellular target(s), the effect on relaxation activity of DNA topoisomerases I and II was assayed.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Tamoxifen/chemical synthesis , Tamoxifen/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Endometrial Neoplasms/drug therapy , Female , Humans , Molecular Structure , Receptors, Estrogen/metabolism , Tamoxifen/chemistry
3.
Mol Endocrinol ; 26(9): 1542-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22865928

ABSTRACT

The function of the aryl hydrocarbon receptor (AhR) in mediating the biological effect to environmental pollutants is well established. However, accumulated evidence indicates a wide range of physiological and pathological functions mediated by the AhR, suggesting the existence of endogenous AhR ligand(s). The nature of an AhR ligand remain elusive; however, it is known that the AhR is activated by several compounds, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin or the tryptophan photoproduct 6-formylindolo[3,2-b]carbazole. In this study, we show that physiological concentrations of tryptamine (TA) lead to induction of cytochrome P4501A1 transcription through an AhR-dependent mechanism. In addition, we show that activation of the AhR by TA requires a functional monoamino oxidase system, suggesting that TA acts as an AhR proligand possibly by converting to a high-affinity AhR ligand. Taken together, we show a possible mechanism, through which AhR signaling is activated by endogenous conversion of TA involving monoamine oxidases.


Subject(s)
Monoamine Oxidase/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tryptamines/pharmacology , 3T3-L1 Cells , Animals , Carbazoles/pharmacology , Chromatin Immunoprecipitation , Cytochrome P-450 CYP1A1/genetics , HT29 Cells , Humans , Mice , Monoamine Oxidase/genetics , Protein Binding , RNA Interference , Real-Time Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Mol Cell Endocrinol ; 362(1-2): 39-47, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22634562

ABSTRACT

The two estrogen receptor isoforms ERα and ERß mediate biological effects of estrogens, but are also targets for endocrine disruptive chemicals (EDCs), compounds that interfere with hormonal signaling. 3-Methylcholanthrene (3-MC) and dioxin (TCDD) are EDCs and prototypical aryl hydrocarbon receptor (AhR) agonists, and can inhibit ER signaling. However, in contrast to TCDD, 3-MC gives rise to metabolites with estrogenic properties. We compared gene expression profiles in HepG2 cells after exposure to 3-MC, TCDD, and the synthetic estrogen diethylstilbestrol (DES). Interestingly, we observed little overlap between the genetic networks activated by 3-MC and TCDD, two compounds sometimes considered as interchangeable AhR ligands. Like DES, 3-MC induced a number of ER-regulated genes and lead to recruitment of ERα to the promoters of such genes. Interestingly, in contrast to DES, the estrogenic effects exerted by 3-MC were exclusively observed in ERα, but not in ERß-expressing cells, suggesting ER isoform selectivity of 3-MC-derived metabolites.


Subject(s)
Endocrine Disruptors/pharmacology , Gene Regulatory Networks , Methylcholanthrene/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Chromatin Immunoprecipitation , Estrogen Receptor alpha/metabolism , Gene Expression Regulation/drug effects , Genes , Hep G2 Cells , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding , Real-Time Polymerase Chain Reaction , Transcriptional Activation/drug effects , Transcriptome
5.
Mol Endocrinol ; 25(12): 2017-28, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22016564

