Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Oncogene ; 28(4): 575-86, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18997820

ABSTRACT

The breast cancer susceptibility gene 1 (BRCA1) is mutated in approximately 50% of hereditary breast cancers, and its expression is decreased in 30-40% of sporadic breast cancers, suggesting a general role in breast cancer development. BRCA1 physically and functionally interacts with estrogen receptor-alpha (ERalpha) and several transcriptional regulators. We investigated the relationship between cellular BRCA1 levels and tamoxifen sensitivity. Decreasing BRCA1 expression in breast cancer cells by small interfering RNA alleviated tamoxifen-mediated growth inhibition and abolished tamoxifen suppression of several endogenous ER-targeted genes. ER-stimulated transcription and cytoplasmic signaling was increased without detectable changes in ER or ER coregulator expression. Co-immunoprecipitation studies showed that with BRCA1 knockdown, tamoxifen-bound ERalpha was inappropriately associated with coactivators, and not effectively with corepressors. Chromatin immunoprecipitation studies demonstrated that with tamoxifen, BRCA1 knockdown did not change ERalpha promoter occupancy, but resulted in increased coactivator and decreased corepressor recruitment onto the endogenous cyclin D1 promoter. Our results suggest that decreased BRCA1 levels modify ERalpha-mediated transcription and regulation of cell proliferation in part by altering ERalpha-coregulator association. In the presence of tamoxifen, decreased BRCA1 expression results in increased coactivator and decreased corepressor recruitment on ER-regulated gene promoters.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mutation , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tamoxifen/therapeutic use , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
2.
Endocrinology ; 149(1): 139-45, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932215

ABSTRACT

We investigated whether Jun N-terminal kinase (JNK) and p38 mediate gonadotropin subunit transcriptional responses to pulsatile GnRH in normal rat pituitaries. A single pulse of GnRH or vehicle was given to female rats in vivo, pituitaries collected, and phosphorylated JNK and p38 measured. GnRH stimulated an increase in JNK phosphorylation within 5 min, which peaked 15 min after GnRH (3-fold). GnRH also increased p38 phosphorylation 2.3-fold 15 min after stimulus. Rat pituitary cells were given 60-min pulses of GnRH or media plus the JNK inhibitor SP600125 (SP, 20 microM), p38 inhibitor SB203580 (20 microM), or vehicle. In vehicle-treated groups, GnRH pulses increased LHbeta and FSHbeta primary transcript (PT) levels 3-fold. SP suppressed both basal and GnRH-induced increases in FSHbeta PT by half, but the magnitude of responses to GnRH was unchanged. In contrast, SP had no effect on basal LHbeta PT but suppressed the stimulatory response to GnRH. SB203580 had no effect on the actions of GnRH on either LH or FSHbeta PTs. Lbeta-T2 cells were transfected with dominant/negative expression vectors for MAPK kinase (MKK)-4 and/or MKK-7 plus a rat LHbeta promoter-luciferase construct. GnRH stimulated a 50-fold increase in LHbeta promoter activity, and the combination of MKK-4 and -7 dominant/negatives suppressed the response by 80%. Thus, JNK (but not p38) regulates both LHbeta and FSHbeta transcription in a differential manner. For LHbeta, JNK is essential in mediating responses to pulsatile GnRH. JNK also regulates FSHbeta transcription (i.e. maintaining basal expression) but does not play a role in responses to GnRH.


Subject(s)
Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropins/genetics , JNK Mitogen-Activated Protein Kinases/physiology , Pituitary Gland/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured , Male , Periodicity , Phosphorylation/drug effects , Pituitary Gland/metabolism , Promoter Regions, Genetic/drug effects , Protein Subunits/genetics , Rats , Time Factors , Transcription, Genetic/drug effects
3.
Biol Reprod ; 72(3): 523-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15509729

ABSTRACT

This study investigated whether estradiol (E2) or testosterone (T) activate extracellular signal-regulated kinase (ERK) and calcium/calmodulin-dependent kinase II (Ca/CaMK II), as indicated by enzyme phosphorylation in rat pituitaries. In vivo studies used adult female rats given E2, T, or empty silastic capsules (vehicle controls). Twenty-four hours later, the rats were given a single pulse of GnRH (300 ng) or BSA-saline (to controls) and killed 5 min later. GnRH stimulated a two- to three-fold rise in activated Ca/CaMK II, and E2 and T had no effect on Ca/CaMK II activation. In contrast, both GnRH and T stimulated threefold increases in ERK activity, with additive effects seen following the combination of GnRH+T. E2 had no effect on ERK activity. In alpha T3 clonal gonadotrope cells, dihydrotestosterone did not activate ERK alone but enhanced and prolonged the ERK responses to GnRH, demonstrating direct effects on the gonadotrope. Thus, the ERK response to GnRH plus androgen was enhanced in both rat pituitary and alpha T3 cells. In vitro studies with cultured rat pituitary cells examined the effect of GnRH+/-T in the presence of the mitogen-activated protein (MAP) kinase kinase inhibitor, PD-098059 (PD). Results showed that PD suppressed ERK activational and FSH beta transcriptional responses to T. These findings suggest that one site of T regulation of FSH beta transcription is through the selective stimulation of the ERK pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Follicle Stimulating Hormone, beta Subunit/metabolism , Pituitary Gland/enzymology , Testosterone/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cells, Cultured , Enzyme Activation , Estradiol/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Ovariectomy , Phosphorylation , Pituitary Gland/cytology , Rats , Transcriptional Activation
4.
J Neuroendocrinol ; 14(2): 85-94, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11849367

