1
|
Dougherty EJ, Elinoff JM, Ferreyra GA, Hou A, Cai R, Sun J, Blaine KP, Wang S, Danner RL. Mineralocorticoid Receptor (MR) trans-Activation of Inflammatory AP-1 Signaling: DEPENDENCE ON DNA SEQUENCE, MR CONFORMATION, AND AP-1 FAMILY MEMBER EXPRESSION. J Biol Chem 2016; 291:23628-23644. [PMID: 27650495 DOI: 10.1074/jbc.m116.732248] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Indexed: 01/21/2023] Open
Abstract
Glucocorticoids are commonly used to treat inflammatory disorders. The glucocorticoid receptor (GR) can tether to inflammatory transcription factor complexes, such as NFκB and AP-1, and trans-repress the transcription of cytokines, chemokines, and adhesion molecules. In contrast, aldosterone and the mineralocorticoid receptor (MR) primarily promote cardiovascular inflammation by incompletely understood mechanisms. Although MR has been shown to weakly repress NFκB, its role in modulating AP-1 has not been established. Here, the effects of GR and MR on NFκB and AP-1 signaling were directly compared using a variety of ligands, two different AP-1 consensus sequences, GR and MR DNA-binding domain mutants, and siRNA knockdown or overexpression of core AP-1 family members. Both GR and MR repressed an NFκB reporter without influencing p65 or p50 binding to DNA. Likewise, neither GR nor MR affected AP-1 binding, but repression or activation of AP-1 reporters occurred in a ligand-, AP-1 consensus sequence-, and AP-1 family member-specific manner. Notably, aldosterone interactions with both GR and MR demonstrated a potential to activate AP-1. DNA-binding domain mutations that eliminated the ability of GR and MR to cis-activate a hormone response element-driven reporter variably affected the strength and polarity of these responses. Importantly, MR modulation of NFκB and AP-1 signaling was consistent with a trans-mechanism, and AP-1 effects were confirmed for specific gene targets in primary human cells. Steroid nuclear receptor trans-effects on inflammatory signaling are context-dependent and influenced by nuclear receptor conformation, DNA sequence, and the expression of heterologous binding partners. Aldosterone activation of AP-1 may contribute to its proinflammatory effects in the vasculature.
Collapse
Affiliation(s)
- Edward J Dougherty
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Jason M Elinoff
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Gabriela A Ferreyra
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Angela Hou
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Rongman Cai
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Junfeng Sun
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Kevin P Blaine
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Shuibang Wang
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert L Danner
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
2
|
Schöne S, Jurk M, Helabad MB, Dror I, Lebars I, Kieffer B, Imhof P, Rohs R, Vingron M, Thomas-Chollier M, Meijsing SH. Sequences flanking the core-binding site modulate glucocorticoid receptor structure and activity. Nat Commun 2016; 7:12621. [PMID: 27581526 PMCID: PMC5025757 DOI: 10.1038/ncomms12621] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
The glucocorticoid receptor (GR) binds as a homodimer to genomic response elements, which have particular sequence and shape characteristics. Here we show that the nucleotides directly flanking the core-binding site, differ depending on the strength of GR-dependent activation of nearby genes. Our study indicates that these flanking nucleotides change the three-dimensional structure of the DNA-binding site, the DNA-binding domain of GR and the quaternary structure of the dimeric complex. Functional studies in a defined genomic context show that sequence-induced changes in GR activity cannot be explained by differences in GR occupancy. Rather, mutating the dimerization interface mitigates DNA-induced changes in both activity and structure, arguing for a role of DNA-induced structural changes in modulating GR activity. Together, our study shows that DNA sequence identity of genomic binding sites modulates GR activity downstream of binding, which may play a role in achieving regulatory specificity towards individual target genes.
Collapse
Affiliation(s)
- Stefanie Schöne
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, Ihnestrasse 63-73, Berlin 14195, Germany
| | - Marcel Jurk
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, Ihnestrasse 63-73, Berlin 14195, Germany
| | | | - Iris Dror
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089, USA
| | - Isabelle Lebars
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Biologie Structurale, Centre National de la Recherche Scientifique (CNRS) UMR 7104/Institute National de la Santé et de la Recherche Médicale (INSERM) U964/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France
| | - Bruno Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Biologie Structurale, Centre National de la Recherche Scientifique (CNRS) UMR 7104/Institute National de la Santé et de la Recherche Médicale (INSERM) U964/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France
| | - Petra Imhof
- Institute of Theoretical Physics, Free University Berlin, 14195 Berlin, Germany
| | - Remo Rohs
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089, USA
| | - Martin Vingron
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, Ihnestrasse 63-73, Berlin 14195, Germany
| | - Morgane Thomas-Chollier
- Institut de Biologie de l'Ecole Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, U1024, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, F-75005 Paris, France
| | - Sebastiaan H Meijsing
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, Ihnestrasse 63-73, Berlin 14195, Germany
| |
Collapse
|
3
|
Kuo T, Liu PH, Chen TC, Lee RA, New J, Zhang D, Lei C, Chau A, Tang Y, Cheung E, Wang JC. Transcriptional regulation of FoxO3 gene by glucocorticoids in murine myotubes. Am J Physiol Endocrinol Metab 2016; 310:E572-85. [PMID: 26758684 PMCID: PMC4824139 DOI: 10.1152/ajpendo.00214.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
Abstract
Glucocorticoids and FoxO3 exert similar metabolic effects in skeletal muscle. FoxO3 gene expression was increased by dexamethasone (Dex), a synthetic glucocorticoid, both in vitro and in vivo. In C2C12 myotubes the increased expression is due to, at least in part, the elevated rate of FoxO3 gene transcription. In the mouse FoxO3 gene, we identified three glucocorticoid receptor (GR) binding regions (GBRs): one being upstream of the transcription start site, -17kbGBR; and two in introns, +45kbGBR and +71kbGBR. Together, these three GBRs contain four 15-bp glucocorticoid response elements (GREs). Micrococcal nuclease (MNase) assay revealed that Dex treatment increased the sensitivity to MNase in the GRE of +45kbGBR and +71kbGBR upon 30- and 60-min Dex treatment, respectively. Conversely, Dex treatment did not affect the chromatin structure near the -17kbGBR, in which the GRE is located in the linker region. Dex treatment also increased histone H3 and/or H4 acetylation in genomic regions near all three GBRs. Moreover, using chromatin conformation capture (3C) assay, we showed that Dex treatment increased the interaction between the -17kbGBR and two genomic regions: one located around +500 bp and the other around +73 kb. Finally, the transcriptional coregulator p300 was recruited to all three GBRs upon Dex treatment. The reduction of p300 expression decreased FoxO3 gene expression and Dex-stimulated interaction between distinct genomic regions of FoxO3 gene identified by 3C. Overall, our results demonstrate that glucocorticoids activated FoxO3 gene transcription through multiple GREs by chromatin structural change and DNA looping.
Collapse
Affiliation(s)
- Taiyi Kuo
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Patty H Liu
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Tzu-Chieh Chen
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Rebecca A Lee
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Jenny New
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Danyun Zhang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Cassandra Lei
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Andy Chau
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Yicheng Tang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Edna Cheung
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Jen-Chywan Wang
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| |
Collapse
|
4
|
Beck IM, De Bosscher K, Haegeman G. Glucocorticoid receptor mutants: man-made tools for functional research. Trends Endocrinol Metab 2011; 22:295-310. [PMID: 21549614 DOI: 10.1016/j.tem.2011.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 03/20/2011] [Accepted: 03/31/2011] [Indexed: 12/18/2022]
Abstract
The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that can bind to glucocorticoids (GCs). Upon ligand binding, GR sheds its cytoplasmic chaperoning complex and translocates to the nucleus, where it can act as a ligand-dependent transcription factor, transactivating or transrepressing specific gene promoters. Often, GR interacts with specific cofactors to implement a variety of gene promoter effects. GR activity and function is further modulated by post-translational modifications. To assess the diverse aspects of GR mechanisms of activation and gene regulation, researchers continue to use a range of artificial GR mutants. In this review we analyze the characteristics of GR mutants with the aim of assisting the design and interpretation of GR mutant-based experiments.
Collapse
Affiliation(s)
- Ilse M Beck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Physiology, Ghent University, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium.
| | | | | |
Collapse
|
5
|
Kumar R, Calhoun WJ. Differential regulation of the transcriptional activity of the glucocorticoid receptor through site-specific phosphorylation. Biologics 2011; 2:845-54. [PMID: 19707462 PMCID: PMC2727889 DOI: 10.2147/btt.s3820] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Post-translational modifications such as phosphorylation are known to play an important role in the gene regulation by the transcription factors including the nuclear hormone receptor superfamily of which the glucocorticoid receptor (GR) is a member. Protein phosphorylation often switches cellular activity from one state to another. Like many other transcription factors, the GR is a phosphoprotein, and phosphorylation plays an important role in the regulation of GR activity. Cell signaling pathways that regulate phosphorylation of the GR and its associated proteins are important determinants of GR function under various physiological conditions. While the role of many phosphorylation sites in the GR is still not fully understood, the role of others is clearer. Several aspects of transcription factor function, including DNA binding affinity, interaction of transactivation domains with the transcription initiation complex, and shuttling between the cytoplasmic compartments, have all been linked to site-specific phosphorylation. All major phosphorylation sites in the human GR are located in the N-terminal domain including the major transactivation domain, AF1. Available literature clearly indicates that many of these potential phosphorylation sites are substrates for multiple kinases, suggesting the potential for a very complex regulatory network. Phosphorylated GR interacts favorably with critical coregulatory proteins and subsequently enhances transcriptional activity. In addition, the activities and specificities of coregulators may be subject to similar regulation by phosphorylation. Regulation of the GR activity due to phosphorylation appears to be site-specific and dependent upon specific cell signaling cascade. Taken together, site-specific phosphorylation and related kinase pathways play an important role in the action of the GR, and more precise mechanistic information will lead to fuller understanding of the complex nature of gene regulation by the GR- and related transcription factors. This review provides currently available information regarding the role of GR phosphorylation in its action, and highlights the possible underlying mechanisms of action.