ABSTRACT

Glucose transporter 4 (Glut4) is an important regulator of cellular glucose uptake in adipose tissue and skeletal muscle. The estrogen receptors α and ß (ERα and ERß) have been shown to regulate Glut4. However, the regulatory mechanisms are unclear, and there are conflicting results about the effects of the two ER isoforms on Glut4 activity. In this study we investigated how the lack of either ER isoform affects Glut4 expression in differentiated mouse embryonic fibroblasts. Our results demonstrate that Glut4 transcription is markedly reduced in cells lacking ERß, both basally and upon induction by liver X receptor. These changes in Glut4 expression could not be explained by the lack of ERß as ligand-activated transcription factor. They were rather brought about by hypermethylation of one single CpG in the Glut4 promoter in the ERß-deficient cells. This CpG is part of an Sp1-binding site, and Sp1 binding was reduced by its methylation. Treatment with Sp1 inhibitor diminished Glut4 expression in wild-type, but not in ERß-deficient cells, suggesting that reduced recruitment of Sp1 to the Glut4 promoter is responsible for the differences in Glut4 expression. Reintroduction of ERß into ERß-deficient cells partly restored Glut4 transcription and stabilized low DNA methylation after treatment with the DNA demethylating agent 5-Aza-2'-deoxycytidine. Our findings demonstrate the involvement of DNA methylation in Glut4 regulation and imply a novel function for ERß in mediating epigenetic events and thereby regulating gene expression.


Subject(s)
Epigenesis, Genetic , Estrogen Receptor beta/metabolism , Glucose Transporter Type 4/genetics , Adipocytes/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , CpG Islands , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Gene Knockout Techniques , Glucose Transporter Type 4/metabolism , Liver X Receptors , Mice , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic
6.
PLoS One ; 6(10): e25201, 2011.
Article in English | MEDLINE | ID: mdl-21984905

ABSTRACT

XAP2 (also known as aryl hydrocarbon receptor interacting protein, AIP) is originally identified as a negative regulator of the hepatitis B virus X-associated protein. Recent studies have expanded the range of XAP2 client proteins to include the nuclear receptor family of transcription factors. In this study, we show that XAP2 is recruited to the promoter of ERα regulated genes like the breast cancer marker gene pS2 or GREB1 and negatively regulate the expression of these genes in MCF-7 cells. Interestingly, we show that XAP2 downregulates the E2-dependent transcriptional activation in an estrogen receptor (ER) isoform-specific manner: XAP2 inhibits ERα but not ERß-mediated transcription. Thus, knockdown of intracellular XAP2 levels leads to increased ERα activity. XAP2 proteins, carrying mutations in their primary structures, loose the ability of interacting with ERα and can no longer regulate ER target gene transcription. Taken together, this study shows that XAP2 exerts a negative effect on ERα transcriptional activity and may thus prevent ERα-dependent events.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Models, Biological , Protein Binding , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
7.
J Nutr ; 141(9): 1583-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21753063

ABSTRACT

Enterolactone (EL) is an enterolignan produced by gut microbiota from dietary plant lignans. Epidemiological and experimental studies suggest that EL and plant lignans may reduce the risk of breast and prostate cancer as well as cardiovascular disease. These effects are thought to at least in part involve modulation of estrogen receptor activity. Surprisingly little is known about the in vivo estrogenicity of EL. In the present study, we investigated the target tissues of EL, the genes affected by EL treatment, and the response kinetics. Following a single dose of EL, luciferase was significantly induced in reproductive and nonreproductive tissues of male and female 3xERE-luciferase mice, indicating estrogen-like activity. Microarray analysis revealed that EL regulated the expression of only 1% of 17ß-estradiol target genes in the uterus. The majority of these genes were traditional estrogen target genes, but also members of the circadian signaling pathway were affected. Kinetic analyses showed that EL undergoes rapid phase II metabolism and is efficiently excreted. In vivo imaging demonstrated that the estrogen response followed similar, fast kinetics. We conclude that EL activates estrogen signaling in both male and female mice and that the transient responses may be due to the fast metabolism of the compound. Lastly, EL may represent a link among diet, gut microbiota, and circadian signaling.