ABSTRACT

Information on oestrogen action has grown exponentially in the past decade, and recent studies have begun to define the mechanism of ligand-dependent activation and cell-specific effects. Oestrogen-mediated gene transcription in a specific tissue depends on several factors, the most important of which is the presence of at least one of the two nuclear oestrogen receptor (ER) isoforms, ER(alpha) and ERbeta. The presence and levels of specific ER isoform variants, along with receptor coactivator, corepressor and integrator proteins, directly modulate overall nuclear ER activity. The structure of the ligand, including both physiological oestrogens and synthetic oestrogen receptor modulators, influences ER interactions with these other proteins and thus determines the biological response. Furthermore, peptide and neurotransmitter-stimulated intracellular signalling pathways activate specific enzyme cascades and may modify the receptors and their interacting proteins, resulting in either independent or ligand-enhanced ER-mediated responses. Finally, several rapid effects of oestrogen probably occur at the membrane through nongenomic pathways that may or may not require the same ER proteins that are found in the nucleus. This review concentrates on the pituitary-hypothalamic axis and the genomic effects of oestrogen, and discusses the current knowledge of each of these factors in determining oestrogen actions in the neuroendocrine system.


Subject(s)
Estrogens/physiology , Hypothalamo-Hypophyseal System/physiology , Receptors, Estrogen/physiology , Animals , Humans
5.
Oncogene ; 20(47): 6965-9, 2001 Oct 18.
Article in English | MEDLINE | ID: mdl-11687977

ABSTRACT

Both the estrogen receptor (ER) and the progesterone receptor (PR) have two subtypes: ER-alpha and beta, and PR-A and -B, respectively. These subtypes differ in function and expression, and recent reports have correlated changes in the normal proportions of these isoforms with neoplastic states. We investigated ER and PR isoform expression in normal pre- and post-menopausal endometrium, well-differentiated endometrial adenocarcinoma, and poorly differentiated malignant mixed mullerian tumors (MMMTs). Semi-quantitative RT-PCR and immunoblotting were used to measure receptor mRNA and protein expression. Estrogen receptor-alpha/beta mRNA ratios were significantly higher in postmenopausal (27.3) compared to premenopausal endometrium (4.9) mainly as a result of lower ER-beta expression in the former. Compared to age-matched postmenopausal controls, the ER-alpha/beta ratio was reduced in both grade I adenocarcinoma and MMMT specimens (3.3 and 6.8, respectively), due to a selective loss of ER-alpha. The relative abundance of PR-A and PR-B mRNA remained unchanged between all tissue subtypes. Total PR protein, however, was significantly reduced in MMMTs compared to all other groups. Thus, sex steroid receptor expression is significantly and differentially altered in well-differentiated and poorly-differentiated endometrial cancers. Both cancers exhibit decreased ER-alpha expression and the MMMTs also demonstrate a significant loss of PR protein.


Subject(s)
Adenocarcinoma/metabolism , Endometrial Neoplasms/metabolism , Mixed Tumor, Mullerian/metabolism , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Cell Differentiation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Humans , Menopause/metabolism , Middle Aged , Mixed Tumor, Mullerian/genetics , Mixed Tumor, Mullerian/pathology , RNA, Neoplasm/biosynthesis , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics
6.
Mol Endocrinol ; 15(11): 1906-17, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11682622

