Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Oncogene ; 36(30): 4379, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28604749

ABSTRACT

This corrects the article DOI: 10.1038/onc.2016.394.

2.
Oncogene ; 36(30): 4235-4242, 2017 07 27.
Article in English | MEDLINE | ID: mdl-27869167

ABSTRACT

Despite the advances in the diagnosis and treatment of breast cancer, breast cancers still cause significant mortality. For some patients, especially those with triple-negative breast cancer, current treatments continue to be limited and ineffective. Therefore, there remains an unmet need for a novel therapeutic approach. One potential strategy is to target the altered metabolic state that is rewired by oncogenic transformation. Specifically, this rewiring may render certain outside nutrients indispensable. To identify such a nutrient, we performed a nutrigenetic screen by removing individual amino acids to identify possible addictions across a panel of breast cancer cells. This screen revealed that cystine deprivation triggered rapid programmed necrosis, but not apoptosis, in the basal-type breast cancer cells mostly seen in TNBC tumors. In contrast, luminal-type breast cancer cells are cystine-independent and exhibit little death during cystine deprivation. The cystine addiction phenotype is associated with a higher level of cystine-deprivation signatures noted in the basal type breast cancer cells and tumors. We found that the cystine-addicted breast cancer cells and tumors have strong activation of TNFα and MEKK4-p38-Noxa pathways that render them susceptible to cystine deprivation-induced necrosis. Consistent with this model, silencing of TNFα and MEKK4 dramatically reduces cystine-deprived death. In addition, the cystine addiction phenotype can be abrogated in the cystine-addictive cells by miR-200c, which converts the mesenchymal-like cells to adopt epithelial features. Conversely, the introduction of inducers of epithelial-mesenchymal transition (EMT) in cystine-independent breast cancer cells conferred the cystine-addiction phenotype by modulating the signaling components of cystine addiction. Together, our data reveal that cystine-addiction is associated with EMT in breast cancer during tumor progression. These findings provide the genetic and mechanistic basis to explain how cystine deprivation triggers necrosis by activating pre-existing oncogenic pathways in cystine-addicted TNBC with prominent mesenchymal features.


Subject(s)
Cysteine/metabolism , Epithelial-Mesenchymal Transition/physiology , Triple Negative Breast Neoplasms/pathology , Female , Humans , Necrosis/metabolism , Phenotype , Signal Transduction/physiology , Triple Negative Breast Neoplasms/metabolism
3.
Climacteric ; 17 Suppl 2: 60-5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25320023

ABSTRACT

Despite increased survivorship among patients, breast cancer remains the most common cancer among women and is the second leading cause of cancer death in women. The magnitude of this problem provides a strong impetus for new chemopreventative strategies and/or lifestyle changes that reduce cancer incidence. It is of significance, therefore, that several studies positively correlate obesity to the development of breast cancer. Importantly, obesity is also highly associated with elevated cholesterol, and cholesterol itself is a risk factor for breast cancer. Furthermore, patients taking statins demonstrate a lower breast cancer incidence and decreased recurrence. The recent observation that 27-hydroxycholesterol (27HC) is produced in a stoichiometric manner from cholesterol, together with our recent demonstration that it exerts partial agonist activity on both the estrogen and liver X receptors, suggested a potential mechanistic link between hyper-cholesterolemia and breast cancer incidence. Using genetic and pharmacological approaches, we have recently shown that elevation of circulating 27HC significantly increases tumor growth and metastasis in murine models of breast cancer. Further, we have demonstrated in appropriate animal models that the impact of high-fat diet on tumor pathogenesis can be mitigated by statins or by small molecule inhibitors of CYP27A1. These findings suggest that pharmacological or dietary modifications that lower total cholesterol, and by inference 27HC, are likely to reduce the impact of obesity/metabolic syndrome on breast cancer incidence.