Subject(s)
4-Butyrolactone/analogs & derivatives , CLOCK Proteins/metabolism , Circadian Clocks/genetics , Estrogens/metabolism , Lignans/pharmacology , Phytoestrogens/pharmacology , Signal Transduction/drug effects , 4-Butyrolactone/blood , 4-Butyrolactone/pharmacology , Animals , CLOCK Proteins/genetics , Circadian Clocks/drug effects , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Lignans/blood , Liver/metabolism , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Ovariectomy , Protein Array Analysis , Random Allocation , Uterus/metabolism
8.
Toxicology ; 268(3): 132-8, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-19778576

ABSTRACT

The aryl hydrocarbon receptor (AhR), in complex with its binding partner ARNT, mediates the cellular response to xenobiotic compounds such as the environmental pollutant dioxin. In addition, the AhR has important regulatory roles in normal physiology. For instance, there is extensive data showing an intricate relationship between the AhR and estrogen receptor (ER) pathways. This review focuses on the regulatory roles of AhR and ARNT, beyond the response to xenobiotics. In particular, the effects of AhR agonists on the estrogen signaling pathways and the role of ARNT as a modulator of ER activity are discussed.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Receptors, Aryl Hydrocarbon/physiology , Receptors, Estrogen/physiology , Signal Transduction/physiology , Xenobiotics/toxicity , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/agonists , Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Humans , Methylcholanthrene/toxicity , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Estrogen/drug effects , Xenobiotics/metabolism
9.
Cell Mol Life Sci ; 66(24): 3873-94, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19669093

ABSTRACT

Together with the estrogen receptor (ER) alpha, estrogen receptor beta (ER beta ) mediates many of the physiological effects of estrogens. As ER beta is crucially involved in a variety of important physiological processes, its activity should be tightly regulated. ER beta regulation is achieved by hormone binding as well as by posttranslational modifications of the receptor. Furthermore, ER beta expression levels are under circadian control and can be regulated by DNA methylation of the ER beta promoter region. There are also a number of factors that can interfere with ER beta activity, such as phytoestrogens, endocrine disruptive chemicals, and growth factors. In this article, we outline different mechanisms of ER beta regulation and how they are implicated in various diseases. We also discuss how these insights might help to specifically target ER beta in drug design.


Subject(s)
Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Neoplasms/pathology , Alternative Splicing , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Phytoestrogens/metabolism , Protein Binding , Protein Processing, Post-Translational
11.
FEBS J ; 276(11): 3043-59, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19490107

ABSTRACT

Retinoic acid receptor (RAR) and retinoid X receptor are ligand-induced transcription factors that belong to the nuclear receptor family. The receptors are activated by small hydrophobic compounds, such as all-trans-retinoic acid and 9-cis-retinoic acid, respectively. Interestingly, these receptors are also targets for a number of exogenous compounds. In this study, we characterized the biological activity of the 9-cis-substituted retinoic acid metabolite, S-4-oxo-9-cis-13,14-dihydro-retinoic acid (S-4o9cDH-RA). The endogenous levels of this metabolite in wild-type mice and rats were found to be higher than those of all-trans-retinoic acid, especially in the liver. Using cell-based luciferase reporter systems, we showed that S-4o9cDH-RA activates the transcription of retinoic acid response element-containing genes in several cell types, both from a simple 2xDR5 element and from the promoter of the natural retinoid target gene RARbeta2. In addition, quantitative RT-PCR analysis demonstrated that S-4o9cDH-RA treatment significantly increases the endogenous mRNA levels of the RAR target gene RARbeta2. Utilizing a limited proteolytic digestion assay, we showed that S-4o9cDH-RA induces conformational changes to both RARalpha and RARbeta in the same manner as does all-trans-retinoic acid, suggesting that S-4o9cDH-RA is indeed an endogenous ligand for these receptors. These in vitro results were corroborated in an in vivo system, where S-4o9cDH-RA induced morphological changes similar to those of all-trans-retinoic acid in the developing chicken wing bud. When locally applied to the wing bud, S-4o9cDH-RA induced digit pattern duplications in a dose-dependent fashion. The results illustrate that S-4o9cDH-RA closely mimics all-trans-retinoic acid with regard to pattern respecification. Finally, using quantitative RT-PCR analysis, we showed that S-4o9cDH-RA induces the transcription of several retinoic acid-regulated genes in chick wing buds, including Hoxb8, RARbeta2, shh, Cyp26 and bmp2. Although S-4o9cDH-RA was less potent when compared with all-trans-retinoic acid, the findings clearly demonstrate that S-4o9cDH-RA has the capacity to bind and activate nuclear retinoid receptors and regulate gene transcription both in vitro and in vivo.