ABSTRACT

Steroids may regulate LH subunit gene transcription by modulating hypothalamic GnRH pulse patterns or by acting at the pituitary gonadotrope to alter promoter activity. We tested direct pituitary effects of the androgen dihydrotestosterone (DHT) to modulate the rat LHbeta promoter in transfected LbetaT2 clonal gonadotrope cells and in pituitaries of transgenic mice expressing LHbeta-luciferase. The LHbeta promoter (-617 to +44 bp)-luciferase construct was stimulated in LbetaT2 cells 7- to 10-fold by GnRH. Androgen treatment had little effect on basal promoter activity but suppressed GnRH stimulation by approximately 75%. GnRH stimulation of LHbeta was also suppressed by DHT in isolated pituitary cells from male or female mice with functional nuclear ARs, but not in male littermates with mutant AR. GnRH stimulation of the LHbeta promoter requires interactions between a complex distal response element containing two specificity protein-1 (Sp1) binding sites and a CArG box, and a proximal element with two bipartite binding sites for steroidogenic factor-1 and early growth response protein-1 (Egr-1). DHT effectively suppressed promoter constructs with an intact distal response element. The distal response element does not bind AR, but AR reduces Sp1 binding to this region. Glutathione-S-transferase pull-down studies demonstrated direct interactions of AR with Sp1, which requires the DNA-binding domain of AR, and weaker interactions with Egr-1. We conclude that androgen suppression of the rat LHbeta promoter occurs primarily through direct interaction of AR with Sp1, with some possible role through binding to Egr-1. These interactions result in interference with GnRH-stimulated gene transcription by reducing cooperation between the distal and proximal GnRH response elements.


Subject(s)
Dihydrotestosterone/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Animals , Binding Sites , Cells, Cultured , Female , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Pituitary Gland/cytology , Pituitary Gland/physiology , Promoter Regions, Genetic , Rats , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, LHRH/drug effects , Receptors, LHRH/genetics , Response Elements/drug effects , Response Elements/genetics , Steroidogenic Factor 1 , Suppression, Genetic , Transfection
7.
Endocrinology ; 142(8): 3361-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459779

ABSTRACT

In pituitary and other target tissues, estrogen acts through ERs, which are ligand-activated nuclear transcription factors. ERs can also be activated by intracellular signaling pathways in a ligand-independent manner in some cells. Because the pituitary is the target of several cAMP-activating factors, we examined the ability of cAMP to activate ERs in the alphaT3 gonadotrope cell line. Forskolin, 8-bromo-cAMP, and pituitary adenylate cyclase-activating polypeptide all enhanced ER-dependent promoter activity, which was inhibited by antiestrogen or a pituitary-specific inhibitory ER variant. Activation was PKA dependent and was blocked by the PKA inhibitor H89 or cotransfection of the inhibitor PKI. Although cAMP activated MAPK in alphaT3 cells, inhibition of MAPK with the MEK inhibitor PD98059 did not prevent forskolin-induced ER activation. Similarly, epidermal growth factor did not stimulate ER activity, although it increased MAPK activation. Forskolin-induced activation of ER was enhanced by cotransfection of steroid receptor coactivator-1 and was inhibited by the repressor of ER action, suggesting that cAMP does not alter the normal interactions between ER and cofactors. In contrast to results with estrogen, cAMP treatment did not decrease ER protein levels. These results demonstrate that in the pituitary, cAMP activates ER in a ligand-independent manner exclusively through PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Pituitary Gland/metabolism , Receptors, Estrogen/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP/physiology , Estrogen Receptor alpha , Estrogens/pharmacology , Histone Acetyltransferases , Ligands , Mice , Mitogen-Activated Protein Kinases/physiology , Nuclear Receptor Coactivator 1 , Prohibitins , Receptors, Estrogen/drug effects , Repressor Proteins/pharmacology , Transcription Factors/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
8.
Endocrinology ; 141(6): 2174-84, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10830306

ABSTRACT

Estrogen (E) regulates the synthesis and secretion of several pituitary hormones during the reproductive cycle in a cell- and promoter-specific manner. One mechanism underlying cell specificity is the differential expression of estrogen receptor (ER) isoforms. We used in vivo and in vitro rodent pituitary cell models to examine the expression and regulation of ERalpha, ERbeta, and the pituitary-specific ERalpha isoform, truncated estrogen receptor product-1 (TERP-1). In cycling female rat pituitaries, ERbeta messenger RNA (mRNA) levels fell 40% on the morning of proestrus and were suppressed by E or dihydrotestosterone in ovariectomized females. In lactotrope and gonadotrope cell lines (GH3, RC4B, LbetaT2), progesterone (P) or P plus E also suppressed ERbeta. TERP-1 mRNA increased 3-fold at proestrus and in response to E treatment in vivo and in cell lines. ERalpha mRNA levels were not regulated significantly by any treatment in vivo or in cell lines. However, E suppressed ERalpha protein levels in vivo and in cell lines, and reduction of ERalpha protein levels by E or the antiestrogen ICI182,780 reduced E-stimulated transcriptional activation of the PRL promoter in GH3 cells. TERP-1 and ERbeta protein levels were low to undetectable in cell lines, but E stimulated TERP-1. Because E treatment decreases ERbeta mRNA and ERalpha protein and increases levels of TERP-1 (which can suppress ERalpha/beta activity), the dynamic steroid-induced changes in ER expression in the rat pituitary during the midcycle gondaotropin/PRL surge may provide a means for ovarian steroids to limit positive feedback.