Subject(s)
Breast Neoplasms/etiology , Cholesterol, Dietary/toxicity , Receptors, Estrogen/metabolism , Animals , Autocrine Communication/immunology , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Cholesterol/blood , Cholesterol, Dietary/blood , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Female , Humans , Hydroxycholesterols/blood , Hydroxycholesterols/chemical synthesis , Hyperlipidemias/complications , Hyperlipidemias/drug therapy , Incidence , Neoplasm Recurrence, Local , Obesity/complications , Paracrine Communication/immunology , Risk Factors , Selective Estrogen Receptor Modulators/metabolism
4.
Oncogene ; 33(7): 862-71, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-23435425

ABSTRACT

The androgen receptor (AR) has a critical role in the development and progression of prostate cancer (PC) and is a major therapeutic target in this disease. The transcriptional activity of AR is modulated by the coregulators with which it interacts, and consequently deregulation of cofactor expression and/or activity impacts the expression of genes whose products can have a role in PC pathogenesis. Here we report that E74-like factor 3 (ELF3), a member of the ETS family of transcription factors, is a repressor of AR transcriptional activity. Exogenous expression of ELF3 represses AR transcriptional activity when assessed using reporter-based transfection assays or when evaluated on endogenous AR target genes. Conversely, ELF3 knock down increases the AR transcriptional activity. Biochemical dissection of this activity indicates that it results from the physical interaction between ELF3 and AR and that this interaction inhibits the recruitment of AR to specific androgen response elements within target gene promoters. Significantly, we observed that depletion of ELF3 expression in LNCaP cells promotes cell migration, whereas increased ELF3 expression severely inhibits tumor growth in vitro and in a mouse xenograft model. Taken together, these results suggest that modulation of ELF3 expression and/or AR/ELF3 interaction may have utility in the treatment of PC.


Subject(s)
DNA-Binding Proteins/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/physiology , Receptors, Androgen/metabolism , Transcription Factors/physiology , Animals , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-ets , Repressor Proteins/physiology , Response Elements , Transcription Factors/chemistry , Transcription, Genetic , Tumor Burden
5.
Eur Psychiatry ; 26(5): 313-9, 2011.
Article in English | MEDLINE | ID: mdl-20621454

ABSTRACT

OBJECTIVE: Adding another antipsychotic to a treatment regimen was previously used in evaluating the medication's efficacy. Supplementation of depot antipsychotics with oral antipsychotics is particularly meaningful because depot formulations are typically chosen for patients struggling with adherence to oral antipsychotics. This post-hoc analysis assessed supplementation of olanzapine long-acting injection (olanzapine-LAI) with oral olanzapine. SUBJECTS AND METHODS: We used 12 months of data from an open-label, single-arm extension study of patients with schizophrenia or schizoaffective disorder (N=931) treated with olanzapine-LAI. The prevalence, duration, time to first supplementation, and best predictors of oral supplementation were assessed. RESULTS: Oral supplementation occurred in 21% of patients for a median of 31 days with mean modal dose of 10.8 mg/day. Mean time to first supplementation was shorter for patients who were at least moderately ill at baseline compared to less ill patients (47 vs. 97 days, p<0.001). Best predictors of oral supplementation included a more severe illness profile at baseline, lower olanzapine-LAI dose prior to oral supplementation, supervised living arrangements, and being African-American. CONCLUSION: Supplementation of olanzapine-LAI appears to be infrequent, of relatively short duration, and reserved for more severely ill patients who may require a targeted rescue medication due to signs of impending relapse.