Subject(s)
Receptors, Retinoic Acid/metabolism , Signal Transduction/physiology , Tretinoin/analogs & derivatives , Tretinoin/metabolism , Animals , Cell Line , Cell Line, Tumor , Chick Embryo , Chickens , Electrophoresis, Polyacrylamide Gel , Gene Expression/drug effects , HeLa Cells , Humans , Limb Buds/drug effects , Limb Buds/metabolism , Luciferases/genetics , Luciferases/metabolism , Mice , Molecular Structure , Promoter Regions, Genetic/genetics , Rats , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoic Acid Receptor alpha , Signal Transduction/drug effects , Transfection , Tretinoin/chemistry , Tretinoin/pharmacology , Wings, Animal/drug effects , Wings, Animal/embryology
12.
J Mol Endocrinol ; 43(1): 1-10, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19211731

ABSTRACT

Endocrine disruption refers to the ability of chemicals to interfere with hormonal systems, and has raised considerable concern in recent years. Endocrine disruptive chemicals (EDCs) pose a documented risk to wildlife and have the potential to negatively influence human health. This review focuses on the molecular mechanisms of endocrine disruption and the possible involvement of EDCs in metabolic disorders. The first part describes the role of aryl hydrocarbon receptor (AhR) and nuclear receptors (NRs) in mediating effects of EDCs, in particular, how cross-talk between AhR and NR pathways can lead to endocrine disruption. The second part deals with how these receptors are involved in metabolic functions and how their targeting by EDCs can lead to disturbances in glucose and fat metabolism. The article illustrates that, although there is accumulating data on molecular mechanisms of EDC action as well as on EDC involvement in metabolic disorders, there is still a great demand for data that can unite the mechanistic and the toxicological/epidemiological observations.


Subject(s)
Endocrine Disruptors/toxicity , Metabolic Diseases/chemically induced , Animals , Endocrine Disruptors/poisoning , Humans , Receptors, Cell Surface/metabolism
13.
EXS ; 99: 289-323, 2009.
Article in English | MEDLINE | ID: mdl-19157066

ABSTRACT

Many toxic compounds exert their harmful effects by activating of certain receptors, which in turn leads to dysregulation of transcription. Some of these receptors are so called xenosensors. They are activated by external chemicals and evoke a cascade of events that lead to the elimination of the chemical from the system. Other receptors that are modulated by toxic substances are hormone receptors, particularly the ones of the nuclear receptor family. Some environmental chemicals resemble endogenous hormones and can falsely activate these receptors, leading to undesired activity in the cell. Furthermore, excessive activation of the xenosensors can lead to disturbances of the integrity of the system as well. In this chapter, the concepts of receptor-mediated toxicity and hormone disruption are introduced. We start by describing environmental chemicals that can bind to xenosensors and nuclear hormone receptors. We then describe the receptors most commonly targeted by environmental chemicals. Finally, the mechanisms by which receptor-mediated events can disrupt the system are depicted.


Subject(s)
Endocrine Disruptors/toxicity , Endocrine System/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Endocrine System/metabolism , Endocrine System/physiology , Humans , Models, Biological , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/drug effects , Xenobiotics/toxicity
14.
BMC Evol Biol ; 8: 219, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18655705

