1151
|
Edinger RS, Mambo E, Evans MI. Estrogen-dependent transcriptional activation and vitellogenin gene memory. Mol Endocrinol 1997; 11:1985-93. [PMID: 9415402 DOI: 10.1210/mend.11.13.0037] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The concept of hepatic memory suggests that a gene responds more rapidly to a second exposure of an inducer than it does during the initial activation. To determine how soon estrogen-dependent DNA/protein interactions occur during the primary response, in vivo dimethylsulfate footprinting was carried out using genomic DNA amplified by ligation-mediated PCR. When estrogen was added to disrupted cells from a hormone-naive liver, changes within and around the estrogen response elements occurred within seconds, indicating a direct and rapid effect on this estrogen-responsive promoter that had never before been activated. Because this effect was so rapid relative to the delayed onset of mRNA accumulation during the primary response, run-on transcription assays were used to determine the transcription profiles for four of the yolk protein genes during the primary and secondary responses to estrogen. As with the accumulation of mRNA, the onset of transcription was delayed for all of these genes after a primary exposure to estrogen. Interestingly, after the secondary exposure to estrogen, the vitellogenin I, vitellogenin II, and very low density apolipoprotein II genes displayed a more rapid onset of transcription, whereas the primary and secondary profiles of apolipoprotein B transcription in response to estrogen were identical. Because the apoB gene is constitutively expressed in the absence of estrogen, and the vitellogenins are quiescent before the administration of the hormone, hepatic memory most likely represents a relatively stable event in the transition to an active state of a gene that is committed for tissue-specific expression.
Collapse
Affiliation(s)
- R S Edinger
- Department of Biochemistry, School of Medicine, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown 26506-9142, USA
| | | | | |
Collapse
|
1152
|
Sathya G, Li W, Klinge CM, Anolik JH, Hilf R, Bambara RA. Effects of multiple estrogen responsive elements, their spacing, and location on estrogen response of reporter genes. Mol Endocrinol 1997; 11:1994-2003. [PMID: 9415403 DOI: 10.1210/mend.11.13.0039] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Most highly estrogen-responsive genes possess multiple estrogen-responsive elements (EREs) that act synergistically to activate expression. Synergism between EREs appears to depend on structural features of the EREs and the promoter. To examine the activation process, we cloned single or multiple tandem copies of the consensus ERE into reporter plasmids. These plasmids contained either a chloramphenicol acetyl transferase reporter gene driven by a minimal promoter or a luciferase reporter gene driven by the Simian virus 40 (SV40) promoter. Using MCF-7 human breast cancer cells, we demonstrate that synergism among EREs depends on the number of EREs, their spacing, and the distance of the EREs from the promoter. The induction capacity of EREs falls off slowly with distance from the promoter. Remarkably, multiple EREs can induce effectively and synergize even when they are located more than 2000 nucleotides from the promoter. For EREs located immediately upstream of the promoter, both the distance separating the EREs and the distance to the promoter have to be optimal for synergy. Altering either distance changes the response from synergistic to additive. For distant EREs, presumed to interact by a looping mechanism at the promoter, the length of DNA between the EREs and the promoter is not critical. Synergy among closely spaced EREs that are far from the promoter only requires an optimal distance separating the ERE centers of symmetry. Interestingly, very widely separated EREs can also synergize, presumably also because of their ability to interact by looping. The estrogen response from single or multiple tandem copies of ERE half-palindromes near the SV40 promoter was also tested. The negligible induction capacity of a single half-site was not significantly increased in multiple sites. The biological role of half-EREs is not apparent in the system employed here.
Collapse
Affiliation(s)
- G Sathya
- Department of Biochemistry and Biophysics and the Cancer Center, University of Rochester, School of Medicine and Dentistry, New York 14642, USA
| | | | | | | | | | | |
Collapse
|
1153
|
Cleutjens CB, Steketee K, van Eekelen CC, van der Korput JA, Brinkmann AO, Trapman J. Both androgen receptor and glucocorticoid receptor are able to induce prostate-specific antigen expression, but differ in their growth-stimulating properties of LNCaP cells. Endocrinology 1997; 138:5293-300. [PMID: 9389513 DOI: 10.1210/endo.138.12.5564] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Androgen receptor-positive LNCaP cells were stably transfected with a rat glucocorticoid receptor (GR) expression plasmid. Ligand-binding studies in the generated cell lines revealed high-affinity binding of the cognate ligands to their receptors. Transfection experiments with the newly derived cell lines showed that, like androgen receptor, GR can induce activity of a prostate-specific antigen promoter fragment linked to the luciferase gene. Similarly, dexamethasone can stimulate expression of endogenous prostate-specific antigen messenger RNA. Cell proliferation could be induced by R1881. In contrast, dexamethasone treatment of the GR-positive sublines had no stimulatory effect on cell growth. Using the differential display technique, a so far unknown complementary DNA fragment, designated 21.1, specifically induced by androgens and not by glucocorticoids, has been identified. In conclusion, the newly generated cell lines, together with the parental LNCaP cell line, form an attractive system with which to study the mechanism of specificity of steroid hormone regulation of gene expression.