Subject(s)
Antipsychotic Agents/administration & dosage , Benzodiazepines/administration & dosage , Schizophrenia/drug therapy , Administration, Oral , Adult , Antipsychotic Agents/therapeutic use , Benzodiazepines/therapeutic use , Drug Administration Schedule , Female , Humans , Injections , Male , Middle Aged , Olanzapine , Severity of Illness Index
6.
Endocr Relat Cancer ; 13 Suppl 1: S25-32, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17259555

ABSTRACT

The orphan receptor estrogen-related receptor alpha (ERR alpha) is a member of the nuclear receptor superfamily of ligand-regulated transcription factors. This protein is structurally most related to the canonical estrogen receptor and has been shown to modulate estrogen signaling in some contexts. These observations have heightened interest in ERR alpha as a therapeutic target in both breast and ovarian cancer and in other estrogenopathies. This review details our present understanding of ERR alpha action with a view to highlight specific aspects of its signal-transduction pathway in breast cancer that may be amenable to pharmaceutical manipulation.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Neoplasms/drug therapy , Receptors, Estrogen/antagonists & inhibitors , Animals , Bone and Bones/physiology , Energy Metabolism/physiology , Homeostasis , Humans , Models, Biological , Neoplasms/etiology , Receptor Cross-Talk , Receptors, Estrogen/physiology , ERRalpha Estrogen-Related Receptor
7.
Mol Endocrinol ; 16(3): 487-96, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11875107

ABSTRACT

Some aspects of ligand-regulated transcription activation by the estrogen receptor (ER) are associated with the estrogen-dependent formation of a hydrophobic cleft on the receptor surface. At least in vitro, this cleft is required for direct interaction of ER with an alpha helix, containing variants of the sequence LXXLL, found in many coactivators. In cells, it is unknown whether ER interactions with the different LXXLL-containing helices are uniformly similar or whether they vary with LXXLL sequence or activating ligand. Using fluorescence resonance energy transfer (FRET), we confirm in the physiological environment a direct interaction between the estradiol (E2)-bound ER and LXXLL peptides expressed in living cells as fusions with spectral variants of the green fluorescent protein. This interaction was blocked by a single amino acid mutation in the hydrophobic cleft. No FRET was detected when cells were incubated with the antiestrogenic ligands tamoxifen and ICI 182,780. E2, diethylstilbestrol, ethyl indenestrol A, and 6,4'-dihydroxyflavone all promoted FRET and activated ER-dependent transcription. Measurement of the level of FRET of ER with different LXXLL-containing peptides suggested that the orientations or affinities of the LXXLL interactions with the hydrophobic cleft were globally similar but slightly different for some activating ligands.


Subject(s)
Energy Transfer , Estradiol/analogs & derivatives , Receptors, Estrogen/metabolism , Spectrometry, Fluorescence , Cell Line , Diethylstilbestrol/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha , Fulvestrant , Green Fluorescent Proteins , Ligands , Luminescent Proteins/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Structure, Secondary , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Response Elements , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/metabolism , Tamoxifen/pharmacology , Transcription, Genetic , Red Fluorescent Protein
8.
Cancer Res ; 61(24): 8712-7, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751389

ABSTRACT

The pathophysiological mechanism(s) by which androgen independence develops in prostate cancer remains to be determined. The identification in many prostate cancer specimens of a mutant androgen receptor, T877A, with altered ligand specificity has provided an explanation for some treatment failures. The T877A mutant androgen receptor recognizes a number of nonandrogenic compounds, including certain estrogens, progestins, and even antiandrogens as androgens. However, a comprehensive screen for hormonal agents which display agonist activity on this mutant has not been performed. In this study, we characterized this clinically important receptor mutant further and found that it can be activated by a wide range of compounds, including a number of endogenous glucocorticoids. Among the most clinically relevant compounds identified are DOC and corticosterone, both of which can effectively activate the mutant receptor at concentrations normally found in blood. Dexamethasone, a synthetic glucocorticoid frequently used in various contexts for prostate cancer therapy, is also recognized as an androgen by the mutant receptor. These unexpected findings suggest the need to: (a) reassess the role of adrenally derived glucocorticoids in prostate cancer disease progression; and (b) recognize the potential for iatrogenic stimulation of disease progression with certain glucocorticoid interventions.


