Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Endocrinology ; 161(1)2020 01 01.
Article in English | MEDLINE | ID: mdl-31754709

ABSTRACT

To limit excessive glucocorticoid secretion following hypothalamic-pituitary-adrenal (HPA) axis stimulation, circulating glucocorticoids inhibit corticotropin-releasing hormone (CRH) expression in paraventricular nucleus (PVN) neurons. As HPA function differs between sexes and depends on circulating estradiol (E2) levels in females, we investigated sex/estrous stage-dependent glucocorticoid regulation of PVN Crh. Using NanoString nCounter technology, we first demonstrated that adrenalectomized (ADX'd) diestrous female (low E2), but not male or proestrous female (high E2), mice exhibited a robust decrease in PVN CRH mRNA following 2-day treatment with the glucocorticoid receptor (GR) agonist RU28362. Immunohistochemical analysis of PVN CRH neurons in Crh-IRES-Cre;Ai14 mice, where TdTomato fluorescence permanently tags CRH-expressing neurons, showed similarly abundant co-expression of GR-immunoreactivity in males, diestrous females, and proestrous females. However, we identified sex/estrous stage-related glucocorticoid regulation or expression of GR transcriptional coregulators. Out of 17 coregulator genes examined using nCounter multiplex analysis, mRNAs that were decreased by RU28362 in ADX'd mice in a sex/estrous stage-dependent fashion included: GR (males = diestrous females > proestrous females), signal transducer and activator of transcription 3 (STAT3) (males < diestrous = proestrous), and HDAC1 (males < diestrous > proestrous). Steroid receptor coactivator 3 (SRC-3), nuclear corepressor 1 (NCoR1), heterogeneous nuclear ribonucleoprotein U (hnrnpu), CREB binding protein (CBP) and CREB-regulated transcription coactivator 2 (CRTC2) mRNAs were lower in ADX'd diestrous and proestrous females versus males. Additionally, most PVN CRH neurons co-expressed methylated CpG binding protein 2 (MeCP2)-immunoreactivity in diestrous female and male Crh-IRES-Cre;Ai14 mice. Our findings collectively suggest that GR's sex-dependent regulation of PVN Crh may depend upon differences in the GR transcriptional machinery and an underlying influence of E2 levels in females.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Estradiol/blood , Glucocorticoids/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Adrenalectomy , Androstanols/pharmacology , Animals , Corticotropin-Releasing Hormone/genetics , Estrous Cycle , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/agonists , Hypothalamus/cytology , Male , Mice , Mice, Inbred Strains , Neurons/drug effects , Neurons/physiology , RNA, Messenger , Sex Factors , Vagina/cytology
2.
J Steroid Biochem Mol Biol ; 174: 128-132, 2017 11.
Article in English | MEDLINE | ID: mdl-28847748

ABSTRACT

Although ERα activation properties have been intensively studied, this is not the case for their repressive properties. In this report, the ERα ligand binding domain (LBD) is shown to interact both with a deacetylase function and with HDAC1 and HDAC3. Ligands do not affect binding to the deacetylase activity or to HDAC1. In distinction, E2 reduced LBD binding to HDAC3, whereas Tmx had no effect. Knock-down of either HDAC1 or 3 led to increased transcriptional activity by both HDACs, presumably by decreased repression. In distinction, only HDAC3 knock-down led to increased activity in the presence of Tmx. In summary, ERα differentially interacts with HDACs 1 and 3 to regulate transcriptional activity.


Subject(s)
Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Tamoxifen/metabolism , Estrogen Receptor alpha/genetics , HeLa Cells , Humans , MCF-7 Cells
3.
Mol Cell Endocrinol ; 454: 158-164, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28655627

ABSTRACT

The hypothalamic-pituitary-adrenal (HPA) axis plays a critical role in mounting a stress response and maintaining homeostasis. A dysregulated HPA axis and elevated levels of CRH are associated with a number of disorders. Although extensive research has been devoted to understanding molecular events associated with stimulated CRH gene, less is known about the mechanisms that restrain CRH expression. Using a cell culture system, we report here two molecular aspects of CRH gene regulation that are required for maintenance of basal level of CRH gene expression. These are a specific CpG methylation at a single CpG, and adequate levels of the methyl CpG binding protein 2 (MeCP2). The single site methylation allows the recruitment of MeCP2 to the CRH gene promoter region, and MeCP2 knockdown leads to increased expression of CRH gene. Taken together, the results indicate that site-specific methylation and MeCP2 are required for maintenance of basal levels of CRH gene expression.