Collapse
Affiliation(s)
- C B Cleutjens
- Department of Pathology, Erasmus University, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
1154
|
Savoldi G, Fenaroli A, Ferrari F, Rigaud G, Albertini A, Di Lorenzo D. The glucocorticoid receptor regulates the binding of C/EPBbeta on the alpha-1-acid glycoprotein promoter in vivo. DNA Cell Biol 1997; 16:1467-76. [PMID: 9428795 DOI: 10.1089/dna.1997.16.1467] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A complex interaction between the Glucocorticoid Receptor (GR), C/EBPbeta, and other transcription factors activate the Alpha-1 Acid Glycoprotein (AGP) promoter in HTC(JZ-1) rat hepatoma culture cells. This effect is mediated by the so-called Steroid Responsive Unit (SRU) of the AGP promoter that contains several binding sites for C/EBP transcription factors, some of which overlap with the Glucocorticoid Responsive Element (GRE). Our in vivo footprinting experiments revealed that the GRE- and the C/EBP-binding sites were already occupied glucocorticoid dependently in HTC(JZ-1) cells 10 min after dexamethasone administration (10(-6) M). Furthermore, local changes in the chromatine structure shown by the appearance of DNAse I hypersensitive sites (HS sites) also took place. These changes were probably dependent on a tissue-specific organization of the chromatine at the SRU because they were not detectable in a different glucocorticoid-responsive cell line (PC12) that did not express AGP. Here, we have also shown that withdrawal of dexamethasone or addition of the anti-glucocorticoid RU486 were able to revert the pattern induced by dexamethasone in vivo. The disappearance of the protected region and the hypersensitive sites, typical of the hormone activated promoter, confirmed the necessity of the GR to be bound by the agonist and the inability of the GR-antagonist complex to bind the DNA. By functional assays, we showed that the occupancy of the SRU by these transcriptional proteins in vivo correlated with the activation of the AGP gene transcription. With these results, we have shown that one of the functions of the GR to activate transcription of the AGP gene is to recruit C/EBPbeta and to maintain it bound at its target DNA sequences (SRU). This process was not accomplished by RU486.
Collapse
Affiliation(s)
- G Savoldi
- Laboratory of Hormonology and Toxicology, Civic Hospital of Brescia, Italy
| | | | | | | | | | | |
Collapse
|
1155
|
Li H, Leo C, Schroen DJ, Chen JD. Characterization of receptor interaction and transcriptional repression by the corepressor SMRT. Mol Endocrinol 1997; 11:2025-37. [PMID: 9415406 DOI: 10.1210/mend.11.13.0028] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) and N-CoR (nuclear receptor corepressor) are two related transcriptional corepressors that contain separable domains capable of interacting with unliganded nuclear receptors and repressing basal transcription. To decipher the mechanisms of receptor interaction and transcriptional repression by SMRT/N-CoR, we have characterized protein-protein interacting surfaces between SMRT and nuclear receptors and defined transcriptional repression domains of both SMRT and N-CoR. Deletional analysis reveals two individual nuclear receptor domains necessary for stable association with SMRT and a C-terminal helix essential for corepressor dissociation. Coordinately, two SMRT domains are found to interact independently with the receptors. Functional analysis reveals that SMRT contains two distinct repression domains, and the corresponding regions in N-CoR also repress basal transcription. Both repression domains in SMRT and N-CoR interact weakly with mSin3A, which in turn associates with a histone deacetylase HDAC1 in a mammalian two-hybrid assay. Far-Western analysis demonstrates a direct protein-protein interaction between two N-CoR repression domains with mSin3A. Finally we demonstrate that overexpression of full-length SMRT further represses basal transcription from natural promoters. Together, these results support a role of SMRT/N-CoR in corepression through the utilization of multiple mechanisms for receptor interactions and transcriptional repression.
Collapse
Affiliation(s)
- H Li
- Department of Pharmacology and Molecular Toxicology, University of Massachusetts Medical School, Worcester 01655-0126, USA
| | | | | | | |
Collapse
|
1156
|
Tenbaum S, Baniahmad A. Nuclear receptors: structure, function and involvement in disease. Int J Biochem Cell Biol 1997; 29:1325-41. [PMID: 9570131 DOI: 10.1016/s1357-2725(97)00087-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear hormone receptors are acting as transcription factors in the cell nucleus. They regulate gene expression of hormonal regulated target genes. The role of hormone in the transcriptional process is to modulate and change the nuclear receptor functionality. Receptors contain a DNA binding domain that enables them to bind to hormone response elements of target genes. Nuclear hormone receptors bind to lipophilic hormones produced by the organisms' endocrine system, which links the secretion of hormones directly to regulation of gene expression of responsive tissues. In recent years increasing numbers of naturally occurring mutations of a variety of nuclear hormone receptor genes were identified in patients showing abnormalities in hormonal response. Here, we present an overview of nuclear receptors and their mutant forms which cause human syndromes or are associated with cancer progression. The major scope of this article is to give an overview on the structural-functional relationship and based on that, to understand the effects of naturally occurring receptor mutants on the molecular level. Thereby, functional aberrations of naturally occurring receptors for androgen, glucocorticoids, mineralocorticoid, estrogen, vitamin D3, retinoic acid, and thyroid hormone as well as the orphan receptor DAX1 are discussed.