Collapse
|
6
|
Sharma R, Prichard D, Majer F, Byrne AM, Kelleher D, Long A, Gilmer JF. Ursodeoxycholic acid amides as novel glucocorticoid receptor modulators. J Med Chem 2010; 54:122-30. [PMID: 21158453 DOI: 10.1021/jm100860s] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ursodeoxycholic acid (UDCA) is used for the treatment of hepatic inflammatory diseases. Recent studies have shown that UDCA's biological effects are partly glucocorticoid receptor (GR) mediated. UDCA derivatives were synthesized and screened for ability to induce GR translocation in a high content analysis assay using the esophageal cancer SKGT-4 cell line. UDCA derivatives induced GR translocation in a time dependent manner with equal efficacy to that of dexamethasone (Dex) and with greatly increased potency relative to UDCA. The cyclopropylamide 1a suppressed TNF-α induced NF-κB activity and it induced GRE transactivation. 1a was unable to displace Dex from the GR ligand binding domain (LBD) in a competition experiment but was capable of coactivator recruitment in a time-resolved fluorescence energy transfer assay (TR-FRET). This represents a novel mechanism of action for a GR modulator. These derivatives could result in a new class of GR modulators.
Collapse
Affiliation(s)
- Ruchika Sharma
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
7
|
Chan IH, Privalsky ML. A conserved lysine in the thyroid hormone receptor-alpha1 DNA-binding domain, mutated in hepatocellular carcinoma, serves as a sensor for transcriptional regulation. Mol Cancer Res 2010; 8:15-23. [PMID: 20053725 DOI: 10.1158/1541-7786.mcr-09-0425] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nuclear receptors are hormone-regulated transcription factors that play key roles in normal physiology and development; conversely, mutant nuclear receptors are associated with a wide variety of neoplastic and endocrine disorders. Typically, these receptor mutants function as dominant negatives and can interfere with wild-type receptor activity. Dominant-negative thyroid hormone receptor (TR) mutations have been identified in over 60% of the human hepatocellular carcinomas analyzed. Most of these mutant TRs are defective for corepressor release or coactivator binding in vitro, accounting for their transcriptional defects in vivo. However, two HCC-TR mutants that function as dominant-negative receptors in cells display near-normal properties in vitro, raising questions about the molecular basis behind their transcriptional defects. We report here that a single amino acid substitution, located at the same position in the DNA-binding domain of both mutants, is responsible for their impaired transcriptional activation and dominant-negative properties. Significantly, this amino acid, K74 in TRalpha, is highly conserved in all known nuclear receptors and seems to function as an allosteric sensor that regulates the transcriptional activity of these receptors in response to binding to their DNA recognition sequences. We provide evidence that these two human hepatocellular carcinoma mutants have acquired dominant-negative function as a result of disruption of this allosteric sensing. Our results suggest a novel mechanism by which nuclear receptors can acquire transcriptional defects and contribute to neoplastic disease.
Collapse
Affiliation(s)
- Ivan H Chan
- Department of Microbiology, One Shields Avenue, University of California at Davis, Davis, CA 95616, USA
| | | |
Collapse
|
8
|
Chan IH, Privalsky ML. Thyroid hormone receptor mutants implicated in human hepatocellular carcinoma display an altered target gene repertoire. Oncogene 2009; 28:4162-74. [PMID: 19749797 PMCID: PMC2787677 DOI: 10.1038/onc.2009.265] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Thyroid hormone receptors (TRs) are hormone-regulated transcription factors that control multiple aspects of normal physiology and development. Mutations in TRs have been identified at high frequency in certain cancers, including human hepatocellular carcinomas (HCCs). The majority of HCC-TR mutants bear lesions within their DNA recognition domains, and we have hypothesized that these lesions change the mutant receptors' target gene repertoire in a way crucial to their function as oncoproteins. Using stable cell transformants and expression array analysis, we determined that mutant TRs isolated from two different HCCs do, as hypothesized, display a target gene repertoire distinct from that of their normal TR progenitors. Only a subset of genes regulated by wild-type TRs was regulated by the corresponding HCC-TR mutants. More surprisingly, the HCC-TR mutants also gained the ability to regulate additional target genes not recognized by the wild-type receptors, and were not simply restricted to repression, but could also activate a subset of their target genes. We conclude that the TR mutants isolated from HCC have sustained multiple alterations from their normal progenitors that include not only changes in their transcriptional outputs, but also changes in the genes they target; both are likely to contribute to neoplasia.
Collapse
Affiliation(s)
- I H Chan
- Department of Microbiology, College of Biological Sciences, University of California at Davis, Davis, CA 95616, USA
| | | |
Collapse
|
9
|
Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 2009; 89:147-91. [PMID: 19126757 DOI: 10.1152/physrev.00010.2008] [Citation(s) in RCA: 1211] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The incidence of the metabolic syndrome has taken epidemic proportions in the past decades, contributing to an increased risk of cardiovascular disease and diabetes. The metabolic syndrome can be defined as a cluster of cardiovascular disease risk factors including visceral obesity, insulin resistance, dyslipidemia, increased blood pressure, and hypercoagulability. The farnesoid X receptor (FXR) belongs to the superfamily of ligand-activated nuclear receptor transcription factors. FXR is activated by bile acids, and FXR-deficient (FXR(-/-)) mice display elevated serum levels of triglycerides and high-density lipoprotein cholesterol, demonstrating a critical role of FXR in lipid metabolism. In an opposite manner, activation of FXR by bile acids (BAs) or nonsteroidal synthetic FXR agonists lowers plasma triglycerides by a mechanism that may involve the repression of hepatic SREBP-1c expression and/or the modulation of glucose-induced lipogenic genes. A cross-talk between BA and glucose metabolism was recently identified, implicating both FXR-dependent and FXR-independent pathways. The first indication for a potential role of FXR in diabetes came from the observation that hepatic FXR expression is reduced in animal models of diabetes. While FXR(-/-) mice display both impaired glucose tolerance and decreased insulin sensitivity, activation of FXR improves hyperglycemia and dyslipidemia in vivo in diabetic mice. Finally, a recent report also indicates that BA may regulate energy expenditure in a FXR-independent manner in mice, via activation of the G protein-coupled receptor TGR5. Taken together, these findings suggest that modulation of FXR activity and BA metabolism may open new attractive pharmacological approaches for the treatment of the metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Philippe Lefebvre
- Institut National de la Sante et de la Recherche Medicale, Lille, France
| | | | | | | | | |
Collapse
|
10
|
Shimizu N, Yoshikawa N, Wada T, Handa H, Sano M, Fukuda K, Suematsu M, Sawai T, Morimoto C, Tanaka H. Tissue- and context-dependent modulation of hormonal sensitivity of glucocorticoid-responsive genes by hexamethylene bisacetamide-inducible protein 1. Mol Endocrinol 2008; 22:2609-23. [PMID: 18801933 DOI: 10.1210/me.2008-0101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Physiological and pharmacological processes mediated by glucocorticoids involve tissue- and context-specific regulation of glucocorticoid-responsive gene expression via glucocorticoid receptor (GR). However, the molecular mechanisms underlying such highly coordinated regulation of glucocorticoid actions remain to be studied. We here addressed this issue using atp1a1 and scnn1a, both of which are up-regulated in response to corticosteroids in human embryonic kidney-derived 293 cells, but resistant in liver-derived HepG2 cells. Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) represses gene expression via, at least, two distinct mechanisms, i.e. positive transcription elongation factor b sequestration and direct interaction with GR, and is relatively high in HepG2 cells compared with 293 cells. Given this, we focused on the role of HEXIM1 in transcriptional regulation of these GR target genes. In HepG2 cells, hormone resistance of atp1a1 and scnn1a was diminished by either knockdown of HEXIM1 or overexpression of GR. Such a positive effect of exogenous expression of GR was counteracted by concomitant overexpression of HEXIM1, indicating the balance between GR and HEXIM1 modulates hormonal sensitivity of these genes. In support of this, the hormone-dependent recruitment of RNA polymerase II onto atp1a1 promoter was in parallel with that of GR. Moreover, we revealed that not positive transcription elongation factor b-suppressing activity but direct interaction with GR of HEXIM1 plays a major role in suppression of promoter recruitment of the receptor and subsequent atp1a1 and scnn1a gene activation. Collectively, we may conclude that HEXIM1 may participate in tissue-selective determination of glucocorticoid sensitivity via direct interaction with GR at least in certain gene sets including atp1a1 and scnn1a.
Collapse
Affiliation(s)
- Noriaki Shimizu
- Division of Clinical Immunology, Advanced Clinical Research Center, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Haffner MC, Jurgeit A, Berlato C, Geley S, Parajuli N, Yoshimura A, Doppler W. Interaction and functional interference of glucocorticoid receptor and SOCS1. J Biol Chem 2008; 283:22089-96. [PMID: 18524780 DOI: 10.1074/jbc.m801041200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytokine and glucocorticoid (GC) hormone signaling act in an integrated fashion to control inflammation and immune response. Here we establish a new mode of interaction of these two pathways and propose Suppressor of Cytokine Signaling (SOCS)-1 as an essential player in mediating cross-talk. We observed that glucocorticoid receptor (GR) and SOCS1 form an intracellular complex through an interaction, which required the SH2 domain of SOCS1 and the ligand binding domain of GR. Furthermore, GC stimulation was found to increase the nuclear level of SOCS1. SOCS1 binding to the GR did not require ligand binding of the receptor; however, it was abolished after long term GC stimulation, suggesting a functional role of the interaction for the early phase of GC action. The interaction between GR and SOCS1 appeared to negatively influence the transcription of the two GR-regulated genes, FKBP5 and MKP1, because the GC-dependent expression of these genes was inhibited by the SOCS1 inducer IFNgamma and enhanced in SOCS1-deficient murine embryonic fibroblasts as compared with IFNgamma treated wild-type cells. Our results suggest a prominent role of SOCS1 in the early phase of cross-talk between GR and cytokine signaling.