Subject(s)
Androgens , Glucocorticoids/pharmacology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , Desoxycorticosterone/pharmacology , Dexamethasone/pharmacology , Humans , Male , Mutation , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Transfection , Tumor Cells, Cultured
9.
Endocrinology ; 142(11): 4721-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606437

ABSTRACT

We have developed a transgenic mouse that functions as a reporter of ER activity, termed ER action indicator (ERIN), by incorporating a transgene with an estrogen-responsive promoter (three copies of the vitellogenin estrogen response element with a minimal thymidine kinase promoter) linked to the reporter gene beta-galactosidase. Evaluation of ER activity in female ERIN mice demonstrated estrogen-inducible expression of the reporter gene in the uterus, pituitary, and hypothalamus; established targets of estrogen action. Importantly, we also identified ER activity in a number of nonclassical estrogen target tissues, including kidney, liver, adrenal, and thyroid gland. ERIN provides a system to measure the same end point (transgene regulation) in different target tissues, permitting separation of the contributions of cell- and promoter-specific factors in determining ER pharmacology. In this regard we observed that on this specific promoter the pituitary gland was 25-fold more sensitive than the uterus to the estrogen diethylstilbestrol, implying the existence of cell-specific factors that influence ligand sensitivity. Our studies also identified considerable difference in the efficacy and potency of ER ligands in the uterus when ER transcriptional activity was assayed vs. uterine weight gain. Specifically, we observed that the environmental estrogen bisphenol A was a potent agonist in stimulating ER transcriptional activity, whereas it exhibited little uterotropic activity. In contrast to bisphenol A, tamoxifen significantly increased uterine weight, but minimally induced ER reporter activity in this tissue. Given the results of these studies, we believe that ERIN will be a useful model to evaluate ER ligand pharmacology and will assist in defining the cellular and molecular mechanisms that determine agonist and antagonist activity.


Subject(s)
Estrogens/physiology , Mice, Transgenic/genetics , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Xenobiotics/pharmacology , 3T3 Cells , Animals , Benzhydryl Compounds , Diethylstilbestrol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Estrogens, Non-Steroidal/pharmacology , Female , Gene Expression , Genes, Reporter/drug effects , Ligands , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Phenols/pharmacology , Receptors, Estrogen/genetics , Tamoxifen/pharmacology , Tissue Distribution , Uterus/anatomy & histology
10.
J Biol Chem ; 276(38): 35684-92, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11473106

ABSTRACT

The human estrogen receptor alpha-isoform (ERalpha) is a nuclear transcription factor that displays a complex pharmacology. In addition to classical agonists and antagonists, the transcriptional activity of ERalpha can be regulated by selective estrogen receptor modulators, a new class of drugs whose relative agonist/antagonist activity is determined by cell context. It has been demonstrated that the binding of different ligands to ERalpha results in the formation of unique ERalpha-ligand conformations. These conformations have been shown to influence ERalpha-cofactor binding and, therefore, have a profound impact on ERalpha pharmacology. In this study, we demonstrate that the nature of the bound ligand also influences the stability of ERalpha, revealing an additional mechanism by which the pharmacological activity of a compound is determined. Of note we found that although all ERalpha-ligand complexes can be ubiquitinated and degraded by the 26 S proteasome in vivo, the mechanisms by which they are targeted for proteolysis appear to be different. Specifically, for agonist-activated ERalpha, an inverse relationship between transcriptional activity and receptor stability was observed. This relationship does not extend to selective estrogen receptor modulators and pure antagonists. Instead, it appears that with these compounds, the determinant of receptor stability is the ligand-induced conformation of ERalpha. We conclude that the different conformational states adopted by ERalpha in the presence of different ligands influence transcriptional activity directly by regulating cofactor binding and indirectly by modulating receptor stability.