Collapse
Affiliation(s)
- S Tenbaum
- Genetisches Institut der Justus-Liebig Universität, Giessen, Germany
| | | |
Collapse
|
1157
|
Seol W, Chung M, Moore DD. Novel receptor interaction and repression domains in the orphan receptor SHP. Mol Cell Biol 1997; 17:7126-31. [PMID: 9372944 PMCID: PMC232569 DOI: 10.1128/mcb.17.12.7126] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
SHP (short heterodimer partner) is a novel orphan receptor that lacks a conventional DNA binding domain and interacts with other members of the nuclear hormone receptor superfamily. We have characterized the SHP sequences required for interaction with other superfamily members, and have defined an SHP repressor domain. In the mammalian two-hybrid system, a fusion of full-length SHP to the GAL4 DNA binding domain shows 9-cis-retinoic acid-dependent interaction with a VP16-retinoid X receptor alpha (RXR alpha) fusion. By deletion analysis, sequences required for this RXR interaction map to the central portion of SHP (amino acids 92 to 148). The same region is required for interaction with RXR in vitro and in the yeast two-hybrid system, and results from the yeast system suggest that the same SHP sequences are required for interaction with other members of the nuclear hormone receptor superfamily such as thyroid hormone receptor and retinoic acid receptor. In mammalian cells, a GAL4-SHP fusion protein shows about 10-fold-decreased transcriptional activation relative to GAL4 alone, and fusion of SHP to the C terminus of a GAL4-VP16 fusion to generate a triple chimera also results in a strong decrease in transactivation activity. Sequences required for this repressor function were mapped to the C terminus of SHP. This region is distinct from that required for corepressor interaction by other members of the nuclear hormone receptor superfamily, and SHP did not interact with N-CoR in either the yeast or mammalian two-hybrid system. Together, these results identify novel receptor interaction and repressor domains in SHP and suggest two distinct mechanisms for inhibition of receptor signaling pathways by SHP.
Collapse
Affiliation(s)
- W Seol
- Department of Molecular Biology, Massachusetts General Hospital, Boston 02114, USA
| | | | | |
Collapse
|
1158
|
Ikonen T, Palvimo JJ, Jänne OA. Interaction between the amino- and carboxyl-terminal regions of the rat androgen receptor modulates transcriptional activity and is influenced by nuclear receptor coactivators. J Biol Chem 1997; 272:29821-8. [PMID: 9368054 DOI: 10.1074/jbc.272.47.29821] [Citation(s) in RCA: 278] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Identical N-terminal deletions in the wild-type rat androgen receptor (rAR) and a constitutively active rAR (ARDelta641-902) devoid of the ligand-binding domain (LBD) resulted in dissimilar consequences in transcriptional activation: deletion of residues 149-295 abolished wild-type AR activity, but did not influence that of ARDelta641-902. The activity of the N-terminal transactivation domain is thus controlled by the hormone-occupied LBD, suggesting that the N- and C-terminal regions of rAR communicate. Consistent with this idea, a strong androgen-dependent interaction between the N-terminal region and LBD was demonstrated in a mammalian two-hybrid system using GAL4 and VP16 fusion proteins. This interaction can be direct or indirect. Several nuclear receptor coactivators (CBP, F-SRC-1, SRC-1, and RIP140) that interact with other steroid receptors were tested as potential mediators of the N- and C-terminal interaction of rAR using the mammalian two-hybrid system. CBP or F-SRC-1 not only enhanced AR-mediated transactivation, but also facilitated the androgen-dependent interaction between the N- and C-terminal domains, implying that part of the coactivator-dependent transcriptional activation occurs via this mechanism. In contrast, SRC-1, a coactivator for the progesterone receptor, inhibited both AR-mediated transactivation and interaction between the N and C termini. Recruitment of coregulators may involve AR domains other than the LBD, as F-SRC-1 and CBP enhanced, but SRC-1 repressed, the transcriptional activity of ARDelta641-902. Collectively, interplay between the N-terminal region and LBD of rAR results in the formation of a transactivation complex that includes coregulators and that is mandatory for optimal activation of androgen-induced promoters.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chromosome Mapping
- DNA-Binding Proteins
- Histone Acetyltransferases
- Humans
- Ligands
- Nuclear Receptor Coactivator 1
- Nuclear Receptor Coactivator 3
- Promoter Regions, Genetic
- Rats
- Receptors, Androgen/chemistry
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Glucocorticoid/metabolism
- Receptors, Progesterone/metabolism
- Recombinant Fusion Proteins/metabolism
- Saccharomyces cerevisiae Proteins
- Trans-Activators/metabolism
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- T Ikonen
- Department of Physiology, Institute of Biomedicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | |
Collapse
|
1159
|