Subject(s)
Corticotropin-Releasing Hormone/genetics , CpG Islands/genetics , DNA Methylation/genetics , Methyl-CpG-Binding Protein 2/metabolism , Animals , Azacitidine/pharmacology , Base Sequence , Cell Line , Corticotropin-Releasing Hormone/metabolism , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Ligands , Promoter Regions, Genetic , Rats , Receptors, Glucocorticoid/metabolism
4.
PLoS One ; 11(9): e0161430, 2016.
Article in English | MEDLINE | ID: mdl-27588681

ABSTRACT

E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, cyclooxygenase- 2 (COX-2) expression. Although inflammatory processes in the brain are usually associated with microglia and astrocytes, we found that the COX-2 gene (cox-2) was expressed in a neuronal context, specifically in an amygdalar cell line (AR-5). Given that COX-2 has been reported to be in neurons in the brain, and that the amygdala is a site involved in neurodegenerative and neuropsychiatric processes, we investigated mechanisms by which E2 could down-regulate cox-2 expression in the AR-5 line. These cells express estrogen receptors alpha (ERα) and beta (ERß), and as shown here cox-2. At the level of RNA, E2 and the ERß selective ligand diarylpropionitrile (DPN) both attenuated gene expression, whereas the ERα selective ligand propyl pyrazole triol (PPT) had no effect. Neither ligand increased ERß at the cox-2 promoter. Rather, DPN decreased promoter occupancy of NF-κB p65 and histone 4 (H4) acetylation. Treatment with the non-specific HDAC inhibitor Trichostatin A (TSA) counteracted DPN's repressive effects on cox-2 expression. In keeping with the TSA effect, E2 and DPN increased histone deacetylase one (HDAC1) and switch-independent 3A (Sin3A) promoter occupancy. Lastly, even though E2 increased CpG methylation, DPN did not. Taken together, the pharmacological data indicate that ERß contributes to neuronal cox-2 expression, as measured by RNA levels. Furthermore, ER ligands lead to increased recruitment of HDAC1, Sin3A and a concomitant reduction of p65 occupancy and Ac-H4 levels. None of the events, however, are associated with a significant recruitment of ERß at the promoter. Thus, ERß directs recruitment to the cox-2 promoter, but does so in the absence of being recruited itself.


Subject(s)
Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Neurons/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Cell Line , Cyclooxygenase 2/genetics , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Gene Expression/drug effects , Neurons/metabolism , Nitriles/pharmacology , Phenols/pharmacology , Propionates/pharmacology , Pyrazoles/pharmacology , Rats
5.
BMC Genomics ; 14: 413, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23800343

ABSTRACT

BACKGROUND: Corticotropin-releasing hormone (CRH) plays an important role in regulating the mammalian stress response. Two of the most extensively studied neuronal populations that express CRH are in the hypothalamus and amygdala. Both regions are involved in the stress response, but the amygdala is also involved in mediating response to fear and anxiety. Given that both hypothalamus and amygdala have overlapping functions, but their CRH-expressing neurons may respond differently to a given perturbation, we sought to identify differentially expressed genes between two neuronal cell types, amygdalar AR-5 and hypothalamic IVB cells. Thus, we performed a microarray analysis. Our hypothesis was that we would identify differentially expressed transcription factors, coregulators and chromatin-modifying enzymes. RESULTS: A total of 31,042 genes were analyzed, 10,572 of which were consistently expressed in both cell lines at a 95% confidence level. Of the 10,572 genes, 2,320 genes in AR-5 were expressed at ≥ 2-fold relative to IVBs, 1,104 genes were expressed at ≥2-fold in IVB relative to AR-5 and 7,148 genes were expressed at similar levels between the two cell lines. The greatest difference was in six mitochondrial DNA-encoded genes, which were highly abundant in AR-5 relative to IVB cells. The relative abundance of these genes ranged from 413 to 885-fold according to the microarray results. Differential expression of these genes was verified by RTqPCR. The differentially expressed mitochondrial genes were cytochrome b (MT-CYB), cytochrome c oxidase subunit 1 and 2 (MT-CO1 and MT-CO2) and NADH-ubiquinone oxidoreductase chain 1, 2, and 3 (MT-ND1, MT-ND2, MT-ND3). CONCLUSION: As expected, the array revealed differential expression of transcription factors and coregulators; however the greatest difference between the two cell lines was in genes encoded by the mitochondrial genome. These genes were abundant in AR-5 relative to IVBs. At present, the reason for the marked difference is unclear. The cells may differ in mtDNA copy number, number of mitochondria, or regulation of the mitochondrial genome. The specific functions served by having such different levels of mitochondrial expression have not been determined. It is possible that the greater expression of the mitochondrial genes in the amygdalar cells reflects higher energy requirements than in the hypothalamic cell line.