Subject(s)
Estrogen Antagonists/pharmacology , Receptors, Estrogen/metabolism , Ubiquitins/metabolism , Estrogen Receptor alpha , Humans , Ligands , Receptors, Estrogen/agonists , Receptors, Estrogen/drug effects , Tumor Cells, Cultured
12.
J Biol Chem ; 276(28): 26324-31, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11342538

ABSTRACT

The ubiquitin-protein ligase (E3), hRPF1/Nedd4, is a component of the ubiquitin-proteasome pathway responsible for substrate recognition and specificity. Although previously characterized as a regulator of the stability of cytoplasmic proteins, hRPF1/Nedd4 has also been suggested to have a role in the nucleus. However, in light of the cytoplasmic localization of hRPF1/Nedd4, it is unclear whether bona fide nuclear substrates of hRPF1/Nedd4 exist, and if so, what mechanism may allow a cytoplasmic ubiquitin ligase to manifest nuclear activity. Our search for nuclear substrates led to the identification of the human proline-rich transcript, brain-expressed (hPRTB) protein, the ubiquitination and degradation of which is regulated by hRPF1/Nedd4. Interestingly, hPRTB colocalizes with the splicing factor SC35 in nuclear speckles. Finally, we demonstrate that hRPF1/Nedd4 is indeed capable of entering the nucleus; however, the presence of a functional Rev-like nuclear export sequence in hRPF1/Nedd4 ensures a predominant cytoplasmic localization. Cumulatively, these findings highlight a nuclear role for the ubiquitin ligase hRPF1/Nedd4 and underscore cytoplasmic/nuclear localization as an important regulatory component of hRPF1/Nedd4-substrate recognition.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Nucleus/metabolism , Ligases/metabolism , Ubiquitin-Protein Ligases , Amino Acid Sequence , Endosomal Sorting Complexes Required for Transport , HeLa Cells , Humans , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Nuclear Proteins/metabolism , Protein Transport , Substrate Specificity , Ubiquitins/metabolism
13.
Cancer Res ; 61(7): 2917-22, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306468

ABSTRACT

Tamoxifen inhibits estrogen receptor (ER) transcriptional activity by competitively inhibiting estradiol binding and inducing conformational changes in the receptor that may prevent its interaction with coactivators. In bone, the cardiovascular system, and some breast tumors, however, tamoxifen exhibits agonist activity, suggesting that the tamoxifen-ER complex is not recognized identically in all cells. We used phage display to demonstrate that the antiestrogen GW5638 induces a unique structural change in the ER. The biological significance of this conformational change was revealed in studies that demonstrated that tamoxifen-resistant breast tumor explants are not cross-resistant to GW5638. Because of these properties, this drug is currently being developed as a potential therapeutic for tamoxifen-resistant breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Cinnamates/pharmacology , Estrogen Receptor Modulators/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Receptors, Estrogen/drug effects , Stilbenes/pharmacology , Tamoxifen/pharmacology , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , Drug Interactions , Drug Resistance, Neoplasm , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Protein Conformation , Receptors, Estrogen/metabolism , Xenograft Model Antitumor Assays
14.
Ann N Y Acad Sci ; 949: 16-35, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11795348

ABSTRACT

The term Selective Estrogen Receptor Modulators (SERMs) has been used of late to describe a group of pharmaceuticals that manifest estrogen receptor (ER) agonist activity in some tissues, but that oppose estrogen action in others. Whereas the name describing this class of drugs is new, the concept is not. Indeed, compounds exhibiting tissue-selective ER agonist/antagonist properties have been around for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of these drugs and develop them as treatments for estrogenopathies where it is desirable to direct therapy to a specific estrogen-responsive target organ. This realization has provided the impetus for research in this area and has pushed the development and clinical use of this class of drugs. The objective of this review is to describe how the medical need for SERMs arose and how recent studies of the mechanism of action of the currently available drugs are paving the way for the development of novel drugs with improved selectivity.