ABSTRACT

BACKGROUND: The origin of nuclear receptors (NRs) and the question whether the ancestral NR was a liganded or an unliganded transcription factor has been recently debated. To obtain insight into the evolution of the ligand binding ability of estrogen receptors (ER), we comparatively characterized the ER from the protochordate amphioxus (Branchiostoma floridae), and the ER from lamprey (Petromyzon marinus), a basal vertebrate. RESULTS: Extensive phylogenetic studies as well as signature analysis allowed us to confirm that the amphioxus ER (amphiER) and the lamprey ER (lampER) belong to the ER group. LampER behaves as a "classical" vertebrate ER, as it binds to specific DNA Estrogen Responsive Elements (EREs), and is activated by estradiol (E2), the classical ER natural ligand. In contrast, we found that although amphiER binds EREs, it is unable to bind E2 and to activate transcription in response to E2. Among the 7 natural and synthetic ER ligands tested as well as a large repertoire of 14 cholesterol derivatives, only Bisphenol A (an endocrine disruptor with estrogenic activity) bound to amphiER, suggesting that a ligand binding pocket exists within the receptor. Parsimony analysis considering all available ER sequences suggest that the ancestral ER was not able to bind E2 and that this ability evolved specifically in the vertebrate lineage. This result does not support a previous analysis based on ancestral sequence reconstruction that proposed the ancestral steroid receptor to bind estradiol. We show that biased taxonomic sampling can alter the calculation of ancestral sequence and that the previous result might stem from a high proportion of vertebrate ERs in the dataset used to compute the ancestral sequence. CONCLUSION: Taken together, our results highlight the importance of comparative experimental approaches vs ancestral reconstructions for the evolutionary study of endocrine systems: comparative analysis of extant ERs suggests that the ancestral ER did not bind estradiol and that it gained the ability to be regulated by estradiol specifically in the vertebrate lineage, before lamprey split.


Subject(s)
Chordata, Nonvertebrate/genetics , Estradiol/metabolism , Evolution, Molecular , Petromyzon/genetics , Receptors, Estrogen/genetics , Amino Acid Sequence , Animals , Benzhydryl Compounds , Cell Line , Chordata, Nonvertebrate/metabolism , Cloning, Molecular , Genes, Reporter , Humans , Molecular Sequence Data , Petromyzon/metabolism , Phenols/metabolism , Phylogeny , Response Elements , Sequence Alignment , Transcriptional Activation
15.
Mol Endocrinol ; 22(2): 304-16, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991765

ABSTRACT

The biological effects of dioxins are mediated by the aryl hydrocarbon receptor (AhR) and its dimerization partner, the AhR nuclear translocator (ARNT), and include interference with hormonal signaling pathways like the response to estrogens. The effects of estrogens are mediated by two estrogen receptor (ER) isoforms, ERalpha and ERbeta, which belong to the family of nuclear receptors. We have previously shown that ARNT can act as coactivator of the ERs. In this study, we show that recruitment of ARNT to AhR or hypoxia-inducible factor-1alpha signaling pathways as well as small interfering RNA-mediated down-regulation of ARNT levels lead to a reduction in ER transcriptional activity. Using chromatin immunoprecipitation assays, we demonstrate that this decrease coincides with reduced recruitment of ARNT to estradiol-regulated promoters. We show further that coactivation by ARNT as well as inhibition by dioxin acts stronger on ERbeta than on ERalpha activity. Additionally, we demonstrate that the effects of ARNT are dependent on the A/B domain of the ERs with the A/B domain of ERbeta being considerably stronger in mediating the coactivating effects of ARNT. Taken together, our studies show that recruitment of ARNT to the AhR after dioxin treatment can account for the antiestrogenic effect of dioxins. Moreover, we show for the first time that the inhibitory effects of dioxin are more pronounced on ERbeta than on ERalpha.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Dioxins/pharmacology , Estrogen Receptor beta/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cell Hypoxia , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Dimerization , Estradiol/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological , Mutation , Polychlorinated Dibenzodioxins/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
16.
Mol Pharmacol ; 73(2): 575-86, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18003862