Ezura Y, Tournay O, Nifuji A, Noda M. Identification of a novel suppressive vitamin D response sequence in the 5'-flanking region of the murine Id1 gene. J Biol Chem 1997; 272:29865-72. [PMID: 9368060 DOI: 10.1074/jbc.272.47.29865] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vitamin D promotes differentiation of cells either by simply enhancing phenotypic gene expression and/or by suppressing expression of inhibitors of differentiation. Previously, we reported that expression of a gene encoding Id1, a negative type helix-loop-helix transcription factor, was transcriptionally suppressed by 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) (1). To identify the sequence required for the negative regulation by 1, 25(OH)2D3, a 1.5-kilobase 5'-flanking region of murine Id1 gene was examined by transiently transfecting luciferase reporter constructs into ROS17/2.8 osteoblastic cells. The transcriptional activity of this construct was repressed by 10(-8) M 1,25(OH)2D3. Deletion analysis revealed that a 57-base pair (bp) upstream response sequence (URS) (-1146/-1090) was required for the suppression by 1,25(OH)2D3. This sequence conferred negative responsiveness to 1,25(OH)2D3 to a heterologous SV40 promoter. The 57-bp URS contained not only Egr-1 consensus sequence (2) but also four direct repeats of a heptamer sequence (C/A)CAGCCC. Electrophoresis mobility shift assay revealed that the 57-bp URS formed specific nuclear protein-DNA complexes, which were neither competed by previously known positive and negative vitamin D response elements nor supershifted by anti-vitamin D receptor antibody, suggesting the absence of vitamin D receptor in these complexes. These results indicate the involvement of the novel 57-bp sequence in the vitamin D suppression of Id1 gene transcription.
Collapse
Affiliation(s)
- Y Ezura
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 101, Japan
| | | | | | | |
Collapse
|
1160
|
Medvedev A, Chistokhina A, Hirose T, Jetten AM. Genomic structure and chromosomal mapping of the nuclear orphan receptor ROR gamma (RORC) gene. Genomics 1997; 46:93-102. [PMID: 9403063 DOI: 10.1006/geno.1997.4980] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The nuclear orphan receptor subfamily ROR/RZR is part of the steroid and thyroid hormone/retinoid receptor superfamily and consists of three different genes, alpha, beta, and gamma. In this study, we determined the genomic structure of mouse ROR gamma and the chromosomal localization of both mouse ROR gamma and human ROR gamma (HGMW-approved symbol RORC). The genomic structure of the mouse ROR gamma gene was derived from the analysis of P1 vector clones containing large genomic fragments encoding ROR gamma. These results revealed that the mROR gamma gene has a complex structure consisting of 11 exons separated by 10 introns spanning more than 21 kb of genomic DNA. The DNA-binding domain is contained in two exons, 3 and 4, each encoding one zinc-finger. The splice site between exon 3 and exon 4 is identical to that found in RAR and TR3 receptors. ROR gamma is expressed as two mRNAs, 2.3 and 3.0 kb in size, that are derived by the use of alternative polyadenylation signals. We show by fluorescence in situ hybridization that the mouse ROR gamma gene is located on chromosome 3, in a region that corresponds to band 3F2.1-2.2. The human ROR gamma was mapped to chromosome region 1q21. The results demonstrate that the ROR gamma genes are located in chromosomal regions that are syntenic between mouse and human.
Collapse
MESH Headings
- Alternative Splicing/genetics
- Amino Acid Sequence
- Animals
- Base Sequence
- Chromosome Mapping
- Chromosomes, Human, Pair 1/genetics
- Cloning, Molecular
- Exons/genetics
- Genes/genetics
- Humans
- Introns/genetics
- Mice
- Molecular Sequence Data
- Nuclear Receptor Subfamily 1, Group F, Member 3
- RNA Processing, Post-Transcriptional/genetics
- RNA, Messenger/analysis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Retinoic Acid
- Receptors, Thyroid Hormone
- Sequence Analysis, DNA
- Transcription, Genetic/genetics
Collapse
Affiliation(s)
- A Medvedev
- Cell Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | |
Collapse
|
1161
|
Eggert M, Michel J, Schneider S, Bornfleth H, Baniahmad A, Fackelmayer FO, Schmidt S, Renkawitz R. The glucocorticoid receptor is associated with the RNA-binding nuclear matrix protein hnRNP U. J Biol Chem 1997; 272:28471-8. [PMID: 9353307 DOI: 10.1074/jbc.272.45.28471] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that is able to modulate gene activity by binding to its response element, interacting with other transcription factors, and contacting several accessory proteins such as coactivators. Here we show that GRIP120, one of the factors we have identified to interact with the glucocorticoid receptor, is identical to the heterogeneous nuclear ribonucleoprotein U (hnRNP U), a nuclear matrix protein binding to RNA as well as to scaffold attachment regions. GR.hnRNP U complexes were identified by blotting and coimmunoprecipitation. The subnuclear distribution of GR and hnRNP U was characterized by indirect immunofluorescent labeling and confocal laser microscopy demonstrating a colocalization of both proteins. Using a nuclear transport-deficient deletion of hnRNP U, nuclear translocation was seen to be dependent on GR and dexamethasone. Transient transfections were used to identify possible interaction domains. Overexpressed hnRNP U interfered with glucocorticoid induction, and the COOH-terminal domains of both proteins were sufficient in mediating the transcriptional interference. A possible functional role for this GR binding-protein in addition to its binding to the nuclear matrix, to RNA, and to scaffold attachment regions is discussed.