Collapse
Affiliation(s)
- Michael C Haffner
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
12
|
Latchoumanin O, Mynard V, Devin-Leclerc J, Dugué MA, Bertagna X, Catelli MG. Reversal of glucocorticoids-dependent proopiomelanocortin gene inhibition by leukemia inhibitory factor. Endocrinology 2007; 148:422-32. [PMID: 17038560 DOI: 10.1210/en.2006-0460] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously have described molecular mechanisms converging at the Nur response element-signal transducer and activator of transcription (STAT) composite site responsible for synergistic activation of the proopiomelanocortin (POMC) gene promoter by leukemia inhibitory factor (LIF) and CRH. In this study, we asked how glucocorticoids (GC), the physiological negative regulators of POMC gene expression, modulate this synergism. In the corticotroph cell line AtT-20, the response of the wild-type promoter to LIF+CRH was barely inhibited by GC, whereas a distal promoter subregion (-414/-293) encompassing the Nur response element-STAT site and devoid of the negative GC-responsive element located in the proximal domain, displayed a cooperative response to LIF+dexamethasone (DEX) and LIF+CRH+DEX treatments. LIF+CRH-stimulated ACTH secretion was also inefficiently inhibited by DEX in the same cell line. This study was focused thereafter on LIF+DEX cooperativity, which may be responsible, on the wild-type promoter, for lack of negative regulation by DEX of the LIF+CRH synergy. The STAT1-3 low-affinity site, in the context of the (-414/-293) subregion of the POMC promoter, was found necessary and sufficient for transcriptional synergism between activated GC receptor (GR) and STAT1-3. Moreover the activities of reporters specific for STAT1-3 or GR were reciprocally enhanced by DEX or LIF. Single and sequential chromatin immunoprecipitations revealed 1) a STAT-dependent corecruitment of coactivators after LIF and LIF+DEX stimulation and 2) a more lasting recruitment of both STAT3 and GR in the same enhanceosome on the endogenous POMC promoter after LIF+DEX joint stimulation than after the single one. Such events may be responsible for a lack of repressive property of GR unmasked on the whole POMC promoter during LIF+CRH stimulation and may contribute to the tonicity of the hypothalamic-pituitary-adrenal axis during inflammatory-infectious diseases.
Collapse
Affiliation(s)
- Olivier Latchoumanin
- Département d'Endocrinologie, Métabolisme et Cancer, Institut Cochin, F-75014 Paris, France
| | | | | | | | | | | |
Collapse
|
13
|
Roessler S, Györy I, Imhof S, Spivakov M, Williams RR, Busslinger M, Fisher AG, Grosschedl R. Distinct promoters mediate the regulation of Ebf1 gene expression by interleukin-7 and Pax5. Mol Cell Biol 2006; 27:579-94. [PMID: 17101802 PMCID: PMC1800812 DOI: 10.1128/mcb.01192-06] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Early differentiation of B lymphocytes requires the function of multiple transcription factors that regulate the specification and commitment of the lineage. Loss- and gain-of-function experiments have provided important insight into the transcriptional control of B lymphopoiesis, whereby E2A was suggested to act upstream of EBF1 and Pax5 downstream of EBF1. However, this simple hierarchy cannot account for all observations, and our understanding of a presumed regulatory network, in which transcription factors and signaling pathways operate, is limited. Here, we show that the expression of the Ebf1 gene involves two promoters that are differentially regulated and generate distinct protein isoforms. We find that interleukin-7 signaling, E2A, and EBF1 activate the distal Ebf1 promoter, whereas Pax5, together with Ets1 and Pu.1, regulates the stronger proximal promoter. In the absence of Pax5, the function of the proximal Ebf1 promoter and accumulation of EBF1 protein are impaired and the replication timing and subcellular localization of the Ebf1 locus are altered. Taken together, these data suggest that the regulation of Ebf1 via distinct promoters allows for the generation of several feedback loops and the coordination of multiple determinants of B lymphopoiesis in a regulatory network.
Collapse
Affiliation(s)
- Stephanie Roessler
- Max Planck Institute of Immunobiology, Department of Cellular and Molecular Immunology, 79108 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen W, Rogatsky I, Garabedian MJ. MED14 and MED1 differentially regulate target-specific gene activation by the glucocorticoid receptor. Mol Endocrinol 2006; 20:560-72. [PMID: 16239257 DOI: 10.1210/me.2005-0318] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Mediator subunits MED14 and MED1 have been implicated in transcriptional regulation by the glucocorticoid receptor (GR) by acting through its activation functions 1 and 2. To understand the contribution of these Mediator subunits to GR gene-specific regulation, we reduced the levels of MED14 and MED1 using small interfering RNAs in U2OS-hGR osteosarcoma cells and examined the mRNA induction by dexamethasone of four primary GR target genes, interferon regulatory factor 8 (IRF8), ladinin 1, IGF-binding protein 1 (IGFBP1), and glucocorticoid-inducible leucine zipper (GILZ). We found that the GR target genes differed in their requirements for MED1 and MED14. GR-dependent mRNA expression of ladinin 1 and IRF8 required both MED1 and MED14, whereas induction of IGFBP1 mRNA by the receptor was dependent upon MED14, but not MED1. In contrast, GILZ induction by GR was largely independent of MED1 and MED14, but required the p160 cofactor transcriptional intermediary factor 2. Interestingly, we observed higher GR occupancy at GILZ than at the IGFBP1 or IRF8 glucocorticoid response element (GREs). In contrast, recruitment of MED14 compared with GR at IGFBP1 and IRF8 was higher than that observed at GILZ. At GILZ, GR and RNA polymerase II were recruited to both the GRE and the promoter, whereas at IGFBP1, RNA polymerase II occupied the promoter, but not the GRE. Thus, MED14 and MED1 are used by GR in a gene-specific manner, and the requirement for the Mediator at GILZ may be bypassed by increased GR and RNA polymerase II occupancy at the GREs. Our findings suggest that modulation of the Mediator subunit activities would provide a mechanism for promoter selectivity by GR.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Microbiology, New York University Cancer Institute, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | | | | |
Collapse
|
15
|
Chan IH, Privalsky ML. Thyroid hormone receptors mutated in liver cancer function as distorted antimorphs. Oncogene 2006; 25:3576-88. [PMID: 16434963 PMCID: PMC2701908 DOI: 10.1038/sj.onc.1209389] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aberrant thyroid hormone receptors (TRs) are found in over 70% of the human hepatocellular carcinomas (HCCs) analysed. To better understand the role(s) of these TR mutants in this neoplasia, we analysed a panel of HCC mutant receptors for their molecular properties. Virtually all HCC-associated TR mutants tested retained the ability to repress target genes in the absence of T3, yet were impaired in T3-driven gene activation and functioned as dominant-negative inhibitors of wild-type TR activity. Intriguingly, the HCC TRalpha1 mutants exerted dominant-negative interference at all T3 concentrations tested, whereas the HCC TRbeta1 mutants were dominant-negatives only at low and intermediate T3 concentrations, reverting to transcriptional activators at higher hormone levels. The relative affinity for the SMRT versus N-CoR corepressors was detectably altered for several of the HCC mutant TRs, suggesting changes in corepressor preference and recruitment compared to wild type. Several of the TRalpha HCC mutations also altered the DNA recognition properties of the encoded receptors, indicating that these HCC TR mutants may regulate a distinct set of target genes from those regulated by wild-type TRs. Finally, whereas wild-type TRs interfere with c-Jun/AP-1 function in a T3-dependent fashion and suppress anchorage-independent growth when ectopically expressed in HepG2 cells, at least certain of the HCC mutants did not exert these inhibitory properties. These alterations in transcriptional regulation and DNA recognition appear likely to contribute to oncogenesis by reprogramming the differentiation and proliferative properties of the hepatocytes in which the mutant TRs are expressed.
Collapse
Affiliation(s)
- I H Chan
- Section of Microbiology, University of California at Davis, 95616, USA
| | | |
Collapse
|
16
|
Kieslinger M, Folberth S, Dobreva G, Dorn T, Croci L, Erben R, Consalez GG, Grosschedl R. EBF2 Regulates Osteoblast-Dependent Differentiation of Osteoclasts. Dev Cell 2005; 9:757-67. [PMID: 16326388 DOI: 10.1016/j.devcel.2005.10.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Revised: 09/20/2005] [Accepted: 10/17/2005] [Indexed: 11/22/2022]
Abstract
Communication between bone-depositing osteoblasts and bone-resorbing osteoclasts is required for bone development and homeostasis. Here, we identify EBF2, a member of the early B cell factor (EBF) family of transcription factors that is expressed in osteoblast progenitors, as a regulator of osteoclast differentiation. We find that mice homozygous for a targeted inactivation of Ebf2 show reduced bone mass and an increase in the number of osteoclasts. These defects are accompanied by a marked downregulation of the osteoprotegerin (Opg) gene, encoding a RANK decoy receptor. EBF2 binds to sequences in the Opg promoter and transactivates the Opg promoter in synergy with the Wnt-responsive LEF1/TCF:beta-catenin pathway. Taken together, these data identify EBF2 as a regulator of RANK-RANKL signaling and osteoblast-dependent differentiation of osteoclasts.
Collapse
Affiliation(s)
- Matthias Kieslinger
- Gene center and Institute for Biochemistry, University of Munich, 81377 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The v-Erb A oncoprotein of avian erythroblastosis virus is derived from c-Erb A, a hormone-activated transcription factor. Notably, v-Erb A has sustained multiple mutations relative to c-Erb A and functions as a constitutive transcriptional repressor. We report here an analysis of the contributions of these different mutations to v-Erb A function. Our experiments demonstrate that two amino-acid differences between v-Erb A and c-Erb A, located in the 'I-box,' alter the dimerization properties of the viral protein, resulting in more stable homodimer formation, increased corepressor binding, and increased target gene repression. An additional amino-acid difference between v- and c-Erb A, located in helix 3 of the hormone binding domain, renders corepressor binding by the viral protein more resistant to release by thyroid hormone. Finally, we report that a C-terminal truncation in v-Erb A not only inhibits exchange of corepressor and coactivator, as previously noted, but also permits v-Erb A to recruit both SMRT and N-CoR corepressors, whereas c-Erb A is selective for N-CoR. The latter two mutations in v-Erb A also impair its ability to suppress c-Jun function in response to T3 hormone. We propose that the acquisition of oncogenic potential by the v-Erb A protein was a multistep process involving a series of mutations that alter the transcriptional repressive properties of the viral protein through multiple mechanisms.