ABSTRACT

The biological effects of 17beta-estradiol (E(2)) are mediated by the two estrogen receptor (ER) isoforms ERalpha and ERbeta. These receptors are ligand-inducible transcription factors that belong to the nuclear receptor superfamily. These receptors are also targets for a broad range of natural and synthetic compounds that induce ER activity, including dietary compounds, pharmaceuticals, and various types of environmental pollutants such as bisphenols and polychlorinated hydroxy-biphenyls. Here, we study the effect of the combustion byproduct 3-methylcholanthrene (3-MC) on ERalpha and ERbeta. 3-MC is a compound identified previously as an activator of the aryl hydrocarbon receptor (AhR). Activation of AhR is traditionally associated with an inhibition of the E(2) signaling network. In this study, we demonstrate that 3-MC is a cell-specific activator or inhibitor of E(2) signaling pathways. We show that 3-MC acts as a repressor in some cells, presumably via the AhR, whereas it is a potent activator of ER activity in other cells. It is interesting that we demonstrate that the estrogenic effects of 3-MC are dependent on the ability of cells to metabolize parental 3-MC to alternative compounds. In summary, our results suggest that exposure to AhR ligands like 3-MC can lead to either activation or repression of E(2) signaling, depending on the cellular context.


Subject(s)
Cell Membrane/metabolism , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Methylcholanthrene/pharmacology , Signal Transduction/physiology , Cell Line , Cell Membrane/drug effects , Cell Membrane/genetics , Estradiol/metabolism , Estradiol/physiology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Humans , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
17.
Mol Cell Biol ; 28(2): 784-93, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18039858

ABSTRACT

Circadian regulation of gene expression plays a major role in health and disease. The precise role of the circadian system remains to be clarified, but it is known that circadian proteins generate physiological rhythms in organisms by regulating clock-controlled target genes. The estrogen receptor beta (ERbeta) is, together with ERalpha, a member of the nuclear receptor superfamily and a key mediator of estrogen action. Interestingly, recent studies show that disturbed circadian rhythmicity in humans can increase the risk of reproductive malfunctions, suggesting a link between the circadian system and ER-mediated transcription pathways. Here, we identify a novel level of regulation of estrogen signaling where ERbeta, but not ERalpha, is controlled by circadian clock proteins. We show that ERbeta mRNA levels fluctuate in different peripheral tissues following a robust circadian pattern, with a peak at the light-dark transition, which is maintained under free-running conditions. Interestingly, this oscillation is abolished in clock-deficient BMAL1 knockout mice. Circadian control of ERbeta expression is exerted through a conserved E-box element in the ERbeta promoter region that recruits circadian regulatory factors. Furthermore, using small interfering RNA-mediated knockdown assays, we show that the expression levels of the circadian regulatory factors directly influence estrogen signaling by regulating the intracellular levels of endogenous ERbeta.


Subject(s)
Estrogen Receptor beta/metabolism , Gene Expression Regulation , Trans-Activators/metabolism , ARNTL Transcription Factors , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Circadian Rhythm , Estrogen Receptor beta/genetics , Humans , Lung/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Period Circadian Proteins , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Sequence Alignment , Sequence Homology , Signal Transduction , Trans-Activators/genetics , Transcription, Genetic/genetics
18.
Endocrinology ; 148(10): 4875-86, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17628008

ABSTRACT

Numerous dietary compounds can modify gene expression by binding to the members of the nuclear receptor superfamily of transcription factors. For example, dietary polyphenols, such as soy isoflavones genistein and daidzein, modulate the activity of the estrogen receptors (ERs)-alpha and ERbeta. An additional class of dietary polyphenols that modulate cellular signaling pathways are lignans, compounds that are common constituents of Western diets. In this study, we show that a metabolite of dietary lignans, enterolactone, at physiological concentrations, activates ER-mediated transcription in vitro with preference for ERalpha. The effects of enterolactone are mediated by the ER ligand binding domain and are susceptible to antiestrogen treatment. Furthermore, the affinity of enterolactone toward ERalpha, measured by a novel ligand binding assay, is augmented in cell culture conditions. Moreover, our results demonstrate for the first time that enterolactone has estrogenic activity in vivo. In transgenic estrogen-sensitive reporter mice, enterolactone induces tissue-specific estrogen-responsive reporter gene expression as well as promotes uterine stromal edema and expression of estrogen-responsive endogenous genes (CyclinD1 and Ki67). Taken together, our data show that enterolactone is a selective ER agonist inducing ER-mediated transcription both in vitro in different cell lines and in vivo in the mouse uterus.