Collapse
Affiliation(s)
- M Eggert
- Genetisches Institut der Justus-Liebig-Universität, Heinrich-Buff-Ring 58-62, D-35392, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1162
|
Milhon J, Lee S, Kohli K, Chen D, Hong H, Stallcup MR. Identification of amino acids in the tau 2-region of the mouse glucocorticoid receptor that contribute to hormone binding and transcriptional activation. Mol Endocrinol 1997; 11:1795-805. [PMID: 9369447 DOI: 10.1210/mend.11.12.0018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The tau 2-region of steroid hormone receptors is a highly conserved region located at the extreme N-terminal end of the hormone-binding domain. A protein fragment encoding tau 2 has been shown to function as an independent transcriptional activation domain; however, because this region is essential for hormone binding, it has been difficult to determine whether the tau 2-region also contributes to the transactivation function of intact steroid receptors. In this study a series of amino acid substitutions were engineered at conserved positions in the tau 2-region of the mouse glucocorticoid receptor (mGR, amino acids 533-562) to map specific amino acid residues that contribute to the hormone-binding function, transcriptional activation, or both. Substitution of alanine or glycine for some amino acids (mutations E546G, P547A, and D555A) reduced or eliminated hormone binding, but the transactivation function of the intact GR and/or the minimum tau 2-fragment was unaffected for each of these mutants. Substitution of alanine for amino acid S561 reduced transactivation activity in the intact GR and the minimum tau 2-fragment but had no effect on hormone binding. The single mutation L550A and the double amino acid substitution L541G+L542G affected both hormone binding and transactivation. The fact that the S561A and L550A substitutions each caused a loss of transactivation activity in the minimum tau 2-fragment and the full-length GR indicated that the tau 2-region does contribute to the overall transactivation function of the full-length GR. Overall, the N-terminal portion of the tau 2-region (mGR 541-547) was primarily involved in hormone binding, whereas the C-terminal portion of the tau 2-region (mGR 548-561) was primarily involved in transactivation.
Collapse
Affiliation(s)
- J Milhon
- Department of Pathology, University of Southern California, Los Angeles 90033, USA
| | | | | | | | | | | |
Collapse
|
1163
|
Stoecklin E, Wissler M, Moriggl R, Groner B. Specific DNA binding of Stat5, but not of glucocorticoid receptor, is required for their functional cooperation in the regulation of gene transcription. Mol Cell Biol 1997; 17:6708-16. [PMID: 9343435 PMCID: PMC232525 DOI: 10.1128/mcb.17.11.6708] [Citation(s) in RCA: 133] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Prolactin and glucocorticoid hormone are signals which regulate the transcription of milk protein genes in mammary epithelial cells. We have investigated the molecular mechanisms by which these hormones cooperate in the induction of transcription. Both hormones activate latent transcription factors in the cytoplasm of mammary epithelial cells. Prolactin exerts its effect through binding to the extracellular domain of the prolactin receptor and through receptor dimerization. This leads to the activation of a protein tyrosine kinase (Jak2), which is noncovalently associated with the cytoplasmic domain of the prolactin receptor. Jak2 phosphorylates the signal transducer and transcription activator (Stat5) which causes its dimerization and nuclear translocation where Stat5 specifically binds to sequence elements in the promoter regions of milk protein genes. In comparison, the glucocorticoid receptor is activated by a lipophilic steroid ligand in the cytoplasm which causes allosteric changes in the molecule, dimerization, and nuclear localization. It has been demonstrated that Stat5 and the glucocorticoid receptor form a molecular complex which cooperates in the induction of transcription of the beta-casein gene. We have defined the DNA sequence requirements for this cooperative mechanism and have delimited the functional domains in Stat5 and the glucocorticoid receptor that are necessary for the functional interaction. We find that the Stat5 response element (Stat5RE) within the beta-casein gene promoter is sufficient to elicit the cooperative action of Stat5 and the glucocorticoid receptor on transcription. Activation of Stat5 through phosphorylation of tyrosine 694 is an absolute prerequisite for transcription. Deletion of the transactivation domain of Stat5 results in a molecule which cannot mediate transactivation by itself but can still cooperate with the glucocorticoid receptor. Mutated variants of the glucocorticoid receptor with a nonfunctional DNA binding domain or a DNA binding domain contributed by the estrogen receptor are still able to cooperate with Stat5 in transcriptional induction. Deletion of the ligand binding domain of the glucocorticoid receptor does not impede cooperation with Stat5, whereas deletion of the AF-1 transactivation domain does prevent cooperation. Our results indicate that the glucocorticoid receptor acts as a ligand-dependent coactivator of Stat5 independently of its DNA binding function.