Collapse
Affiliation(s)
- Sangho Lee
- Section of Microbiology, Division of Biological Sciences, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Martin L Privalsky
- Section of Microbiology, Division of Biological Sciences, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
- Correspondence: ML Privalsky; E-mail:
| |
Collapse
|
18
|
Geserick C, Meyer HA, Haendler B. The role of DNA response elements as allosteric modulators of steroid receptor function. Mol Cell Endocrinol 2005; 236:1-7. [PMID: 15876478 DOI: 10.1016/j.mce.2005.03.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Accepted: 03/23/2005] [Indexed: 11/25/2022]
Abstract
Steroid receptors are ligand-activated transcription factors which control the expression of their target genes by binding to specific DNA elements. Consensus response elements have been delineated for the glucocorticoid, androgen, progesterone and mineralocorticoid receptors on one hand (steroid response element, SRE) and for the estrogen receptor on the other hand (estrogen response element, ERE). Small variations in these sequences not only affect the binding but may also have a dramatic impact on the transcriptional activity of steroid receptors. It has now become obvious that DNA response elements do not merely tether regulatory proteins to control regions of target genes but may additionally impart conformational changes onto the DNA-binding domain as well as to neighbouring domains of steroid receptors. This in turn will create unique platforms for selective recruitment of cofactors and possibly for induction of modifications in local chromatin architecture. An additional level of complexity is added by the frequent presence of multiple response elements in gene promoter regions. The allosteric effects of DNA response elements on steroid receptors may be essential for differential gene expression and this offers interesting perspectives for the identification of selective modulators.
Collapse
|
19
|
Abstract
Steroidal glucocorticoids are commonly used due to their powerful antiinflammatory activity. However, despite their excellent efficacy, severe side effects frequently limit the use of these drugs. The search for novel glucocorticoids with reduced side effects has been intensified by the discovery of new molecular details regarding the function of the glucocorticoid receptor. These new insights may pave the way for novel, safer therapies that retain the efficacy of currently prescribed steroids.
Collapse
Affiliation(s)
- Jonathan Rosen
- Department of Molecular and Cell Biology, Ligand Pharmaceuticals, 10275 Science Center Drive, San Diego, California 92121, USA
| | | |
Collapse
|
20
|
Riml S, Schmidt S, Ausserlechner MJ, Geley S, Kofler R. Glucocorticoid receptor heterozygosity combined with lack of receptor auto-induction causes glucocorticoid resistance in Jurkat acute lymphoblastic leukemia cells. Cell Death Differ 2005; 11 Suppl 1:S65-72. [PMID: 15017388 DOI: 10.1038/sj.cdd.4401413] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Glucocorticoids (GC) induce apoptosis in malignant lymphoblasts, but the mechanism of this process as well as that of the clinically important GC resistance is unknown. We investigated GC resistance in Jurkat T-ALL cells in which ectopic GC receptor (GR) restores GC sensitivity, suggesting deficient GR expression. Jurkat cells expressed one wild-type and one mutated (R477H) GR allele. GR(R477H) ligand-binding-dependent nuclear import, as revealed by live-cell microscopy of YFP-tagged GR, was unaffected. Transactivation and transrepression were markedly impaired; however, GR(R477H) did not act in a dominant-negative manner, that is, did not prevent cell death, when introduced into a GC-sensitive cell line by retroviral gene transfer. Contrary to another GR heterozygous, but GC-sensitive, T-ALL model (CCRF-CEM), Jurkats expressed lower basal GR levels and did not auto-induce their GR, as revealed by 'real-time' RT-PCR and immunoblotting. Absent GR auto-induction could not be restored by transgenic GR and, hence, was not caused by reduced basal GR levels. Thus, inactivation of one GR gene results in haploinsufficiency if associated with lack of GR auto-induction.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Apoptosis/physiology
- Bacterial Proteins/genetics
- Bacterial Proteins/metabolism
- Base Sequence
- Blotting, Western
- COS Cells
- Cell Line, Tumor
- Chlorocebus aethiops
- Dexamethasone/pharmacology
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Genes, Dominant/genetics
- Glucocorticoids/pharmacology
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Heterozygote
- Humans
- Jurkat Cells
- Kinetics
- Luciferases, Renilla/genetics
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Mammary Tumor Virus, Mouse/genetics
- Microscopy, Confocal
- Point Mutation
- Protein Transport/drug effects
- Protein Transport/genetics
- Radioligand Assay
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/genetics
- Transcriptional Activation/drug effects
- Transcriptional Activation/genetics
- Transfection
- Triamcinolone/metabolism
Collapse
Affiliation(s)
- S Riml
- Division of Molecular Pathophysiology, Institute of Pathophysiology, University of Innsbruck Medical School, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria
| | | | | | | | | |
Collapse
|
21
|
Shah N, Scanlan TS. Design and evaluation of novel nonsteroidal dissociating glucocorticoid receptor ligands. Bioorg Med Chem Lett 2005; 14:5199-203. [PMID: 15380227 DOI: 10.1016/j.bmcl.2004.07.052] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 07/23/2004] [Accepted: 07/23/2004] [Indexed: 11/17/2022]
Abstract
A novel class of phenylpyrazole fused Wieland-Miescher ketone derivatives are high affinity, receptor specific, selective modulators of glucocorticoid receptor (GR) mediated transcription in vitro, dissociating transactivation, AP-1 repression, and NF-kappaB repression from each other.
Collapse
Affiliation(s)
- Nilesh Shah
- Graduate Group in Biophysics, University of California--San Francisco, San Francisco, CA 94143-2280, USA
| | | |
Collapse
|
22
|
Martens C, Bilodeau S, Maira M, Gauthier Y, Drouin J. Protein-protein interactions and transcriptional antagonism between the subfamily of NGFI-B/Nur77 orphan nuclear receptors and glucocorticoid receptor. Mol Endocrinol 2004; 19:885-97. [PMID: 15591535 DOI: 10.1210/me.2004-0333] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (Gc) act through the glucocorticoid receptor (GR) to enhance or repress transcription of glucocorticoid-responsive genes depending on the promoter and cellular context. Repression of proopiomelanocortin (POMC) gene expression by Gc was proposed to use different mechanisms. We described the POMC promoter Nur response element (NurRE) as a target for Gc repression. NGFI-B (Nur77), an orphan nuclear receptor, and two related factors, Nurr1 and NOR1, bind the NurRE as homo- or heterodimers to enhance POMC gene expression in response to CRH. Gc antagonize CRH-stimulated as well as NGFI-B-dependent transcription. We now show that GR antagonizes NurRE-dependent transcription induced by all members of the Nur77 subfamily and that these nuclear receptors can all interact directly with GR. Transcriptional antagonism as well as direct protein-protein interaction between NGFI-B and GR take place primarily via their respective DNA binding domains, although DNA binding itself and the GR homodimerization interface are not involved. In vivo, GR and Nur factors can be coimmunoprecipitated whereas GR is recruited to the POMC promoter upon glucocorticoid action. Thus, our data suggest a mechanism for transrepression between two nuclear receptors, GR and NGFI-B, that is unique, although quite similar to that proposed for transrepression between GR and activator protein 1 (AP-1) or nuclear factor-kappaB (NFkappaB).
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cells, Cultured
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/metabolism
- Down-Regulation
- Glucocorticoids/pharmacology
- Humans
- Immunoprecipitation
- Molecular Sequence Data
- Mutation
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Pro-Opiomelanocortin/genetics
- Promoter Regions, Genetic
- Protein Structure, Tertiary
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Steroid/antagonists & inhibitors
- Receptors, Steroid/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Christine Martens
- Laboratoire de génétique moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, Quebec, Canada H2W 1R7
| | | | | | | | | |
Collapse
|
23
|
Necela BM, Cidlowski JA. A single amino acid change in the first zinc finger of the DNA binding domain of the glucocorticoid receptor regulates differential promoter selectivity. J Biol Chem 2004; 279:39279-88. [PMID: 15220338 DOI: 10.1074/jbc.m405489200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian species are well known to differ in their sensitivity to glucocorticoids, but the molecular basis for this difference remains largely uncharacterized. To address this issue, the transcriptional activity of the mouse and human glucocorticoid receptor (GR) was analyzed on two model glucocorticoid-responsive promoters. Mouse GR (mGR) displayed unique promoter discrimination in response to a range of glucocorticoids, with enhanced activity on a simple glucocorticoid response element (GRE)-based promoter and diminished activity on the complex mouse mammary tumor virus promoter compared with human GR (hGR). Promoter discrimination between mGR and hGR was mapped to a single amino acid change at residue 437 (glycine to valine) of mGR and to sequence differences within individual GREs of the different promoters. Mouse GR displayed higher activation on GREs with a guanine rather than a thymine at the -6 position. Binding studies indicated mGR (mGR437V) displayed a weaker affinity for GREs containing a thymine at the -6 position than a mGR mutant containing a glycine at residue 437 (mGR437G). Despite distinct transcriptional activities, both receptors had similar affinities for response elements that contain a guanine at the -6 position. Our findings support a model by which the presence of a valine residue at position 437 of mGR induces a conformational change that leads to alterations in affinity and/or transcriptional activation in a promoter-dependent context.
Collapse
Affiliation(s)
- Brian M Necela
- Laboratory of Signal Transduction, NIEHS, and Department of Health and Human Services, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
24
|
Uht RM, Webb P, Nguyen P, Price Jr RH, Valentine C, Favre H, Kushner PJ. A conserved lysine in the estrogen receptor DNA binding domain regulates ligand activation profiles at AP-1 sites, possibly by controlling interactions with a modulating repressor. NUCLEAR RECEPTOR 2004; 2:2. [PMID: 15132742 PMCID: PMC446215 DOI: 10.1186/1478-1336-2-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 05/07/2004] [Indexed: 11/25/2022]
Abstract
Background Estrogen receptors alpha and beta (ERα and ERβ) differentially activate genes with AP-1 elements. ERα activates AP-1 targets via activation functions with estrogens (the AF-dependent pathway), whereas ERβ, and a short version of ERα (ERα 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 (ERα.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 ERβ, K170A, or in ERα DBD-LBD leads to estrogen-dependent AP-1 activation and loss of the usually potent anti-estrogen effects. Each of numerous K206 substitutions in ERα, 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 ERβ and ERα DBD-LBD), and prevents ERα 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.