Subject(s)
Receptors, Estrogen/agonists , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Estradiol/pharmacology , Female , Humans , Ligands , Molecular Sequence Data , Peptide Fragments/chemistry , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/physiology , Sequence Alignment , Tamoxifen/pharmacology
15.
Mol Endocrinol ; 14(12): 2010-23, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11117531

ABSTRACT

The biological actions of estrogen are manifest through two genetically distinct estrogen receptors (ER alpha and ER beta) that display nonidentical expression patterns in target tissues. The phenotypic alterations in response to estrogens in mice disrupted for either or both of these receptors are not identical, suggesting that each subtype plays a unique role in ER-action. However, the lack of subtype-specific agonists and antagonists has made it difficult to define the processes that are regulated by ER alpha and/or ER beta. Previously, we have reported the identification and characterization of a series of LXXLL-containing peptide antagonists that block estrogen signaling by preventing the association of ER alpha with required coactivators. As expected, given the similarity of the coactivator binding pockets among nuclear receptors, most of the peptide antagonists identified inhibited the activity of multiple receptors. However, by altering sequences flanking the core LXXLL motif, some receptor selectivity was afforded. Building on this observation, we have screened combinatorial phage libraries, expressing peptides in the format X7LXXLLX7, for peptides that interact in a specific manner with ER beta. Using this approach, a series of highly specific, potent peptide antagonists have been identified that efficiently inhibit ER beta-mediated estrogen signaling when introduced into target cells. Interestingly, in cells where both ER subtypes were expressed, these ER beta antagonists were capable of attenuating ER action, suggesting that ER alpha and ER beta do indeed form functional heterodimeric complexes. We believe that suitably formulated versions of these peptides can be used to study ER beta action in vitro and in vivo. In addition, the unanticipated specificity of the peptides identified should serve as an impetus to investigate the use of this approach to develop peptide antagonists of other nuclear receptors and unrelated transcription factors.


Subject(s)
Estrogen Antagonists/metabolism , Peptides/metabolism , Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Amino Acid Motifs , Cell Line , Dimerization , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Ligands , Peptide Library , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/antagonists & inhibitors , Sequence Homology, Amino Acid , Transcriptional Activation/drug effects , Two-Hybrid System Techniques
16.
Mol Endocrinol ; 14(12): 2024-39, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11117532

ABSTRACT

Ligand binding to estrogen receptor (ER) is presumed to regulate the type and timing of ER interactions with different cofactors. Using fluorescence microscopy in living cells, we characterized the recruitment of five different green fluorescent protein (GFP)-labeled ER-interacting peptides to the distinct subnuclear compartment occupied by blue fluorescent protein (BFP)-labeled ER alpha. Different ligands promoted the recruitment of different peptides. One peptide was recruited in response to estradiol (E2), tamoxifen, raloxifene, or ICI 182,780 incubation whereas other peptides were recruited specifically by E2 or tamoxifen. Peptides containing different sequences surrounding the ER-interacting motif LXXLL were recruited with different time courses after E2 addition. Complex temporal kinetics also were observed for recruitment of the full-length, ER cofactor glucocorticoid receptor-interacting protein 1 (GRIP1); rapid, E2-dependent recruitment of GRIP1 was blocked by mutation of the GRIP1 LXXLL motifs to LXXAA whereas slower E2 recruitment persisted for the GRIP1 LXXAA mutant. This suggested the presence of multiple, temporally distinct GRIP 1 recruitment mechanisms. E2 recruitment of GRIP1 and LXXLL peptides was blocked by coincubation with excess ICI 182,780. In contrast, preformed E2/ER/GRIP1 and E2/ER/LXXLL complexes were resistant to subsequent ICI 182,780 addition whereas ICI 182,780 dispersed preformed complexes containing the GRIP1 LXXAA mutant. This suggested that E2-induced LXXLL binding altered subsequent ligand/ER interactions. Thus, alternative, ligand-selective recruitment and dissociation mechanisms with distinct temporal sequences are available for ER alpha action in vivo.