Collapse
Affiliation(s)
- E Stoecklin
- Institute for Experimental Cancer Research, Tumor Biology Center Freiburg, Germany
| | | | | | | |
Collapse
|
1164
|
Jiang G, Lee U, Sladek FM. Proposed mechanism for the stabilization of nuclear receptor DNA binding via protein dimerization. Mol Cell Biol 1997; 17:6546-54. [PMID: 9343418 PMCID: PMC232508 DOI: 10.1128/mcb.17.11.6546] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hepatocyte nuclear factor 4 (HNF-4) defines a new subgroup of nuclear receptors that exist in solution and bind DNA exclusively as homodimers. We recently showed that the putative ligand binding domain (LBD) of HNF-4 is responsible for dimerization in solution and prevents heterodimerization with other receptors. In this report, the role of the LBD in DNA binding by HNF-4 is further investigated by using electrophoretic mobility shift analysis. A comparison of constructs containing either the DNA binding domain (DBD) alone or the DBD plus the LBD of HNF-4 showed that dimerization via the DBD was sufficient to provide nearly the full DNA binding affinity of the full-length HNF-4. In contrast, dimerization via the DBD was not sufficient to produce a stable protein-DNA complex, whereas dimerization via the LBD increased the half-life of the complex by at least 100-fold. Circular permutation analysis showed that full-length HNF-4 bent DNA by approximately 80 degrees while the DBD bent DNA by only 24 degrees. Nonetheless, analysis of other constructs indicated that the increase in stability afforded by the LBD could be explained only partially by an increased ability to bend DNA. Coimmunoprecipitation studies, on the other hand, showed that dimerization via the LBD produced a protein-protein complex that was much more stable than the corresponding protein-DNA complex. These results led us to propose a model in which dimerization via the LBD stabilizes the receptor on DNA by converting an energetically favorable two-step dissociation event into an energetically unfavorable single-step event. Implications of this one-step model for other nuclear receptors are discussed.
Collapse
Affiliation(s)
- G Jiang
- Environmental Toxicology Graduate Program, University of California, Riverside 92521, USA
| | | | | |
Collapse
|
1165
|
Abstract
Two human glucocorticoid receptor (GR) isoforms, GRalpha and GRbeta, are derived from the same gene by alternative splicing involving exon 9 of the GR locus. The non-ligand binding isoform GRbeta was proposed to act as a transdominant negative inhibitor of GRalpha, thus modulating glucocorticoid responsiveness of target tissues. To study GRbeta in mice we characterized the genomic region around exon 9 of the murine GR gene. Sequence analysis revealed that the presumed exon 9beta contained an open reading frame of 59 amino acids. In contrast, human exon 9beta encoded only 15 amino acids. Using reverse transcriptase polymerase chain reaction the absence of GRbeta mRNA was demonstrated in all adult mouse tissues examined. To exclude the possibility that the polymerase chain reaction conditions employed were not suitable for the amplification of GRbeta mRNA, we synthesized an artificial template corresponding to the presumed GRbeta mRNA spanning exons 7, 8, and 9beta. Various amounts of this template were added to brain cDNA preparations and as little as 25 molecules were detectable under the polymerase chain reaction conditions chosen. Since GRbeta is not conserved across species its physiological significance in humans appears questionable.
Collapse
Affiliation(s)
- C Otto
- Division Molecular Biology of the Cell I, German Cancer Research Centre, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | |
Collapse
|
1166
|
Abstract
Androgens can rescue axotomized motor neurons from cell death. Here we examine a possible mechanism for this trophic action in juvenile Xenopus laevis: regulation of a calcium-binding protein, calbindin, after axotomy. Western analysis revealed that a monoclonal antibody to calbindin D specifically recognizes a single approximately 28 kDa band in X. laevis CNS and rat cerebellum. Retrograde transport of peroxidase combined with immunohistochemistry demonstrated that somata, axons, and synaptic terminals of laryngeal motor neurons in nucleus (N.) IX-X of X. laevis are calbindin-positive. The number of calbindin-positive cells was compared in the intact and axotomized sides of N.IX-X of gonadectomized males that were either hormonally untreated or DHT-treated for 1 month. Although axotomy decreased the number of calbindin-positive cells by 86% in hormonally untreated males, the decrease was only 56% in DHT-treated animals. Compared with hormonally untreated animals, the number of calbindin-labeled cells in N.IX-X of DHT-treated males was increased in both the intact (14%) and axotomized sides (75%). We conclude that axotomy decreases and that DHT enhances calbindin immunoreactivity in N.IX-X. Axotomy-induced decrease in calbindin immunoreactivity precedes cell loss in N.IX-X and may impair the capacity of motor neurons to regulate cytoplasmic calcium. Androgen-mediated maintenance of calbindin expression is thus a candidate cellular mechanism for trophic maintenance of hormone target neurons.