Subject(s)
Amygdala/cytology , Cell Nucleus/genetics , Hypothalamus/cytology , Mitochondria/genetics , Transcriptome , Animals , Cell Line , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Corticotropin-Releasing Hormone/genetics , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Transcription Factors/metabolism
6.
Endocrinology ; 154(5): 1802-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23515287

ABSTRACT

Testosterone has been shown to suppress the acute stress-induced activation of the hypothalamic-pituitary-adrenal axis; however, the mechanisms underlying this response remain unclear. The hypothalamic-pituitary-adrenal axis is regulated by a neuroendocrine subpopulation of medial parvocellular neurons in the paraventricular nucleus of the hypothalamus (PVN). These neurons are devoid of androgen receptors (ARs). Therefore, a possibility is that the PVN target neurons respond to a metabolite in the testosterone catabolic pathway via an AR-independent mechanism. The dihydrotestosterone metabolite, 5α-androstane-3ß,17ß-diol (3ß-diol), binds and activates estrogen receptor-ß (ER-ß), the predominant ER in the PVN. In the PVN, ER-ß is coexpressed with oxytocin (OT). Therefore, we tested the hypothesis that 3ß-diol regulates OT expression through ER-ß activation. Treatment of ovariectomized rats with estradiol benzoate or 3ß-diol for 4 days increased OT mRNA selectively in the midcaudal, but not rostral PVN compared with vehicle-treated controls. 3ß-Diol treatment also increased OT mRNA in the hypothalamic N38 cell line in vitro. The functional interactions between 3ß-diol and ER-ß with the human OT promoter were examined using an OT promoter-luciferase reporter construct (OT-luc). In a dose-dependent manner, 3ß-diol treatment increased OT-luc activity when cells were cotransfected with ER-ß, but not ER-α. The 3ß-diol-induced OT-luc activity was reduced by deletion of the promoter region containing the composite hormone response element (cHRE). Point mutations of the cHRE also prevented OT-luc activation by 3ß-diol. These results indicate that 3ß-diol induces OT promoter activity via ER-ß-cHRE interactions.


Subject(s)
Androstane-3,17-diol/pharmacology , Estrogen Receptor beta/physiology , Oxytocin/genetics , Promoter Regions, Genetic/drug effects , Transcriptional Activation/drug effects , Androgens/metabolism , Androstane-3,17-diol/metabolism , Animals , Cells, Cultured , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Humans , Mice , Ovariectomy , Oxytocin/metabolism , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , Response Elements/drug effects , Response Elements/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/genetics
7.
Neuroendocrinology ; 96(2): 111-8, 2012.
Article in English | MEDLINE | ID: mdl-22538291

ABSTRACT

It is now well established that estrogens can influence a panoply of physiological and behavioral functions. In many instances, the effects of estrogens are mediated by the 'classical' actions of two different estrogen receptors (ERs), ERα or ERß. ERα and ERß appear to have opposing actions in the control of stress responses and modulate different neurotransmitter or neuropeptide systems. Studies elucidating the molecular mechanisms for such regulatory processes are currently in progress. Furthermore, the use of ERα and ERß knockout mouse lines has allowed the exploration of the importance of these receptors in behavioral responses such as anxiety-like and depressive-like behaviors. This review examines some of the recent advances in our knowledge of hormonal control of neuroendocrine and behavioral responses to stress and underscore the importance of these receptors as future therapeutic targets for control of stress-related signaling pathways.