Subject(s)
Cell Nucleus/metabolism , Estradiol/analogs & derivatives , Peptides/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Compartmentation , Cell Line , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Fulvestrant , Green Fluorescent Proteins , Indicators and Reagents/metabolism , Kinetics , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macromolecular Substances , Molecular Sequence Data , Nuclear Receptor Coactivator 2 , Protein Structure, Tertiary , Receptors, Estrogen/agonists , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology
17.
Mol Pharmacol ; 58(4): 852-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10999957

ABSTRACT

We previously demonstrated differential interactions of the methoxychlor metabolite 2,2-bis(p-hydroxyphenyl)-1,1, 1-trichloroethane (HPTE) with estrogen receptor alpha (ERalpha), ERbeta, and the androgen receptor (AR). In this study, we characterize the ERalpha, ERbeta, and AR activity of structurally related methoxychlor metabolites. Human hepatoma cells (HepG2) were transiently transfected with human ERalpha, ERbeta, and AR plus an appropriate steroid-responsive luciferase reporter vector. After transfection, cells were treated with various concentrations of HPTE or structurally related compounds in the presence (for detecting antagonism) and absence (for detecting agonism) of 17beta-estradiol and dihydrotestosterone. The monohydroxy analog of methoxychlor, as well as monohydroxy and dihydroxy analogs of 2, 2-bis(p-hydroxyphenyl)-1,1-dichloroethylene, had ERalpha agonist activity and ERbeta and AR antagonist activity similar to HPTE. The trihydroxy metabolite of methoxychlor displayed only weak ERalpha agonist activity and did not alter ERbeta or AR activities. Replacement of the trichloroethane or dichloroethylene group with a methyl group resulted in a compound with ERalpha and ERbeta agonist activity that retained antiandrogenic activities. This study identifies some of the structural requirements for ERalpha and ERbeta activity and demonstrates the complexity involved in determining the mechanism of action of endocrine-active chemicals that simultaneously act as agonists or antagonists through one or more hormone receptors.


Subject(s)
Methoxychlor/pharmacology , Phenols/pharmacology , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Dose-Response Relationship, Drug , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Humans , Insecticides/chemistry , Insecticides/pharmacology , Methoxychlor/chemistry , Phenols/chemistry , Receptors, Androgen/drug effects , Receptors, Estrogen/drug effects , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured
18.
Mol Cell Biol ; 20(9): 3102-15, 2000 May.
Article in English | MEDLINE | ID: mdl-10757795

ABSTRACT

The human progesterone receptor (PR) exists as two functionally distinct isoforms, hPRA and hPRB. hPRB functions as a transcriptional activator in most cell and promoter contexts, while hPRA is transcriptionally inactive and functions as a strong ligand-dependent transdominant repressor of steroid hormone receptor transcriptional activity. Although the precise mechanism of hPRA-mediated transrepression is not fully understood, an inhibitory domain (ID) within human PR, which is necessary for transrepression by hPRA, has been identified. Interestingly, although ID is present within both hPR isoforms, it is functionally active only in the context of hPRA, suggesting that the two receptors adopt distinct conformations within the cell which allow hPRA to interact with a set of cofactors that are different from those recognized by hPRB. In support of this hypothesis, we identified, using phage display technology, hPRA-selective peptides which differentially modulate hPRA and hPRB transcriptional activity. Furthermore, using a combination of in vitro and in vivo methodologies, we demonstrate that the two receptors exhibit different cofactor interactions. Specifically, it was determined that hPRA has a higher affinity for the corepressor SMRT than hPRB and that this interaction is facilitated by ID. Interestingly, inhibition of SMRT activity, by either a dominant negative mutant (C'SMRT) or histone deacetylase inhibitors, reverses hPRA-mediated transrepression but does not convert hPRA to a transcriptional activator. Together, these data indicate that the ability of hPRA to transrepress steroid hormone receptor transcriptional activity and its inability to activate progesterone-responsive promoters occur by distinct mechanisms. To this effect, we observed that hPRA, unlike hPRB, was unable to efficiently recruit the transcriptional coactivators GRIP1 and SRC-1 upon agonist binding. Thus, although both receptors contain sequences within their ligand-binding domains known to be required for coactivator binding, the ability of PR to interact with cofactors in a productive manner is regulated by sequences contained within the amino terminus of the receptors. We propose, therefore, that hPRA is transcriptionally inactive due to its inability to efficiently recruit coactivators. Furthermore, our experiments indicate that hPRA interacts efficiently with the corepressor SMRT and that this activity permits it to function as a transdominant repressor.