Subject(s)
Estradiol/pharmacology , Gene Expression Regulation , Pituitary Gland/metabolism , Receptors, Estrogen/genetics , Animals , Cell Line , Dihydrotestosterone/pharmacology , Estrogen Receptor alpha , Female , Gene Expression Regulation/drug effects , In Situ Hybridization , Male , Mice , Orchiectomy , Ovariectomy , Pituitary Gland/chemistry , Progesterone/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Endocrinol ; 14(4): 472-85, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10770486

ABSTRACT

GnRH pulses regulate gonadotropin subunit gene transcription in a frequency-dependent, subunit-specific manner. The alpha-subunit gene is stimulated by constant GnRH and by rapid to intermediate pulse frequencies, while stimulation of LHbeta subunit gene transcription requires intermediate frequency pulses. We have defined the GnRH-responsive elements of the rat LH subunit gene promoters by deletion/mutation analysis and transfection studies in rat pituitary cells and two clonal gonadotrope cell lines. The alpha-subunit gene GnRH-responsive region lies between -411 and -375 bp. The region contains two Ets-domain protein binding sites, and mutating either site obliterates the response. DNA protein binding studies demonstrate the two sites are not equivalent, and that Ets-1 does not mediate this response. Studies of the LHbeta promoter reveal a major GnRH-responsive region between -456 and -342 bp. Within this region, two Sp1 binding sites contribute to the GnRH response, and the 3'Sp1 site is also critical for basal expression. The 5'Sp1 site partially overlaps a CArG box, and mutating the CArG element specifically eliminates the response to pulsatile GnRH. DNA containing this mutation cannot form intermediate mobility complexes with nuclear proteins, but retains Sp1 binding. Mutation of the 3'Sp1 site and either the 5'Sp1 or CArG element partially restores GnRH stimulation, suggesting a downstream element contributes to the full GnRH response. These studies demonstrate that unique composite elements and transcription factors are responsible for GnRH stimulation of the LH subunit genes and may contribute to their differential responses to GnRH pulses.


Subject(s)
Glycoprotein Hormones, alpha Subunit/genetics , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/genetics , Promoter Regions, Genetic , Response Elements , Animals , Base Sequence , Binding Sites , Consensus Sequence , DNA/chemistry , DNA/metabolism , DNA Mutational Analysis , Gene Deletion , Humans , Mice , Mutagenesis , Nuclear Proteins/metabolism , Rats , Transfection
10.
J Biol Chem ; 275(10): 7158-66, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10702284

ABSTRACT

The estrogen receptor (ER) is a ligand-activated transcription factor that acts as a homodimer. Truncated estrogen receptor product-1 (TERP-1) is a pituitary-specific, estrogen-induced, isoform of rat ERalpha that is transcribed from a unique start site and contains only the C-terminal region of the full-length receptor. TERP-1 does not affect transcription directly but suppresses ligand-activated ERalpha and ERbeta activity. Because TERP-1 contains a dimerization domain and part of the coactivator binding pocket, we hypothesized that it modulates ER function by direct interactions with full-length ER or the steroid receptor coactivator, SRC-1. Localization studies demonstrate that TERP-1 is present in the cytoplasm and nucleus of transfected cells and colocalizes with nuclear ER. Protein binding studies show that TERP-1 forms heterodimers with both ERalpha and ERbeta and inhibits ERalpha binding to its cognate DNA response element. TERP-1 also binds SRC-1, and increasing levels of SRC-1 decrease the TERP-1-ERalpha interactions, in agreement with the rescue of TERP-1-suppressed ERalpha transcriptional activity by SRC-1. Mutational analysis of TERP-1 and ERalpha in the activation helix and the AF-2 dimerization helix indicates that TERP-1 acts predominantly through dimerization with ERalpha. Therefore, TERP-1 suppression of ER transcription occurs primarily by formation of inactive heterodimers and secondarily by competition for coactivators.