Collapse
Affiliation(s)
- Rosalie M Uht
- Departments of Pathology, & Biochemistry and Molecular Genetics University of Virginia, School of Medicine, MR5 Rm. 3123, 415 Lane Rd., Charlottesville, VA 22908-0904, USA
| | - Paul Webb
- Center for Diabetes and Endocrinology, University of California San Francisco, CA 94143-0540, USA
| | - Phuong Nguyen
- Center for Diabetes and Endocrinology, University of California San Francisco, CA 94143-0540, USA
| | | | - Cathleen Valentine
- Department of Medicine, Room C430, 2200 Post St., University of California San Francisco, San Francisco CA 94115-1640, USA
| | - Helene Favre
- Department of Medicine, Room C430, 2200 Post St., University of California San Francisco, San Francisco CA 94115-1640, USA
| | - Peter J Kushner
- Department of Medicine, Room C430, 2200 Post St., University of California San Francisco, San Francisco CA 94115-1640, USA
| |
Collapse
|
25
|
Baxter JD, Funder JW, Apriletti JW, Webb P. Towards selectively modulating mineralocorticoid receptor function: lessons from other systems. Mol Cell Endocrinol 2004; 217:151-65. [PMID: 15134814 DOI: 10.1016/j.mce.2003.10.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although there is clinical utility in blocking mineralocorticoid receptor (MR) action, the usefulness of available MR antagonists is limited because of cross-reactivity with the androgen and progesterone receptors (spironolactone) or possibly by low affinity for MR (eplerenone). MR binds aldosterone and physiologic glucocorticoids, such as cortisol, which both can act as MR agonists in epithelial tissues. However, in preliminary studies aldosterone and cortisol appear to induce different conformations in non-epithelial tissues; in the cardiomyocyte, cortisol usually acts as an MR antagonist, whereas in vascular smooth muscle cortisol mimics aldosterone actions if it can access MR, just as it does in the kidney. Thus, there are needs for improved MR antagonists with higher selectivity and potency and, if possible, for compounds that lock MR into specific desirable conformations. Efforts are underway to modulate selectively the action of many nuclear receptors, and insights from one nuclear receptor may be applicable to others given the similarities in structure and function. We have used traditional approaches aided by X-ray crystallography to obtain several classes of selective ligands that modulate thyroid receptor (TR) action. We describe the properties of these selective TR modulators here, and discuss the possibility that similar approaches to ligand design may yield MR interacting compounds with improved specificity and, possibly, tissue specificity.
Collapse
MESH Headings
- Aldosterone/metabolism
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/metabolism
- Crystallography, X-Ray
- Epithelium/physiology
- Eplerenone
- Hydrocortisone/administration & dosage
- Hydrocortisone/metabolism
- Mineralocorticoid Receptor Antagonists/chemistry
- Myocytes, Cardiac/metabolism
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/analysis
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/metabolism
- Receptors, Mineralocorticoid/agonists
- Receptors, Mineralocorticoid/chemistry
- Receptors, Mineralocorticoid/metabolism
- Receptors, Thyroid Hormone/antagonists & inhibitors
- Receptors, Thyroid Hormone/chemistry
- Receptors, Thyroid Hormone/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Spironolactone/administration & dosage
- Spironolactone/analogs & derivatives
- Spironolactone/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- John D Baxter
- Diabetes Center and Metabolic Research Unit, University of California-San Francisco, San Francisco, CA 94143-0540, USA.
| | | | | | | |
Collapse
|
26
|
Dobreva G, Dambacher J, Grosschedl R. SUMO modification of a novel MAR-binding protein, SATB2, modulates immunoglobulin mu gene expression. Genes Dev 2004; 17:3048-61. [PMID: 14701874 PMCID: PMC305257 DOI: 10.1101/gad.1153003] [Citation(s) in RCA: 206] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nuclear matrix attachment regions (MARs) are regulatory DNA sequences that are important for higher-order chromatin organization, long-range enhancer function, and extension of chromatin modifications. Here we characterize a novel cell type-specific MAR-binding protein, SATB2, which binds to the MARs of the endogenous immunoglobulin micro locus in pre-B cells and enhances gene expression. We found that SATB2 differs from the closely related thymocyte-specific protein SATB1 by modifications of two lysines with the small ubiquitive related modifier (SUMO), which are augmented specifically by the SUMO E3 ligase PIAS1. Mutations of the SUMO conjugation sites of SATB2 enhance its activation potential and association with endogenous MARs in vivo, whereas N-terminal fusions with SUMO1 or SUMO3 decrease SATB2-mediated gene activation. Sumoylation is also involved in targeting SATB2 to the nuclear periphery, raising the possibility that this reversible modification of a MAR-binding protein may contribute to the modulation of subnuclear DNA localization.
Collapse
Affiliation(s)
- Gergana Dobreva
- Gene Center and Institute of Biochemistry, University of Munich, Munich 81377, Germany
| | | | | |
Collapse
|
27
|
Krieg AJ, Krieg SA, Ahn BS, Shapiro DJ. Interplay between estrogen response element sequence and ligands controls in vivo binding of estrogen receptor to regulated genes. J Biol Chem 2003; 279:5025-34. [PMID: 14617632 DOI: 10.1074/jbc.m307076200] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To examine the role of the estrogen response element (ERE) sequence in binding of liganded estrogen receptor (ER) to promoters, we analyzed in vivo interaction of liganded ER with the imperfect ERE in the pS2 gene and the composite estrogen-responsive unit (ERU) in the proteinase inhibitor 9 (PI-9) gene. In transient transfections of ER-positive HepG2-ER7 cells, PI-9 was strongly induced by estrogen, moxestrol (MOX), and 4-hydroxytamoxifen (OHT). PI-9 was not induced by raloxifene or ICI 182,780. Quantitative reverse transcriptase-PCR showed that moxestrol strongly induced cellular PI-9 and pS2 mRNAs, whereas OHT moderately induced PI-9 mRNA and weakly induced pS2 mRNA. Chromatin immunoprecipitation experiments demonstrated strong and similar association of 17beta-estradiol-hERalpha and MOX-hERalpha with the PI-9 ERU and with the pS2 ERE. Binding of MOX-hERalpha to the PI-9 ERU and the pS2 ERE was rapid and continuous. Although MOX-hERalpha bound strongly to the PI-9 ERU and less well to the pS2 ERE in chromatin immunoprecipitation, gel shift assays showed that estrogen-hERalpha binds with higher affinity to the deproteinized pS2 ERE than to the PI-9 ERU. Across a broad range of OHT concentrations, OHT-hERalpha associated strongly with the pS2 ERE and weakly with the PI-9 ERU. ICI-hERalpha bound poorly to the PI-9 ERU and effectively to the pS2 ERE. Raloxifene-hERalpha and MOX-hERalpha exhibited similar binding to the PI-9 ERU and the pS2 ERE. These studies demonstrate that ER ligand and ERE sequence work together to regulate in vivo binding of ER to estrogen-responsive promoters.
Collapse
Affiliation(s)
- Adam J Krieg
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801-3602, USA
| | | | | | | |
Collapse
|
28
|
Stockner T, Sterk H, Kaptein R, Bonvin AMJJ. Molecular dynamics studies of a molecular switch in the glucocorticoid receptor. J Mol Biol 2003; 328:325-34. [PMID: 12691744 DOI: 10.1016/s0022-2836(03)00316-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The glucocorticoid receptor (GR) is a hormone dependent nuclear receptor that regulates gene transcription when bound to the glucocorticoid response element (GRE). The GRE acts as an allosteric effector, inducing a structural change in the GR DNA-binding domain (GR DBD) upon binding, thereby switching the GR to an active conformation. A similar conformational change can be induced by two single point mutations: Ser459Ala and Pro493Arg. Structural and dynamical aspects of the conformational switch have been investigated by molecular dynamics simulations. Our results indicate that these two mutants, which share a similar phenotype, exert their action at a structural level through different mechanisms. In the Arg(493) mutant, the D-loop and the second helix are stabilized in a conformation that preforms the protein-protein dimer interface. In the Ala(459) mutant, the structurally important hydrogen bond between Arg(496) and Ser(459) is missing, which leads to a core reorganization and a reorientation of the second helical region. Although remote, both in sequence and three dimensional structure, these two mutations induce structural changes that are ultimately reflected in similar regions of the GR DBD structure, namely the D-loop and the short second helical region. These correspond to hot area of the GR DBD that are important both for DNA-binding and for the proper formation of the protein-protein interface. The conformational rearrangements in these area are proposed to decrease unfavorable protein-DNA and protein-protein contacts and allow unspecific DNA-binding leading to the squelching phenotype of the mutants. The GR DBD can thus exist in two states, a transcriptionally active and an inactive state. Switching between these states can be accomplished either by GRE binding or by the described mutations.
Collapse
Affiliation(s)
- Thomas Stockner
- Department of NMR Spectroscopy, Karl Franzens Universität Graz, Heinrichstrasse 28, 8010 Graz, Austria
| | | | | | | |
Collapse
|
29
|
DeFranco DB. Functional implications of glucocorticoid receptor trafficking. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2003:91-109. [PMID: 12355731 DOI: 10.1007/978-3-662-04660-9_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- D B DeFranco
- University of Pittsburgh, Department of Biological Sciences, Pittsburgh, PA 15260, USA. dod1+@pitt.edu
| |
Collapse
|
30
|
Huss JM, Kopp RP, Kelly DP. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem 2002; 277:40265-74. [PMID: 12181319 DOI: 10.1074/jbc.m206324200] [Citation(s) in RCA: 406] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The transcriptional coactivator PPARgamma coactivator-1alpha (PGC-1alpha) has been characterized as a broad regulator of cellular energy metabolism. Although PGC-1alpha functions through many transcription factors, the PGC-1alpha partners identified to date are unlikely to account for all of its biologic actions. The orphan nuclear receptor estrogen-related receptor alpha (ERRalpha) was identified in a yeast two-hybrid screen of a cardiac cDNA library as a novel PGC-1alpha-binding protein. ERRalpha was implicated previously in regulating the gene encoding medium-chain acyl-CoA dehydrogenase (MCAD), which catalyzes the initial step in mitochondrial fatty acid oxidation. The cardiac perinatal expression pattern of ERRalpha paralleled that of PGC-1alpha and MCAD. Adenoviral-mediated ERRalpha overexpression in primary neonatal cardiac mycoytes induced endogenous MCAD expression. Furthermore, PGC-1alpha enhanced the transactivation of reporter plasmids containing an estrogen response element or the MCAD gene promoter by ERRalpha and the related isoform ERRgamma. In vitro binding experiments demonstrated that ERRalpha interacts with PGC-1alpha via its activation function-2 homology region. Mutagenesis studies revealed that the LXXLL motif at amino acid position 142-146 of PGC-1alpha (L2), necessary for PGC-1alpha interactions with other nuclear receptors, is not required for the PGC-1alpha.ERRalpha interaction. Rather, ERRalpha binds PGC-1alpha primarily through a Leu-rich motif at amino acids 209-213 (Leu-3) and utilizes additional LXXLL-containing domains as accessory binding sites. Thus, the PGC-1alpha.ERRalpha interaction is distinct from that of other nuclear receptor PGC-1alpha partners, including PPARalpha, hepatocyte nuclear factor-4alpha, and estrogen receptor alpha. These results identify ERRalpha and ERRgamma as novel PGC-1alpha interacting proteins, implicate ERR isoforms in the regulation of mitochondrial energy metabolism, and suggest a potential mechanism whereby PGC-1alpha selectively binds transcription factor partners.