Subject(s)
Estrogens/metabolism , Neurosecretory Systems/physiology , Receptors, Estrogen/metabolism , Stress, Psychological/pathology , Animals , Estrogens/pharmacology , Humans , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
8.
Endocrinology ; 153(5): 2353-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22434086

ABSTRACT

The endocrine component of the stress response is regulated by glucocorticoids and sex steroids. Testosterone down-regulates hypothalamic-pituitary-adrenal (HPA) axis activity; however, the mechanisms by which it does so are poorly understood. A candidate testosterone target is the oxytocin gene (Oxt), given that it too inhibits HPA activity. Within the paraventricular nucleus of the hypothalamus, oxytocinergic neurons involved in regulating the stress response do not express androgen receptors but do express estrogen receptor-ß (ERß), which binds the dihydrotestosterone metabolite 3ß,17ß-diol (3ß-diol). Testosterone regulation of the HPA axis thus appears to involve the conversion to the ERß-selective ligand 5α-androstane, 3ß-diol. To study mechanisms by which 3ß-diol could regulate Oxt expression, we used a hypothalamic neuronal cell line derived from embryonic mice that expresses Oxt constitutively and compared 3ß-diol with estradiol (E2) effects. E2 and 3ß-diol elicited a phasic response in Oxt mRNA levels. In the presence of either ligand, Oxt mRNA levels were increased for at least 60 min and returned to baseline by 2 h. ERß occupancy preceded an increase in Oxt mRNA levels in the presence of 3ß-diol but not E2. In tandem with ERß occupancy, 3ß-diol increased occupancy of the Oxt promoter by cAMP response element-binding protein and steroid receptor coactivator-1 at 30 min. At the same time, 3ß-diol led to the increased acetylation of histone H4 but not H3. Taken together, the data suggest that in the presence of 3ß-diol, ERß associates with cAMP response element-binding protein and steroid receptor coactivator-1 to form a functional complex that drives Oxt gene expression.


Subject(s)
Androstane-3,17-diol/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Oxytocin/genetics , Androstane-3,17-diol/metabolism , Animals , Cell Line , Cells, Cultured , Estradiol/metabolism , Estradiol/pharmacology , Gene Expression/drug effects , Hypothalamus/metabolism , Mice , Neurons/metabolism , Oxytocin/metabolism , Promoter Regions, Genetic/drug effects
9.
Physiol Behav ; 104(2): 312-20, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21463644

ABSTRACT

The paraventricular nucleus of the hypothalamus (PVH) plays a central role in regulating the hypothalamic-pituitary-adrenal (HPA) axis. Medial parvocellular neurons of the PVH (mpPVH) integrate sensory and humoral inputs to maintain homeostasis. Humoral inputs include glucocorticoids secreted by the adrenals, which down-regulate HPA activation. A primary glucocorticoid target is the population of mpPVH neurons that synthesize and secrete corticotropin-releasing factors, the most potent of which is corticotropin-releasing hormone (CRH). Although CRH gene (crh) expression is known to be down-regulated by glucocorticoids, the mechanisms by which this process occurs are still poorly understood. To begin this study we postulated that glucocorticoid repression of crh involves HDAC recruitment to the region of the crh proximal promoter. To evaluate this hypothesis, we treated hypothalamic cells that express CRH with the HDAC inhibitor trichostatin A (TSA). As predicted, treatment with TSA led to increased CRH mRNA levels and crh promoter activity. Although co-treatment with Dex (10(-7)M) reduced the TSA effect on mRNA levels, it failed to reduce promoter activity; however co-transfection of HDAC1 but not 3 restored Dex inhibition. A distinction between HDAC1 and 3 was also apparent with respect to crh promoter occupancy. Dex led to increased HDAC1 but not HDAC3 occupancy. In vivo studies revealed that CRH-immunoreactive (-ir) neurons contained HDAC1- and HDAC3-ir. Collectively, these data point to a role for HDAC1 in the physiologic regulation of crh.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Down-Regulation/physiology , Histone Deacetylase 1/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , RNA, Messenger/metabolism , Adrenalectomy , Analysis of Variance , Animals , Cell Line , Chromatin Immunoprecipitation , Corticotropin-Releasing Hormone/metabolism , Dexamethasone/pharmacology , Down-Regulation/drug effects , Drug Interactions , Glucocorticoids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Male , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley
10.
Endocrinology ; 149(1): 346-57, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17947358