Subject(s)
Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/physiology , Repressor Proteins , Transcriptional Activation , Animals , Binding, Competitive , COS Cells , Cell Nucleus/chemistry , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/physiology , Dimerization , Estrogen Receptor alpha , Estrogen Receptor beta , Rats , Receptors, Estrogen/chemistry , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/physiology , Response Elements , Transcription Factors/physiology
11.
Cancer Res ; 60(2): 245-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667568

ABSTRACT

Epidemiological data suggest a protective effect for estrogen replacement therapy on colon cancer. The estrogen receptor (ER) is required for the action of estrogen. The ER-beta isoform is functionally similar to ER-alpha but has a distinct pattern of expression and transcriptional response to selective estrogen response modulators. Our goal was to investigate the presence of ER-alpha and ER-beta in normal and malignant colon tissue. Human colon cancer tissue and adjacent normal colon tissue were harvested from five male and six female patients undergoing segmental colon resection for colon cancer. Western blot analysis revealed very low levels of ER-alpha protein in tumor and normal colon tissue. In both male and female patients, malignant colon tissue showed a selective loss of ER-beta protein expression when compared to normal colon tissue in the same patient. Semiquantitative reverse transcription-PCR revealed no difference in ER-beta mRNA levels between normal and malignant colon tissue. Malignant transformation of the colon is associated with a marked diminution of ER-beta protein expression, possibly through a posttranscriptional mechanism.


Subject(s)
Colonic Neoplasms/pathology , Receptors, Estrogen/analysis , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/surgery , Endometrium/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Humans , Male , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , beta 2-Microglobulin/analysis
12.
Rev Endocr Metab Disord ; 1(1-2): 35-42, 2000 Jan.
Article in English | MEDLINE | ID: mdl-11704990

ABSTRACT

T3 suppression of TSH subunit gene transcription is an important step in maintaining thyroid hormone homeostasis, and recent investigations have increased our understanding of this process. Thyrotrope-specific proteins play a critical role in TSH subunit gene expression, and influence T3-mediated regulatory mechanisms. The structure and placement of the TSH gene TREs define suppressive regulation by T3, and this process is favored by the TR isoforms expressed in the pituitary. Elimination of TR beta function compromises the pituitary response to T3. TR beta 2, the isoform specifically expressed in pituitary and neural tissue, contains a transferable domain that both increases T3-independent gene transcription and enhances T3-suppressed transcription. The functional interaction of TR beta 2 with other regulatory proteins is distinct from that of other TR isoforms, and likely plays a critical role in pituitary physiology and in pituitary resistance to thyroid hormone. The development of novel thyrotrope cell lines will allow investigators to define new proteins and molecular mechanisms that distinguish negative from positive T3 transcriptional regulation.


Subject(s)
Gene Expression/drug effects , Pituitary Hormones/genetics , Triiodothyronine/pharmacology , Animals , Humans , Pituitary Gland/physiology , Protein Isoforms/physiology , Receptors, Thyroid Hormone/physiology , Response Elements , Thyrotropin/genetics
13.
Horm Behav ; 36(2): 98-108, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506534

ABSTRACT

The midbrain periaqueductal gray (PAG) has been strongly implicated in numerous behaviors heavily influenced by the gonadal steroids estrogen and testosterone, including reproductive behavior, autonomic regulation, and antinociception. However, the location of receptors for these steroids within the PAG has not been carefully characterized. Immunocytochemical techniques were used to map the distribution of neurons immunoreactive for the androgen (AR) and estrogen receptor (alpha subtype; ERalpha) along the rostrocaudal axis of the PAG in the male rat. The results show that the PAG contains a large population of both androgen and estrogen receptor containing neurons. Neurons immunoreactive for either receptor were concentrated within the caudal two-thirds of the PAG. At midlevels of the PAG, ERalpha and AR immunoreactive neurons were located primarily within the dorsomedial and lateral PAG. In the caudal third of the PAG, immunoreactive cells were distributed primarily within the dorsal half. The distributions of ERalpha and AR were remarkably similar, and it is likely that some PAG neurons contain receptors for both gonadal steroids, similar to what has been previously reported for the male rat hypothalamus. The results of this study suggest that the PAG may provide the anatomical substrate for steroid mediated changes in nociceptive thresholds and reproductive behavior.


Subject(s)
Periaqueductal Gray/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Animals , Brain Mapping , Immunohistochemistry , Male , Periaqueductal Gray/cytology , Photomicrography , Rats , Rats, Sprague-Dawley , Sexual Behavior, Animal/physiology
14.
Gynecol Oncol ; 74(1): 38-47, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385549

ABSTRACT

Breast cancer tissue has been shown to contain alternatively spliced estrogen receptor alpha (ER-alpha) mRNA variants, which have altered biological activities compared to the full-length ER-alpha. The development of endometrial cancer, as well as drug resistance in breast cancer patients undergoing tamoxifen therapy, may represent altered ER-alpha function secondary to specific exon deletions. While the literature is replete with ER mRNA variant data, little information is available regarding the presence and function of endometrial ER variant proteins. We evaluated the presence of human ER-alpha mRNA and protein variants in six premenopausal, six postmenopausal, and six endometrial carcinoma samples. Reverse transcription-polymerase chain reaction, DNA hybridization, and sequencing techniques identified exon 4, exon 5, and exon 7 mRNA splice variants in all patients as well as MCF-7 and Ishikawa cell lines. Presence of translated proteins for full-length ER-alpha, as well as splice variants, was investigated by Western blot analysis using antibodies directed against the N-terminus, hinge region, and C-terminus portions of the ER. These experiments confirmed the presence of immunopositive protein bands of approximately 64-66 kDa in all patients corresponding to wild-type ER-alpha. A protein band migrating at 41 kDa, consistent with an exon 5 splice variant, was only seen in endometrial adenocarcinoma samples. Premenopausal and postmenopausal endometrial samples did not contain detectable amounts of ER splice variant protein. Human ER-alpha mRNA variants are present in all human endometrial samples, but detectable levels of variant proteins are only observed in patients with endometrial adenocarcinoma.