Collapse
Affiliation(s)
- Janice M Huss
- Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
31
|
Lethimonier C, Tujague M, Kern L, Ducouret B. Peptide insertion in the DNA-binding domain of fish glucocorticoid receptor is encoded by an additional exon and confers particular functional properties. Mol Cell Endocrinol 2002; 194:107-16. [PMID: 12242033 DOI: 10.1016/s0303-7207(02)00181-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The trout glucocorticoid receptor (rtGR) contains an additional sequence of nine amino acids located between the two zinc fingers of the DNA-binding domain (DBD) (Endocrinology 136 (1995) 3774). Polymerase chain reaction on trout genomic DNA and sequencing were performed in the DBD region, demonstrating that this peptide is encoded by an additional exon of 27 nucleotides between the two exons encoding the two zinc fingers of other nuclear receptors. This additional sequence in the rtGR confers a better binding affinity of the receptor to a single GRE, as shown by gel shift experiments with GST-DBDrtGR fusion proteins, deleted or not of the nine amino acids (Delta9). This higher affinity is correlated with a higher constitutive transcriptional activity of the receptor on a reporter gene driven by a single GRE, but not with the ligand-induced transcriptional activity. Nevertheless, on a double GRE, the wild type and rtGR-Delta9 are equally active on both constitutive or dexamethasone-induced transcriptional activity. This original DBD structure could have emerged during evolution such as to allow better regulation of glucocorticoid dependent genes in relation to the large spectrum of cortisol physiological functions in fish.
Collapse
Affiliation(s)
- C Lethimonier
- Endocrinologie Moléculaire de la Reproduction, UMR-CNRS 6026 Bât 13, Campus de Beaulieu, Université de Rennes 1, 35042 Rennes cedex, France
| | | | | | | |
Collapse
|
32
|
Keeton EK, Fletcher TM, Baumann CT, Hager GL, Smith CL. Glucocorticoid receptor domain requirements for chromatin remodeling and transcriptional activation of the mouse mammary tumor virus promoter in different nucleoprotein contexts. J Biol Chem 2002; 277:28247-55. [PMID: 12029095 DOI: 10.1074/jbc.m203898200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The glucocorticoid receptor (GR) contains several activation domains, tau1 (AF-1), tau2, and AF-2, which were initially defined using transiently transfected reporter constructs. Using domain mutations in the context of full-length GR, this study defines those domains required for activation of the mouse mammary tumor virus (MMTV) promoter in two distinct nucleoprotein configurations. A transiently transfected MMTV template with a disorganized, accessible chromatin structure was largely dependent on the AF-2 domain for activation. In contrast, activation of an MMTV template in organized, replicated chromatin requires both domains but has a relatively larger dependence on the tau1 domain. Domain requirements for GR-induced chromatin remodeling of the latter template were also investigated. Mutation of the AF-2 helix 12 domain partially inhibits the induction of nuclease hypersensitivity, but the inhibition was relieved in the absence of tau1, suggesting the occurrence of an important interaction between the two domains. Further mutational analysis indicates that GR-induced chromatin remodeling requires the ligand-binding domain in the region of helix 3. Our study shows that the GR activation surfaces required for transcriptional modulation of a target promoter were determined in part by its chromatin structure. Within a particular cellular environment the GR appears to possess a significant degree of versatility in the mechanism by which it activates a target promoter.
Collapse
MESH Headings
- Alanine
- Amino Acid Substitution
- Animals
- Base Sequence
- Binding Sites
- Chromatin/physiology
- Chromatin/ultrastructure
- Cloning, Molecular
- Dexamethasone/pharmacology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Models, Molecular
- Mutagenesis, Site-Directed
- Nuclear Proteins/metabolism
- Nucleoproteins/metabolism
- Oligodeoxyribonucleotides
- Promoter Regions, Genetic
- Protein Structure, Secondary
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Interferon/metabolism
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
- Serine
- Templates, Genetic
- Transfection
- Tumor Cells, Cultured
Collapse
|
33
|
Liu MM, Albanese C, Anderson CM, Hilty K, Webb P, Uht RM, Price RH, Pestell RG, Kushner PJ. Opposing action of estrogen receptors alpha and beta on cyclin D1 gene expression. J Biol Chem 2002; 277:24353-60. [PMID: 11986316 DOI: 10.1074/jbc.m201829200] [Citation(s) in RCA: 332] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
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.
Collapse
Affiliation(s)
- Meng-Min Liu
- Department of Medicine, University of California, San Francisco, California 94112-1640, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Claudel T, Sturm E, Duez H, Torra IP, Sirvent A, Kosykh V, Fruchart JC, Dallongeville J, Hum DW, Kuipers F, Staels B. Bile acid-activated nuclear receptor FXR suppresses apolipoprotein A-I transcription via a negative FXR response element. J Clin Invest 2002; 109:961-71. [PMID: 11927623 PMCID: PMC150929 DOI: 10.1172/jci14505] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Serum levels of HDL are inversely correlated with the risk of coronary heart disease. The anti-atherogenic effect of HDL is partially mediated by its major protein constituent apoA-I. In this study, we identify bile acids that are activators of the nuclear receptor farnesoid X receptor (FXR) as negative regulators of human apoA-I expression. Intrahepatocellular accumulation of bile acids, as seen in patients with progressive familial intrahepatic cholestasis and biliary atresia, was associated with diminished apoA-I serum levels. In human apoA-I transgenic mice, treatment with the FXR agonist taurocholic acid strongly decreased serum concentrations and liver mRNA levels of human apoA-I, which was associated with reduced serum HDL levels. Incubation of human primary hepatocytes and hepatoblastoma HepG2 cells with bile acids resulted in a dose-dependent downregulation of apoA-I expression. Promoter mutation analysis and gel-shift experiments in HepG2 cells demonstrated that bile acid-activated FXR decreases human apoA-I promoter activity by a negative FXR response element mapped to the C site. FXR bound this site and repressed transcription in a manner independent of retinoid X receptor. The nonsteroidal synthetic FXR agonist GW4064 likewise decreased apoA-I mRNA levels and promoter activity in HepG2 cells.
Collapse
Affiliation(s)
- Thierry Claudel
- Unité de Recherche 545, Institut National de la Santé et de la Recherche Médicale, Département d'Athérosclérose, Institut Pasteur de Lille, and the Faculté de Pharmacie, Université de Lille II, Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Claudel T, Sturm E, Duez H, Torra IP, Sirvent A, Kosykh V, Fruchart JC, Dallongeville J, Hum DW, Kuipers F, Staels B. Bile acid-activated nuclear receptor FXR suppresses apolipoprotein A-I transcription via a negative FXR response element. J Clin Invest 2002. [DOI: 10.1172/jci0214505] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
36
|
Shah OJ, Iniguez-Lluhi JA, Romanelli A, Kimball SR, Jefferson LS. The activated glucocorticoid receptor modulates presumptive autoregulation of ribosomal protein S6 protein kinase, p70 S6K. J Biol Chem 2002; 277:2525-33. [PMID: 11705993 DOI: 10.1074/jbc.m105935200] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Protein metabolism in eukaryotic organisms is defined by a synthesis-degradation equilibrium that is subject to regulation by hormonal and nutritional signals. In mammalian tissues such as skeletal muscle, glucocorticoid hormones specify a catabolic response that influences both protein synthetic and protein degradative pathways. With regard to the former, glucocorticoids attenuate mRNA translation at two levels: translational efficiency, i.e. translation initiation, and translational capacity, i.e. ribosome biogenesis. Glucocorticoids may impair translational capacity through the ribosomal S6 protein kinase (p70 S6K), a recognized glucocorticoid target and an effector of ribosomal protein synthesis. We demonstrate here that the reduction in growth factor-activated p70 S6K activity by glucocorticoids depends upon a functional glucocorticoid receptor (GR) and that the GR is both necessary and sufficient to render p70 S6K subject to glucocorticoid regulation. Furthermore, the DNA binding and transcriptional activation but not repression properties of the GR are indispensable for p70 S6K regulation. Finally, a mutational analysis of the p70 S6K carboxyl terminus indicates that this region confers glucocorticoid sensitivity, and thus glucocorticoids may facilitate autoinhibition of the enzyme ultimately reducing the efficiency with which T389 is phosphorylated.