Subject(s)
Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Transcription, Genetic , Amino Acid Sequence , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Hormone Antagonists/pharmacology , Humans , Kinetics , Mifepristone/pharmacology , Molecular Sequence Data , Nuclear Receptor Co-Repressor 2 , Peptides/metabolism , Plasmids , Progesterone Congeners/pharmacology , Promegestone/pharmacology , Protein Binding , Protein Isoforms , Receptors, Progesterone/antagonists & inhibitors , Recombinant Proteins/metabolism , Repressor Proteins/metabolism
19.
J Biol Chem ; 275(21): 16302-8, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10748020

ABSTRACT

A transcriptional coactivator of the peroxisome proliferator-activated receptor-gamma (PPARgamma), PPARgamma-coactivator-1(PGC-1) interacts in a constitutive manner with the hinge domain of PPARgamma and enhances its transcriptional activity. In this study we demonstrate that PGC-1 is a coactivator of estrogen receptor-alpha (ERalpha)-dependent transcriptional activity. However the mechanism by which PGC-1 interacts with ERalpha is different from that of PPARgamma. Specifically, it was determined that the carboxyl terminus of PGC-1 interacts in a ligand-independent manner with the ERalpha hinge domain. In addition, an LXXLL motif within the amino terminus of PGC-1 was shown to interact in an agonist-dependent manner with the AF2 domain within the carboxyl terminus of ERalpha. The ability of PGC-1 to associate with and potentiate the transcriptional activity of an ERalpha-AF2 mutant that is unable to interact with the p160 class of coactivators suggests that this coactivator may have a unique role in estrogen signaling. It is concluded from these studies that PGC-1 is a bona fide ERalpha coactivator, which may serve as a convergence point between PPARgamma and ERalpha signaling.


Subject(s)
Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Estrogen Receptor alpha , Humans , Mutation , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured
20.
J Soc Gynecol Investig ; 7(1 Suppl): S10-5, 2000.
Article in English | MEDLINE | ID: mdl-10732322

ABSTRACT

Until 1986, our understanding of estrogen receptor (ER) action was based on information derived from in vitro biochemical analyses and in vivo correlations. With the cloning of the human ER cDNA, the reconstitution of ER responsive transcription units in heterologous cells has permitted the genetic dissection of the ER signal transduction pathway. The recent discovery of ER beta and a multitude of adaptor proteins (coactivators and corepressors) has expanded the potential explanation for tissue-selective activities. The current concept of ER action includes a rheostat-like action of the receptor due to conformational changes in the ligand receptor complex that depend on the nature of the bound ligand. This conformational change also determines subsequent adaptor protein interactions. Recognition of the tissue-specific activities of tamoxifen, the first selective ER modulator (SERM), led to the development of new SERMs (raloxifene and toremifene) with greater tissue selectivities. A knowledge of the key adaptor proteins expressed within each ER target cell will allow mechanism-based screening of selective ER modulators. These future "designer estrogens" of the next millennium will be used for specific applications in the central nervous, cardiovascular, bone and reproductive systems.


Subject(s)
Estrogen Replacement Therapy , Selective Estrogen Receptor Modulators , Female , Humans , Protein Conformation , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...