Subject(s)
Endometrial Neoplasms/genetics , RNA, Messenger/biosynthesis , Receptors, Estrogen/biosynthesis , DNA, Neoplasm/analysis , Exons , Female , Humans , Receptors, Estrogen/genetics
15.
Mol Endocrinol ; 13(2): 320-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973261

ABSTRACT

Truncated estrogen receptor product-1 (TERP-1) is a naturally occurring rat estrogen receptor (ER) variant transcribed from a unique start site and containing a unique 5'-untranslated region fused to exons 5-8 of ERalpha. TERP-1 is detected only in the pituitary, and TERP-1 mRNA levels are highly regulated during the estrous cycle, exceeding those of the full-length ERalpha on proestrus. These data suggest that TERP-1 may play a role in estrogen- regulated feedback in the pituitary. We examined the ability of TERP-1 to modulate gene transcription in transiently transfected ER-negative (Cos-1) and ER-positive pituitary (alphaT3 and GH3) cell lines. In Cos-1 cells transiently cotransfected with TERP-1 and either ERalpha or ERbeta, low levels of TERP-1 (ratios of < 1:1 with ER) enhanced transcription of model promoters containing estrogen response elements by an average of 3- to 4-fold above that seen with ER alone. At higher concentrations of TERP-1 (> 1:1 with ER) transcription was inhibited. TERP-1 also had a biphasic action on transcription in the alphaT3 and GH3 pituitary cell lines, although the stimulatory action was less pronounced. TERP-1 actions were dependent on ligand-activated ER as TERP-1 did not bind estradiol in transfected Cos-1 cells or in vitro, and estrogen antagonists prevented the stimulatory effects of TERP-1. Coimmunoprecipitation studies suggest that TERP-1 does not bind with high affinity to the full-length ERalpha. However, TERP-1 may compete with ER for binding sites of receptor cofactors because steroid receptor coactivator-1 (SRC-1) rescued the inhibitory actions of TERP-1. The ability of TERP-1 to both enhance and inhibit ER-dependent promoter activity suggests that TERP-1 may play a physiological role in estrogen feedback in the rat pituitary.


Subject(s)
Gene Expression Regulation , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Animals , Binding, Competitive , Blotting, Western , COS Cells , Chlorocebus aethiops , Diethylstilbestrol/metabolism , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogen Antagonists/metabolism , Female , Fulvestrant , Pituitary Gland/physiology , Plasmids/chemistry , Precipitin Tests , Rats , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism
16.
J Neurosci ; 18(22): 9556-63, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9801392

ABSTRACT

Mice, rats, and humans have two types of estrogen receptors, estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta). Estrogen receptor-alpha gene-disrupted (ERalpha-disrupted) mice bear two nonfunctional copies of the ERalpha gene. This mutation blocks the synthesis of full-length ERalpha, renders the animals infertile, and inhibits the induction of female sexual behaviors by estradiol and progesterone. It is likely that many of the processes contributing to the regulation of sexual receptivity by estradiol and progesterone are compromised in ERalpha-disrupted mice. However, given the importance of progesterone in the regulation of sexual receptivity and given the importance of progestin receptors (PRs) in mediating the responses of females to progesterone, we investigated the effects of ERalpha disruption on the induction of PRs by estradiol in the forebrain. We hypothesized that estradiol would induce PRs in wild-type mice but not in ERalpha-disrupted mice. Ovariectomized wild-type and ERalpha-disrupted mice were implanted with either estradiol-filled capsules or empty capsules for 5 d, after which their brains were processed for the immunocytochemical detection of PR. Estradiol increased the number of PR-immunoreactive cells in both wild-type and ERalpha-disrupted mice. The residual responsiveness of ERalpha-disrupted mice to estradiol could be accounted for by an ERbeta-dependent mechanism or another as yet unidentified estrogen receptor; however, because ERalpha-immunoreactivity and PCR product representing the 3' end of ERalpha mRNA were found in at least one PR-containing region of the ERalpha-disrupted mice, an ERalpha splice variant may also mediate the induction of PR-immunoreactivity in ERalpha-disrupted mice.