Collapse
Affiliation(s)
- O Jameel Shah
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033-0850, USA
| | | | | | | | | |
Collapse
|
37
|
Sanyal S, Kim JY, Kim HJ, Takeda J, Lee YK, Moore DD, Choi HS. Differential regulation of the orphan nuclear receptor small heterodimer partner (SHP) gene promoter by orphan nuclear receptor ERR isoforms. J Biol Chem 2002; 277:1739-48. [PMID: 11705994 DOI: 10.1074/jbc.m106140200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The orphan nuclear receptor small heterodimer partner (SHP; NR0B2) interacts with a wide array of nuclear receptors and represses their transcriptional activity. SHP expression is regulated by several other members of the nuclear receptor superfamily, including the orphan receptors SF-1 and LRH-1, and the bile acid receptor FXR. We have found that the SHP promoter is also activated by the estrogen receptor-related receptor gamma (ERRgamma) but not the related ERRalpha and ERRbeta isoforms. SHP and ERRgamma mRNAs are coexpressed in several tissues, including pancreas, kidney, and heart, confirming the potential relevance of this transactivation. ERRgamma transactivation is dependent on only one of five previously characterized DNA-binding sites for SF-1, and this element differs from previously reported ERR response elements. However, treatment with the histone deacetylase inhibitor trichostatin A significantly increased ERRalpha and ERRbeta activity on this element indicating that the lack of activity of ERRalpha and -beta may depend on their association with co-repressor in vivo. Furthermore, using protease sensitivity assays on DNA bound receptors it was demonstrated that DNA sequence of different response elements may cause allosteric modulation of ERR proteins, which in turn may be responsible for the differential activities of these receptors on different response elements. SHP inhibits ERRgamma transactivation and physically interacts with all three members of ERR subfamily, as demonstrated by both yeast two-hybrid and biochemical assays. As with other SHP targets, this interaction is dependent on the AF-2 coactivator-binding site of ERRgamma and the previously described N-terminal receptor interaction domain of SHP. Several recently described SHP mutations associated with moderate obesity in humans block the inhibition of ERRgamma activity. Overall, these results identify a new autoregulatory loop controlling SHP gene expression and significantly extend the potential functional roles of the three ERRs.
Collapse
Affiliation(s)
- Sabyasachi Sanyal
- Department of Biology, Hormone Research Center, Chonnam National University, Kwangju, 500-757, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
38
|
Pearce D, Náray-Fejes-Tóth A, Fejes-Tóth G. Determinants of subnuclear organization of mineralocorticoid receptor characterized through analysis of wild type and mutant receptors. J Biol Chem 2002; 277:1451-6. [PMID: 11677231 DOI: 10.1074/jbc.m105966200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mineralocorticoid receptor (MR) is a hormone-dependent regulator of gene transcription that in the absence of ligand resides both in the cytoplasm and the nucleus. Agonists but not antagonists increase the number of MRs residing in the nucleus and cause aggregation of MRs into distinct clusters. To identify the functional determinants of MR nuclear organization, we examined the localization pattern of wild type MR and a series of mutants in the presence and absence of ligands using fluorescent protein chimeras in living cells. Our data show that although MR DNA binding is not necessary to mediate nuclear localization, it is absolutely required for wild type cluster formation as is an intact N-terminal or C-terminal activation function. In contrast, destabilization of a dimerization motif within the DNA-binding domain has no effect on subnuclear receptor architecture. These data suggest that normal MR cluster formation is dependent on both DNA binding and intact transcriptional activation functions but not on DNA-dependent receptor dimerization. Because dimer mutants bind with high affinity to hormone response element DNA multimers but not to single palindromic DNA sites, we suggest that clusters represent MR aggregates bound to DNA response element multimers in the vicinity of regulated genes.
Collapse
Affiliation(s)
- David Pearce
- Division of Nephrology, Department of Medicine and Cellular & Molecular Pharmacology, University of California, San Francisco, California 94143, USA.
| | | | | |
Collapse
|
39
|
Sachdev S, Bruhn L, Sieber H, Pichler A, Melchior F, Grosschedl R. PIASy, a nuclear matrix-associated SUMO E3 ligase, represses LEF1 activity by sequestration into nuclear bodies. Genes Dev 2001; 15:3088-103. [PMID: 11731474 PMCID: PMC312834 DOI: 10.1101/gad.944801] [Citation(s) in RCA: 429] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2001] [Accepted: 10/17/2001] [Indexed: 11/24/2022]
Abstract
The Wnt-responsive transcription factor LEF1 can activate transcription in association with beta-catenin and repress transcription in association with Groucho. In search of additional regulatory mechanisms of LEF1 function, we identified the protein inhibitor of activated STAT, PIASy, as a novel interaction partner of LEF1. Coexpression of PIASy with LEF1 results in potent repression of LEF1 activity and in covalent modification of LEF1 with SUMO. PIASy markedly stimulates the sumoylation of LEF1 and multiple other proteins in vivo and functions as a SUMO E3 ligase for LEF1 in a reconstituted system in vitro. Moreover, PIASy binds to nuclear matrix-associated DNA sequences and targets LEF1 to nuclear bodies, suggesting that PIASy-mediated subnuclear sequestration accounts for the repression of LEF1 activity.
Collapse
Affiliation(s)
- S Sachdev
- Gene Center and Institute of Biochemistry, University of Munich, 81377 Munich, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Loven MA, Likhite VS, Choi I, Nardulli AM. Estrogen response elements alter coactivator recruitment through allosteric modulation of estrogen receptor beta conformation. J Biol Chem 2001; 276:45282-8. [PMID: 11574541 DOI: 10.1074/jbc.m106211200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Estrogen receptor beta (ERbeta) activates transcription by binding to estrogen response elements (EREs) and coactivator proteins that act as bridging proteins between the receptor and the basal transcription machinery. Although the imperfect vitellogenin B1, pS2, and oxytocin (OT) EREs each differ from the consensus vitellogenin A2 ERE sequence by a single base pair, ERbeta activates transcription of reporter plasmids containing A2, pS2, B1, and OT EREs to different extents. To explain how these differences in transactivation might occur, we have examined the interaction of ERbeta with these EREs and monitored recruitment of the coactivators amplified in breast cancer (AIB1) and transcription intermediary factor 2 (TIF2). Protease sensitivity, antibody interaction, and DNA pull-down assays demonstrated that ERbeta undergoes ERE-dependent changes in conformation resulting in differential recruitment of AIB1 and TIF2 to the DNA-bound receptor. Overexpression of TIF2 or AIB1 in transient transfection assays differentially enhanced ERbeta-mediated transcription of reporter plasmids containing the A2, pS2, B1, and OT EREs. Our studies demonstrate that individual ERE sequences induce changes in conformation of the DNA-bound receptor and influence coactivator recruitment. DNA-induced modulation of receptor conformation may contribute to the ability of ERbeta to differentially activate transcription of genes containing divergent ERE sequences.
Collapse
Affiliation(s)
- M A Loven
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
41
|
Rogatsky I, Zarember KA, Yamamoto KR. Factor recruitment and TIF2/GRIP1 corepressor activity at a collagenase-3 response element that mediates regulation by phorbol esters and hormones. EMBO J 2001; 20:6071-83. [PMID: 11689447 PMCID: PMC125702 DOI: 10.1093/emboj/20.21.6071] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2001] [Revised: 09/09/2001] [Accepted: 09/09/2001] [Indexed: 11/13/2022] Open
Abstract
To investigate determinants of specific transcriptional regulation, we measured factor occupancy and function at a response element, col3A, associated with the collagenase-3 gene in human U2OS osteosarcoma cells; col3A confers activation by phorbol esters, and repression by glucocorticoid and thyroid hormones. The subunit composition and activity of AP-1, which binds col3A, paralleled the intracellular level of cFos, which is modulated by phorbol esters and glucocorticoids. In contrast, a similar AP-1 site at the collagenase-1 gene, not inducible in U2OS cells, was not bound by AP-1. The glucocorticoid receptor (GR) associated with col3A through protein-protein interactions with AP-1, regardless of AP-1 subunit composition, and repressed transcription. TIF2/GRIP1, reportedly a coactivator for GR and the thyroid hormone receptor (TR), was recruited to col3A and potentiated GR-mediated repression in the presence of a GR agonist but not antagonist. GRIP1 mutants deficient in GR binding and coactivator functions were also defective for corepression, and a GRIP1 fragment containing the GR-interacting region functioned as a dominant-negative for repression. In contrast, repression by TR was unaffected by GRIP1. Thus, the composition of regulatory complexes, and the biological activities of the bound factors, are dynamic and dependent on cell and response element contexts. Cofactors such as GRIP1 probably contain distinct surfaces for activation and repression that function in a context-dependent manner.
Collapse
Affiliation(s)
| | - Kol A. Zarember
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 513 Parnassus Avenue, HSW1201, San Francisco, CA 94143-0450, USA
Present address: Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA Corresponding author e-mail:
| | - Keith R. Yamamoto
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 513 Parnassus Avenue, HSW1201, San Francisco, CA 94143-0450, USA
Present address: Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA Corresponding author e-mail:
| |
Collapse
|
42
|
Loven MA, Wood JR, Nardulli AM. Interaction of estrogen receptors alpha and beta with estrogen response elements. Mol Cell Endocrinol 2001; 181:151-63. [PMID: 11476949 DOI: 10.1016/s0303-7207(01)00491-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
To understand how estrogen-responsive genes are regulated, we compared the abilities of estrogen receptors (ERs) alpha and beta to bind to and activate transcription through the consensus vitellogenin A2 ERE and the imperfect pS2, vitellogenin B1, and oxytocin (OT) EREs. Transient transfection experiments demonstrated that ERalpha and ERbeta induced the highest levels of transcription with the A2 ERE, intermediate levels of transcription with the OT ERE, and low levels of transcription with the pS2 and B1 EREs. ERalpha and ERbeta had higher affinities for the A2 ERE than for any of the three imperfect EREs but similar affinities for the pS2, B1, and OT EREs in gel mobility shift assays. ERalpha had a higher affinity and was a more potent activator of transcription than ERbeta. Interestingly, protease sensitivity assays demonstrated that A2, pS2, B1, and OT EREs induced distinct changes in ERalpha and ERbeta conformation thereby providing different functional surfaces for interaction with regulatory proteins involved in control of estrogen-responsive genes.
Collapse
Affiliation(s)
- M A Loven
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 524 Burrill Hall, 407 South Goodwin Ave., Urbana, IL 61801, USA
| | | | | |
Collapse
|
43
|
Wood JR, Likhite VS, Loven MA, Nardulli AM. Allosteric modulation of estrogen receptor conformation by different estrogen response elements. Mol Endocrinol 2001; 15:1114-26. [PMID: 11435612 DOI: 10.1210/mend.15.7.0671] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Estrogen-regulated gene expression is dependent on interaction of the estrogen receptor (ER) with the estrogen response element (ERE). We assessed the ability of the ER to activate transcription of reporter plasmids containing either the consensus vitellogenin A2 ERE or the imperfect pS2, vitellogenin B1, or oxytocin (OT) ERE. The A2 ERE was the most potent activator of transcription. The OT ERE was significantly more effective in activating transcription than either the pS2 or B1 ERE. In deoxyribonuclease I (DNase I) footprinting experiments, MCF-7 proteins protected A2 and OT EREs more effectively than the pS2 and B1 EREs. Limited protease digestion of the A2, pS2, B1, or OT ERE-bound receptor with V8 protease or proteinase K produced distinct cleavage products demonstrating that individual ERE sequences induce specific changes in ER conformation. Receptor interaction domains of glucocorticoid receptor interacting protein 1 and steroid receptor coactivator 1 bound effectively to the A2, pS2, B1, and OT ERE-bound receptor and significantly stabilized the receptor-DNA interaction. Similar levels of the full-length p160 protein amplified in breast cancer 1 were recruited from HeLa nuclear extracts by the A2, pS2, B1, and OT ERE-bound receptors. In contrast, significantly less transcriptional intermediary factor 2 was recruited by the B1 ERE-bound receptor than by the A2 ERE-bound receptor. These studies suggest that allosteric modulation of ER conformation by individual ERE sequences influences the recruitment of specific coactivator proteins and leads to differential expression of genes containing divergent ERE sequences.