ABSTRACT

In the central nervous system, CRH regulates several affective states. Dysregulation of neuronal crh expression in the paraventricular nucleus of the hypothalamus correlates with some forms of depression, and amygdalar crh expression may modulate levels of anxiety. Because estrogens modulate these states, we sought to determine 17beta-estradiol (E2) effects on crh expression. CRH mRNA levels were measured in the AR-5 amygdaloid cell line by RT-PCR analysis. They increased by 1 min of E2 treatment, suggesting that crh behaves as an immediate-early gene. After peaking at 3 min, CRH mRNA returned to basal levels and then increased by 60 min. To dissect some of the molecular mechanisms underlying these events, we measured occupancy of the crh promoter by estrogen receptors (ERs) and coactivators, using chromatin immunoprecipitation. Because this promoter does not contain palindromic estrogen response elements, we targeted the region of a cAMP regulatory element (CRE), implicated in crh regulation. The temporal pattern of the mRNA response was mimicked by recruitment of ERalpha and -beta, phospho-CRE-binding protein, coactivators steroid receptor coactivator-1 and CRE-binding protein-binding protein (CBP), and an increase in histone 3 and 4 acetylation. Lastly, ERalpha and -beta loading were temporally dissociated, peaking at 1 and 3 min, respectively. The ER peaks were associated with coactivators and acetylation patterns. ERalpha associated with phospho-CRE-binding protein, CBP, steroid receptor coactivator-1, and increased acetylated histone 3. ERbeta associated with CBP and increased acetylated histone 4. The tight temporal correlation between E2-induced CRH mRNA levels and promoter occupancy by ERs strongly suggest that E2 regulates crh expression through an ERalpha- and/or ERbeta-CRE alternate pathway.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Cyclic AMP/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation/drug effects , Response Elements , Acetylation , Amygdala/metabolism , Animals , CREB-Binding Protein/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Models, Biological , Nuclear Receptor Coactivator 1 , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Rats , Response Elements/drug effects , Time Factors , Transcription Factors/metabolism
11.
J Neurosci ; 24(47): 10628-35, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15564578

ABSTRACT

The hypothalamic-pituitary-adrenal axis regulates mammalian stress responses by secreting glucocorticoids. The magnitude of the response is in part determined by gender, for in response to a given stressor, circulating glucocorticoids reach higher levels in female rats than in males. This gender difference could result from estrogen regulation of the corticotropin-releasing hormone (CRH) promoter via either of its receptors: estrogen receptor (ER)alpha or ERbeta. Immunocytochemistry revealed that a subset (12%) of medial parvocellular CRH neurons in the rat hypothalamus contain ERbeta but not ERalpha. To determine whether ERs could regulate CRH promoter activity, we cotransfected cells with a CRH promoter construct and either ERalpha or individual ERbeta isoforms. ERalpha weakly stimulated CRH promoter transcriptional activity in a ligand-independent manner. Conversely, all ERbeta isoforms tested stimulated CRH promoter activity with different ligand profiles. ERbeta1 and ERbeta2delta3 displayed constitutive activity (ERbeta1 more than ERbeta2delta3). Ligand-dependent activity of beta isoforms 1 and 2 was altered by an Exon3 splice variant (delta3) or by the additional 18 amino acids in the ligand-binding domain of ERbeta2 isoforms. Lastly, we suggest that ER regulation of CRH takes place through an alternate pathway, one that requires protein-protein interactions with other transcription factors or their associated complexes. However, a pure ER-activator protein-1 alternate pathway does not appear to be involved.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Estrogen Receptor beta/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Animals , Estrogen Receptor alpha/physiology , Female , Gene Expression Regulation , HeLa Cells , Humans , Immunohistochemistry , Paraventricular Hypothalamic Nucleus/cytology , Promoter Regions, Genetic , Protein Isoforms/physiology , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/physiology , Transcription, Genetic
12.
Nucl Recept ; 2(1): 2, 2004 05 07.
Article in English | MEDLINE | ID: mdl-15132742

ABSTRACT

BACKGROUND: Estrogen receptors alpha and beta (ERalpha and ERbeta) differentially activate genes with AP-1 elements. ERalpha activates AP-1 targets via activation functions with estrogens (the AF-dependent pathway), whereas ERbeta, and a short version of ERalpha (ERalpha DBD-LBD) activate only with anti-estrogens (AF-independent pathway). The DNA binding domain (DBD) plays an important role in both pathways, even though neither pathway requires ERE recognition. RESULTS: Mutations of a highly conserved DBD lysine (ERalpha.K206A/G), lead to super-activation of AP-1 through activation function dependent pathways, up to 200 fold. This super-activity can be elicited either through ER AFs 1 or 2, or that of a heterologous activation function (VP16). The homologous substitution in ERbeta, K170A, or in ERalpha DBD-LBD leads to estrogen-dependent AP-1 activation and loss of the usually potent anti-estrogen effects. Each of numerous K206 substitutions in ERalpha, except K206R, eliminates anti-estrogen activation and this loss correlates perfectly with a loss of ability to titrate a repressive function from the RU486 bound progesterone receptor. CONCLUSION: We conclude that ER DBDs contain a complex regulatory function that influences ligand activation profiles at AP-1. This function, which requires the integrity of the conserved lysine, both allows for activation at AP-1 with anti-estrogens (with ERbeta and ERalpha DBD-LBD), and prevents ERalpha from becoming superactive at AP-1 with estrogens. We discuss the possibility that a repressor interaction with the DBD both mediates the AF-independent pathway and dampens the AF dependent pathway. Mutations in the conserved lysine might, by this model, disrupt the binding or function of the repressor.