Subject(s)
Estradiol/pharmacology , Prosencephalon/chemistry , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Ventromedial Hypothalamic Nucleus/chemistry , Animals , Brain Chemistry/drug effects , Female , Gene Expression/physiology , Male , Mice , Mice, Knockout , Ovariectomy , RNA, Messenger/analysis , Receptors, Progesterone/analysis
17.
J Clin Endocrinol Metab ; 83(11): 3965-72, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9814476

ABSTRACT

The physiological effects of estrogen on the pituitary, including cellular proliferation and regulation of hormone synthesis, are mediated by the nuclear estrogen receptor (ER). The ER acts as a dimer to modulate gene transcription and contains specific functional domains encoded in different exons. Two separate, but related, forms of the receptor (ERalpha and ERbeta) exist, with distinct tissue and cell patterns of expression. Additional ER isoforms, generated by alternative messenger ribonucleic acid (mRNA) exon splicing, have been defined in several tissues and are postulated to play a role in tumorigenesis or in modulating the estrogen response. We examined 71 human pituitary adenomas of varying phenotypes and 6 normal pituitary specimens for ER mRNA forms by RT-PCR and hybridization blotting analysis. All prolactinomas (n = 14) contained ERalpha, and several contained ERbeta (5 of 14) mRNA. In comparison, 6 tumors that expressed PRL and GH expressed ERbeta (4 of 6) more frequently than ERalpha (3 of 6). ERbeta mRNA was also found more frequently in null cell (8 of 24 ERalpha and 14 of 24 ERbeta) and gonadotrope (13 of 21 ERalpha and 18 of 21 ERbeta) tumors. Additionally, ERbeta was found in 4 of 6 tumors that contained only GH, although ERalpha was not observed in this tumor type. Expression of the two ER forms within a tumor type was overlapping, but some tumors contained only 1 isoform. Expression of ERalpha mRNA splice variants also varied with cell type. All normal pituitaries contained ERalpha deletions of exon 4, 5, and 7, whereas only 2 of 6 samples contained the exon 2 deletion variant. Although the same ERalpha mRNA variants were observed among the various tumor types, the proportion of specific splice variants expressed varied. For example, most ER-positive prolactinomas expressed ERalpha variants with deletions of exon 2, 4, or 5, whereas gonadotropin tumors preferentially expressed the ERalpha exon 7 deletion variant. A novel ERbeta mRNA splice variant, missing exon 2, was observed in a majority of all ERbeta-positive tumors. Immunoblotting analysis of ERalpha and ERbeta proteins supported the mRNA results. Because ERalpha and ERbeta have different biological responses to selective ER modulators, and the ER deletion variants have biological effects distinct from those of the full-length ER, expression of these isoforms may influence the biological properties of these tumors and affect their ability to respond to estrogen and antiestrogen therapies.


Subject(s)
Adenoma/metabolism , Pituitary Neoplasms/metabolism , Receptors, Estrogen/biosynthesis , Adult , Aged , Estrogen Receptor alpha , Estrogen Receptor beta , Exons , Female , Humans , Male , Middle Aged , Prolactinoma/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
18.
Endocrinology ; 139(10): 4164-74, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751496

ABSTRACT

Hormone-dependent breast cancer responds to primary therapies that block estrogen production or action, but tumor regrowth often occurs 12-18 months later. Additional hormonal treatments that further reduce estrogen synthesis or more effectively block its action cause additional remissions, but the mechanisms responsible for these secondary responses are not well understood. As a working hypothesis, we postulated that primary hormonal therapy induces adaptive changes, resulting in enhanced estrogen receptor (ER) expression and target gene activation and, further, that secondary treatment modalities interfere with these receptor-mediated transcriptional pathways. To test this hypothesis, we used an MCF-7 breast cancer model system involving deprivation of estradiol in culture for a prolonged period. These long-term estradiol-deprived (LTED) cells adapt by acquiring the ability to regrow in the absence of added estradiol. The experimental paradigm involved the comparison of wild-type cells with LTED cells. As endpoints, we directly assessed ER expression at the messenger RNA-, protein-, and ligand-binding levels and ER functionality by quantitating reporter gene activation and expression of endogenous estrogen target gene messenger RNA, as well as ER coactivator levels. Our data demonstrated an adaptive increase in ER expression and in basal ER functionality, as assessed by read-out of three different transfected reporters in LTED, as opposed to wild-type MCF-7 cells. Increased reporter gene read-out was dramatically inhibited by the pure antiestrogen ICI 182,780. As verification that endogenous (as well as transfected) estrogen target genes had enhanced transcription, we found that the basal levels of c-myb and c-myc message were substantially increased in LTED cells and could be inhibited by antiestrogen. Interestingly, the levels of c-myb and c-myc message in the LTED cells seemed to be increased out of proportion to the degree of ER reporter gene activation and were similar to those in wild-type cells maximally stimulated with estradiol. In addition, not all estrogen-responsive genes were activated, because transforming growth factor-alpha message level was not increased in LTED cells. Up-regulation of the steroid receptor coactivator SRC-1 did not seem to mediate the process of enhanced ER-induced transcription. Considering these observations together, we suggest that long-term estradiol deprivation causes adaptive processes that not only involve up-regulation of the ER but also influence the specificity and magnitude of activation of estrogen-responsive genes.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/physiology , Receptors, Estrogen/physiology , Breast Neoplasms/pathology , Cell Division , Female , Genes, myc , Humans , Oncogenes , RNA, Messenger/analysis , Tumor Cells, Cultured
19.
Mol Endocrinol ; 12(3): 451-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9514161