Collapse
Affiliation(s)
- J R Wood
- Department of Molecular and Integrative Physiology, University of Illinois Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- M L Privalsky
- Section of Microbiology, Division of Biological Sciences, One Shield Avenue, University of California at Davis, Davis, CA 95616, USA
| |
Collapse
|
45
|
Doppler W, Windegger M, Soratroi C, Tomasi J, Lechner J, Rusconi S, Cato AC, Almlöf T, Liden J, Okret S, Gustafsson JA, Richard-Foy H, Starr DB, Klocker H, Edwards D, Geymayer S. Expression level-dependent contribution of glucocorticoid receptor domains for functional interaction with STAT5. Mol Cell Biol 2001; 21:3266-79. [PMID: 11287629 PMCID: PMC86973 DOI: 10.1128/mcb.21.9.3266-3279.2001] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The action of the glucocorticoid receptor (GR) on beta-casein gene transcription serves as a well-studied example of a case where the action of the GR is dependent on the activity of another transcription factor, STAT5. We have investigated the domain-requirement of the GR for this synergistic response in transfection experiments employing GR mutants and CV-1 or COS-7 cells. The results were influenced by the expression levels of the GR constructs. At low expression, STAT5-dependent transactivation by mutants of the GR DNA binding domain or N-terminal transactivation domain was impaired and the antiglucocorticoid RU486 exhibited a weak agonistic activity. When the N-terminal region of the GR was exchanged with the respective domain of the progesterone receptor, STAT5-dependent transactivation was reduced at low and high expression levels. Only at high expression levels did the GR exhibit the properties of a coactivator and enhanced STAT5 activity in the absence of a functional DNA binding domain and of GR binding sites in the proximal region of the beta-casein gene promoter. Furthermore, at high GR expression levels RU486 was nearly as efficient as dexamethasone in activating transcription via the STAT5 dependent beta-casein gene promoter. The results reconcile the controversial issue regarding the DNA binding-independent action of the GR together with STAT5 and provide evidence that the mode of action of the GR depends not only on the type of the particular promoter at which it acts but also on the concentration of the GR. GR DNA binding function appears to be mandatory for beta-casein gene expression in mammary epithelial cells, since the promoter function is completely dependent on the integrity of GR binding sites in the promoter.
Collapse
Affiliation(s)
- W Doppler
- Institut für Medizinische Chemie und Biochemie, Universität Innsbruck, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Cross-talk between different transcription factors, notably between the glucocorticoid receptor and AP-1, has been discovered more than 10 years ago: a bona fide transcription factor, without apparent need for its own direct DNA contact, influences the activity of another transcription factor. Recent experiments have added interesting aspects: in addition to major insights into the mechanism of cross-talk, it is now clear that the cross-talk ability of glucocorticoid receptor is essential for mouse development, while the activation of target promoters carrying a glucocorticoid response element (GRE), is surprisingly, dispensable for survival under animal house conditions. Interestingly, the cross-talk function is responsible for almost all regulatory actions of cortisol in the immune system. It is possible that the two functions of the glucocorticoid receptor can be activated separately by specific ligands. Future goals will be to define whether adverse effects of long-term corticosteroid treatment, e.g. osteoporosis, joint necroses, metabolic effects, can be ascribed to GRE-target gene activation and thus be dissociated from the desirable actions in the treatment e.g. of autoimmune disease.
Collapse
Affiliation(s)
- P Herrlich
- Forschungszentrum Karlsruhe, Institute of Toxicology and Genetics, PO Box 3640, D-76021 Karlsruhe, Germany
| |
Collapse
|
47
|
Nissen RM, Yamamoto KR. The glucocorticoid receptor inhibits NFkappaB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev 2000; 14:2314-29. [PMID: 10995388 PMCID: PMC316928 DOI: 10.1101/gad.827900] [Citation(s) in RCA: 401] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Glucocorticoids repress NFkappaB-mediated activation of proinflammatory genes such as interleukin-8 (IL-8) and ICAM-1. Our experiments suggest that the glucocorticoid receptor (GR) confers this effect by associating through protein-protein interactions with NFkappaB bound at each of these genes. That is, we show that the GR zinc binding region (ZBR), which includes the DNA binding and dimerization functions of the receptor, binds directly to the dimerization domain of the RelA subunit of NFkappaB in vitro and that the ZBR is sufficient to associate with RelA bound at NFkappaB response elements in vivo. Moreover, we demonstrate in vivo and in vitro that GR does not disrupt DNA binding by NFkappaB. In transient transfections, we found that the GR ligand binding domain is essential for repression of NFkappaB but not for association with it and that GR can repress an NFkappaB derivative bearing a heterologous activation domain. We used chromatin immunoprecipitation assays in untransfected A549 cells to infer the mechanism by which the tethered GR represses NFkappaB-activated transcription. As expected, we found that the inflammatory signal TNFalpha stimulated preinitiation complex (PIC) assembly at the IL-8 and ICAM-1 promoters and that the largest subunit of RNA polymerase II (pol II) in those complexes became phosphorylated at serines 2 and 5 in its carboxy-terminal domain (CTD) heptapeptide repeats (YSPTSPS); these modifications are required for transcription initiation. Remarkably, GR did not inhibit PIC assembly under repressing conditions, but rather interfered with phosphorylation of serine 2 of the pol II CTD.
Collapse
Affiliation(s)
- R M Nissen
- Departments of Cellular and Molecular Pharmacology, and Biochemistry and Biophysics, PIBS Biochemistry and Molecular Biology Program, University of California, San Francisco, San Francisco, California 94143-0450, USA
| | | |
Collapse
|
48
|
van Tilborg MA, Lefstin JA, Kruiskamp M, Teuben J, Boelens R, Yamamoto KR, Kaptein R. Mutations in the glucocorticoid receptor DNA-binding domain mimic an allosteric effect of DNA. J Mol Biol 2000; 301:947-58. [PMID: 10966797 DOI: 10.1006/jmbi.2000.4001] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two previously isolated mutations in the glucocorticoid receptor DNA-binding domain (DBD), S459A and P493R, have been postulated to mimic DNA-induced conformational changes in the glucocorticoid receptor DBD, thereby constitutively triggering an allosteric mechanism in which binding of specific DNA normally induces the exposure of otherwise silent glucocorticoid receptor transcriptional activation surfaces. Here we report the three-dimensional structure of the free S459A and P493R mutant DBDs as determined by NMR spectroscopy. The free S459A and P493R structures both display the conformational changes in the DBD dimerization interface that are characteristic of the DNA-bound wild-type DBD, confirming that these mutations mimic an allosteric effect of DNA. A transition between two packing arrangements of the DBD hydrophobic core provides a mechanism for long-range transmission of conformational changes, induced either by the mutations or by DNA binding, to protein-protein contact surfaces.
Collapse
Affiliation(s)
- M A van Tilborg
- Bijvoet Center for Biomolecular Research, Padualaan 8, Utrecht, NL3584CH, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
49
|
Iñiguez-Lluhí JA, Pearce D. A common motif within the negative regulatory regions of multiple factors inhibits their transcriptional synergy. Mol Cell Biol 2000; 20:6040-50. [PMID: 10913186 PMCID: PMC86080 DOI: 10.1128/mcb.20.16.6040-6050.2000] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DNA regulatory elements frequently harbor multiple recognition sites for several transcriptional activators. The response mounted from such compound response elements is often more pronounced than the simple sum of effects observed at single binding sites. The determinants of such transcriptional synergy and its control, however, are poorly understood. Through a genetic approach, we have uncovered a novel protein motif that limits the transcriptional synergy of multiple DNA-binding regulators. Disruption of these conserved synergy control motifs (SC motifs) selectively increases activity at compound, but not single, response elements. Although isolated SC motifs do not regulate transcription when tethered to DNA, their transfer to an activator lacking them is sufficient to impose limits on synergy. Mechanistic analysis of the two SC motifs found in the glucocorticoid receptor N-terminal region reveals that they function irrespective of the arrangement of the receptor binding sites or their distance from the transcription start site. Proper function, however, requires the receptor's ligand-binding domain and an engaged dimer interface. Notably, the motifs are not functional in yeast and do not alter the effect of p160 coactivators, suggesting that they require other nonconserved components to operate. Many activators across multiple classes harbor seemingly unrelated negative regulatory regions. The presence of SC motifs within them, however, suggests a common function and identifies SC motifs as critical elements of a general mechanism to modulate higher-order interactions among transcriptional regulators.
Collapse
Affiliation(s)
- J A Iñiguez-Lluhí
- Department of Pharmacology, The University of Michigan Medical School, Ann Arbor, Michigan 48109-0632, USA.
| | | |
Collapse
|
50
|
Abstract
The determination of several structures of nuclear receptor ligand binding domains (LBD) has led to new insights into the mechanism of action of this very important class of receptors. This review describes and compares the different LBD structures and their relationship to the function of the nuclear receptors. The role of the ligand in the LBD structures and the implications of ligand structure on receptor activity are also discussed. Structural information regarding interactions between the LBD and coactivator proteins and the potential role of these interactions in ligand agonism and antagonism is reviewed. Different pathways for nuclear receptor signaling and the use of new ligands to investigate these pathways are also described.
Collapse
Affiliation(s)
- R V Weatherman
- Department of Pharmaceutical Chemistry, University of California, San Francisco 94143-0446, USA
| | | | | |
Collapse
|