Subject(s)
4-Butyrolactone/analogs & derivatives , Diet , Lignans/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , 4-Butyrolactone/biosynthesis , 4-Butyrolactone/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Cell Nucleus/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Gene Expression/drug effects , Genes, Reporter , Humans , Ligands , Lignans/biosynthesis , Lignans/pharmacology , Mice , Mice, Transgenic , Protein Structure, Tertiary/physiology , Receptors, Estrogen/agonists , Tissue Distribution , Transcriptional Activation/drug effects
19.
EMBO Rep ; 7(10): 1035-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16936638

ABSTRACT

Transcriptional control of hypothalamic thyrotropin-releasing hormone (TRH) integrates central regulation of the hypothalamo-hypophyseal-thyroid axis and hence thyroid hormone (triiodothyronine (T(3))) homeostasis. The two beta thyroid hormone receptors, TRbeta1 and TRbeta2, contribute to T(3) feedback on TRH, with TRbeta1 having a more important role in the activation of TRH transcription. How TRbeta1 fulfils its role in activating TRH gene transcription is unknown. By using a yeast two-hybrid screening of a mouse hypothalamic complementary DNA library, we identified a novel partner for TRbeta1, hepatitis virus B X-associated protein 2 (XAP2), a protein first identified as a co-chaperone protein. TR-XAP2 interactions were TR isoform specific, being observed only with TRbeta1, and were enhanced by T(3) both in yeast and mammalian cells. Furthermore, small inhibitory RNA-mediated knockdown of XAP2 in vitro affected the stability of TRbeta1. In vivo, siXAP2 abrogated specifically TRbeta1-mediated (but not TRbeta2) activation of hypothalamic TRH transcription. This study provides the first in vivo demonstration of a regulatory, physiological role for XAP2.


Subject(s)
Hypothalamus/metabolism , Proteins/metabolism , Proteins/physiology , Thyrotropin-Releasing Hormone/metabolism , Transcriptional Activation , Animals , Gene Expression Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Thyroid Hormone Receptors beta/metabolism , Tissue Distribution , Transfection
20.
Proc Natl Acad Sci U S A ; 100(11): 6517-22, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12754377

ABSTRACT

The biological effects of estrogens are mediated by the estrogen receptors ERalpha and ERbeta. These receptors regulate gene expression through binding to DNA enhancer elements and subsequently recruiting factors such as coactivators that modulate their transcriptional activity. Here we show that ARNT (aryl hydrocarbon receptor nuclear translocator), the obligatory heterodimerization partner for the aryl hydrocarbon receptor and hypoxia inducible factor 1alpha, functions as a potent coactivator of ERalpha- and ERbeta- dependent transcription. The coactivating effect of ARNT depends on physical interaction with the ERs and involves the C-terminal domain of ARNT and not the structurally conserved basic helix-loop-helix and PAS (Per-ARNT-Sim) motifs. Moreover, we show that ARNT/ER interaction requires the E2-activated ligand binding domain of ERalpha or ERbeta. These observations, together with the previous role of ARNT as an obligatory partner protein for conditionally regulated basic helix-loop-helix-PAS proteins like the aryl hydrocarbon receptor or hypoxia inducible factor 1alpha, expand the cellular functions of ARNT to include regulation of ERalpha and ERbeta transcriptional activity. ARNT was furthermore recruited to a natural ER target gene promoter in a estrogen-dependent manner, supporting a physiological role for ARNT as an ER coactivator.


Subject(s)
DNA-Binding Proteins , Helix-Loop-Helix Motifs , Receptors, Aryl Hydrocarbon , Receptors, Estrogen/physiology , Transcription Factors/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , COS Cells , Estrogen Receptor alpha , Estrogen Receptor beta , HeLa Cells , Humans , Precipitin Tests , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...