Collapse
|
1167
|
Delaunay F, Khan A, Cintra A, Davani B, Ling ZC, Andersson A, Ostenson CG, Gustafsson J, Efendic S, Okret S. Pancreatic beta cells are important targets for the diabetogenic effects of glucocorticoids. J Clin Invest 1997; 100:2094-8. [PMID: 9329975 PMCID: PMC508401 DOI: 10.1172/jci119743] [Citation(s) in RCA: 226] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abnormalities contributing to the pathogenesis of non-insulin-dependent diabetes mellitus include impaired beta cell function, peripheral insulin resistance, and increased hepatic glucose production. Glucocorticoids are diabetogenic hormones because they decrease glucose uptake and increase hepatic glucose production. In addition, they may directly inhibit insulin release. To evaluate that possible role of glucocorticoids in beta cell function independent of their other effects, transgenic mice with an increased glucocorticoid sensitivity restricted to their beta cells were generated by overexpressing the glucocorticoid receptor (GR) under the control of the insulin promoter. Intravenous glucose tolerance tests showed that the GR transgenic mice had normal fasting and postabsorptive blood glucose levels but exhibited a reduced glucose tolerance compared with their control littermates. Measurement of plasma insulin levels 5 min after intravenous glucose load demonstrated a dramatic decrease in acute insulin response in the GR transgenic mice. These results show that glucocorticoids directly inhibit insulin release in vivo and identify the pancreatic beta cell as an important target for the diabetogenic action of glucocorticoids.
Collapse
Affiliation(s)
- F Delaunay
- Department of Medical Nutrition, Karolinska Institute, Huddinge University Hospital, S-141 86 Huddinge, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1168
|
Minucci S, Horn V, Bhattacharyya N, Russanova V, Ogryzko VV, Gabriele L, Howard BH, Ozato K. A histone deacetylase inhibitor potentiates retinoid receptor action in embryonal carcinoma cells. Proc Natl Acad Sci U S A 1997; 94:11295-300. [PMID: 9326603 PMCID: PMC23446 DOI: 10.1073/pnas.94.21.11295] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/1997] [Accepted: 08/25/1997] [Indexed: 02/05/2023] Open
Abstract
Histone acetylation is thought to have a role in transcription. To gain insight into the role of histone acetylation in retinoid-dependent transcription, we studied the effects of trichostatin A (TSA), a specific inhibitor of histone deacetylase, on P19 embryonal carcinoma cells. We show that coaddition of TSA and retinoic acid (RA) markedly enhances neuronal differentiation in these cells, although TSA alone does not induce differentiation but causes extensive apoptosis. Consistent with the cooperative effect of TSA and RA, coaddition of the two agents synergistically enhanced transcription from stably integrated RA-responsive promoters. The transcriptional synergy by TSA and RA required the RA-responsive element and a functional retinoid X receptor (RXR)/retinoic acid receptor (RAR) heterodimer, both obligatory for RA-dependent transcription. Furthermore, TSA led to promoter activation by an RXR-selective ligand that was otherwise inactive in transcription. In addition, TSA enhanced transcription from a minimum basal promoter, independently of the RA-responsive element. Finally, we show that TSA alone or in combination with RA increases in vivo endonuclease sensitivity within the RA-responsive promoter, suggesting that TSA treatment might alter a local chromatin environment to enhance RXR/RAR heterodimer action. Thus, these results indicate that histone acetylation influences activity of the heterodimer, which is in line with the observed interaction between the RXR/RAR heterodimer and a histone acetylase presented elsewhere.