ABSTRACT

Gonadotropin secretion and gene expression are differentially regulated by hypothalamic GnRH pulses by unknown mechanisms. GnRH stimulates calcium influx through L-type voltage-gated channels and activates phospholipase C, leading to increased protein kinase C (PKC) and mitogen-activated protein kinase activity. We found differential contributions of these pathways to GnRH-stimulated rat LH subunit transcription in pituitary gonadotropes and cell lines. Endogenous transcription of the alpha- and LHbeta-subunits in rat pituitary cells was stimulated by GnRH. Independent PKC activation by phorbol myristate acid stimulated only the alpha-subunit gene. In contrast, an L-channel antagonist (nimodipine) inhibited only LHbeta stimulation by GnRH, and an L-channel agonist (BayK 8644) stimulated only basal LHbeta transcription. GnRH induction of a rat alpha-subunit promoter construct in alphaT3 cells was unaffected by nimodipine or elimination of external calcium, while both treatments eliminated the LHbeta response. Application of a mitogen-activated kinase kinase (MEK) inhibitor (PD098059) decreased basal and GnRH-stimulated alpha-subunit promoter activity and had no effect on LHbeta promoter activity. In pituitary cells from mice bearing an LHbeta promoter-luciferase reporter transgene, GnRH stimulation was inhibited by nimodipine but not by PD098059. Thus, GnRH induction and basal control of the alpha-subunit gene seem to occur through the PKC/mitogen-activated protein kinase pathway, while induction of the LHbeta gene is dependent on calcium influx. Differential signaling from the same receptor may be a mechanism for preferential regulation of transcription.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/metabolism , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/genetics , Transcription, Genetic , Animals , Cell Line , Female , Gonadotropin-Releasing Hormone/pharmacology , Luciferases/genetics , Luciferases/metabolism , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Pituitary Gland/physiology , Promoter Regions, Genetic , Protein Kinase C/metabolism , Rats , Rats, Inbred Strains , Signal Transduction , Transfection , Transgenes
20.
Mol Cell Endocrinol ; 131(2): 147-55, 1997 Aug 08.
Article in English | MEDLINE | ID: mdl-9296373

ABSTRACT

We have identified several estrogen receptor (ER) mRNA isoforms in rat pituitary and characterized their regulation by gonadal steroids. The ER mRNAs correspond to splice variants in which either exon 4, exons 3 and 4, or exons 5 and 6 are deleted. A previously isolated pituitary-specific truncated mRNA, TERP-1, containing a unique 5'-end and exons 5 through 8 of the full-length ER, was also studied. The exon deletion variants were expressed in males and females, in pituitary, uterus, testes, heart, hypothalamus, and liver. An antibody to the ER C-terminus bound to full-length (64 kDa) and smaller (50 55 kDa and 40-45 kDa) ER proteins in uterus and pituitary and a pituitary-specific ER of 20-24 kDa corresponding to TERP-1. Estrogen (E) treatment in vivo stimulated full-length ER 2-3-fold, and TERP-1 7-10-fold, but had no effect on any exon deletion variant. Progesterone treatment, alone or with E, had no consistent effect on any ER mRNA form. TERP-1 mRNA was also dramatically and specifically modulated during the estrous cycle, increasing approximately 500-fold between the morning of diestrous and the afternoon of proestrus. Thus, ER mRNA variants exist in estrogen-responsive tissues; the pituitary contains at least one tissue-specific ER which is regulated by steroids and which may contribute to changes in regulated biological activity.


Subject(s)
Estrogens/pharmacology , Estrus/physiology , Pituitary Gland/metabolism , Progesterone/pharmacology , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Animals , Female , Genetic Variation , Hypothalamus/metabolism , Liver/metabolism , Male , Myocardium/metabolism , Peptide Fragments/genetics , RNA Splicing , Rats , Testis/metabolism , Uterus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...