13.
Gene ; 330: 133-42, 2004 Apr 14.
Article in English | MEDLINE | ID: mdl-15087132

ABSTRACT

Human Kruppel-like factor 5 (hKLF5) is a transcription factor with a potential tumor suppressor function in prostate and breast cancers. In the majority of cancer samples examined, a significant loss of expression for KLF5 has been detected. Whereas hemizygous deletion appears to be responsible for KLF5's reduced expression in about half of the cases, the mechanism for reduction is unknown in the remaining half; gene promoter methylation does not appear to be involved. In this report, we studied the regulation of KLF5 and cloned and functionally characterized a 1944-bp fragment of the 5'-flanking region of the hKLF5 gene. Several mitogens as well as global demethylation induced the expression of KLF5, implicating multiple factors in the regulation of KLF5. KLF5's promoter lacks a TATA box and has a GC-rich region. Deletion mapping in combination with promoter activity assay showed that multiple cis-elements are involved in the transcriptional regulation of KLF5, some of which may play a repressor role whereas some others play an enhancer role. The Sp1 site between position -239 and -219 is essential for a basal promoter activity. Deletion or mutations of this Sp1 site significantly reduced promoter activity in several epithelial cell lines. Electrophoretic mobility shift assays (EMSAs) revealed that the Sp1 site binds Sp1 protein in nucleic extracts of different cell lines. In addition, overexpression of Sp1 protein transactivates KLF5 promoter activity. These findings suggest that Sp1 is a key transcription factor in KLF5's dynamic transcriptional regulation.


Subject(s)
Sp1 Transcription Factor/metabolism , Trans-Activators/genetics , Angiotensin II/pharmacology , Animals , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Cloning, Molecular , Conserved Sequence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Electrophoretic Mobility Shift Assay , Epidermal Growth Factor/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblast Growth Factor 2/pharmacology , Gene Expression/drug effects , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Kruppel-Like Transcription Factors , Luciferases/genetics , Luciferases/metabolism , Male , Metribolone/pharmacology , Mice , Molecular Sequence Data , Oligonucleotides/metabolism , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rodentia , Sequence Deletion , Sequence Homology, Nucleic Acid , Sp1 Transcription Factor/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tretinoin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
14.
J Biol Chem ; 277(27): 24353-60, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-11986316

ABSTRACT

Induction of cyclin D1 gene transcription by estrogen receptor alpha (ERalpha) plays an important role in estrogen-mediated proliferation. There is no classical estrogen response element in the cyclin D1 promoter, and induction by ERalpha has been mapped to an alternative response element, a cyclic AMP-response element at -57, with possible participation of an activating protein-1 site at -954. The action of ERbeta at the cyclin D1 promoter is unknown, although evidence suggests that ERbeta may inhibit the proliferative action of ERalpha. We examined the response of cyclin D1 promoter constructs by luciferase assay and the response of the endogenous protein by Western blot in HeLa cells transiently expressing ERalpha, ERalphaK206A (a derivative that is superactive at alternative response elements), or ERbeta. In each case, ER activation at the cyclin D1 promoter is mediated by both the cyclic AMP-response element and the activating protein-1 site, which play partly redundant roles. The activation by ERbeta occurs only with antiestrogens. Estrogens, which activate cyclin D1 gene expression with ERalpha, inhibit expression with ERbeta. Strikingly, the presence of ERbeta completely inhibits cyclin D1 gene activation by estrogen and ERalpha or even by estrogen and the superactive ERalphaK206A. The observation of the opposing action and dominance of ERbeta over ERalpha in activation of cyclin D1 gene expression has implications for the postulated role of ERbeta as a modulator of the proliferative effects of estrogen.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation/physiology , Receptors, Estrogen/physiology , Amino Acid Substitution , Cyclic AMP Response Element-Binding Protein/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Genes, Reporter , Genetic Vectors , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mutagenesis, Site-Directed , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...