Collapse
Affiliation(s)
- S Minucci
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
1169
|
Pace P, Taylor J, Suntharalingam S, Coombes RC, Ali S. Human estrogen receptor beta binds DNA in a manner similar to and dimerizes with estrogen receptor alpha. J Biol Chem 1997; 272:25832-8. [PMID: 9325313 DOI: 10.1074/jbc.272.41.25832] [Citation(s) in RCA: 219] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The cloning of a novel estrogen receptor beta (denoted ERbeta) has recently been described (Kuiper, G. G. J. M., Enmark, E., Pelto-Huikko, M., Nilsson, S., and Gustafsson, J-A. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 5925-5930 and Mosselman, S., Polman, J. , and Dijkema, R. (1996) FEBS Lett. 392, 49-53). ERbeta is highly homologous to the "classical" estrogen receptor alpha (here referred to as ERalpha), has been shown to bind estrogens with an affinity similar to that of ERalpha, and activates expression of reporter genes containing estrogen response elements in an estrogen-dependent manner. Here we describe functional studies comparing the DNA binding abilities of human ERalpha and beta in gel shift assays. We show that DNA binding by ERalpha and beta are similarly affected by elevated temperature in the absence of ligand or in the presence of 17beta-estradiol and the partial estrogen agonist 4-hydroxy-tamoxifen. In the absence of ligand, DNA binding by ERalpha and beta is rapidly lost at 37 degrees C, while in the presence of 17beta-estradiol and 4-hydroxy-tamoxifen, the loss in DNA binding at elevated temperature is much more gradual. We show that the loss in DNA binding is not due to degradation of the receptor proteins. However, while the complete antagonist ICI 182, 780 does not "protect" human ERalpha (hERalpha) from loss of DNA binding at elevated temperature in vitro, it does appear to protect human ERbeta (hERbeta), suggestive of differences in the way ICI 182, 780 acts on hERalpha and beta. We further report that ERalpha and beta can dimerize with each other, the DNA binding domain of hERalpha being sufficient for dimerization with hERbeta. Cell and promoter-specific transcription activation by ERalpha has been shown to be dependent on the differential action of the N- and C-terminal transcription activation functions AF-1 and AF-2, respectively. The existence of a second estrogen receptor gene and the dimerization of ERalpha and beta add greater levels of complexity to transcription activation in response to estrogens.
Collapse
Affiliation(s)
- P Pace
- Department of Medical Oncology, Imperial College of Medicine, Charing Cross Campus, St. Dunstan's Road, London W6 8RF, United Kingdom
| | | | | | | | | |
Collapse
|
1170
|
Beaulieu M, Lévesque E, Tchernof A, Beatty BG, Bélanger A, Hum DW. Chromosomal localization, structure, and regulation of the UGT2B17 gene, encoding a C19 steroid metabolizing enzyme. DNA Cell Biol 1997; 16:1143-54. [PMID: 9364925 DOI: 10.1089/dna.1997.16.1143] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
UGT2B17 is a UDP-glucuronosyltransferase enzyme expressed in several extrahepatic steroid target tissues, including the human prostate, where it glucuronidates C19 steroids such as dihydrotestosterone (DHT), androsterone (ADT), and androstane-3alpha, 17beta-diol (3alpha-diol). To determine if UGT2B17 is regulated by physiological effectors of the human prostate, DHT and epidermal growth factor (EGF) were demonstrated to specifically down-regulate the steady-state levels of UGT2B17 transcript and protein in LNCaP cells (Guillemette et al., 1997). These results implicate regulation of UGT2B17 at the level of gene transcription, therefore, a P-1-derived artificial chromosome (PAC) clone of 120 kb containing the entire UGT2B17 gene was isolated. The gene is comprised of six exons spanning approximately 30 kb, and fluorescence in situ hybridization of the UGT2B17 PAC clone to normal human lymphocyte chromosomes, mapped the gene to chromosome 4q13. To determine if the 5'-flanking DNA of the UGT2B17 gene is sufficient to confer gene expression, a 2,942-bp fragment was subcloned into a luciferase reporter plasmid and yielded an activity of 25-fold over background when transfected in LNCaP cells. However, transfection of the construct into HK-293, MCF-7, JEG-3, and HepG2 cells yielded only a moderate activity of two- to five-fold over background. Treatment of transfected LNCaP cells with 10 nM R1881, a nonmetabolizable analog of DHT, and 10 ng/ml EGF decreased the luciferase activity by 60%. This suggests that at least part, if not all, of the inhibitory effect of EGF and DHT on UGT2B17 is at the level of transcription. Progressive 5' deletions of the UGT2B17 5'-flanking region in the luciferase constructs alleviated the inhibition by R1881 and EGF, and revealed several potential responsive elements that may confer the observed regulation of the UGT2B17 gene. This study demonstrates regulation of the UGT2B17 gene by physiological effectors of the human prostate and supports the hypothesis that UGT enzymes are involved in steroid metabolism in extrahepatic tissues.
Collapse
Affiliation(s)
- M Beaulieu
- The Medical Research Council Group in Molecular Endocrinology, CHUL Research Center, Laval University, Québec, Canada
| | | | | | | | | | | |
Collapse
|
1171
|
Abstract
The knowledge accumulated about the biochemistry of the synapsis in the last decades completely changes the notion of brain processing founded exclusively over an electrical mechanism, toward that supported by a complex chemical message exchange occurring both locally, at the synaptic site, as well as at other localities, depending on the solubility of the involved chemical substances in the extracellular compartment. These biochemical transactions support a rich symbolic processing of the information both encoded by the genes and provided by actual data collected from the surrounding environment, by means of either special molecular or cellular receptor systems. In this processing, molecules play the role of symbols and chemical affinity shared by them specifies the syntax for symbol manipulation in order to process and to produce chemical messages. In this context, neurons are conceived as message-exchanging agents. Chemical strings are produced and stored at defined places, and ionic currents are used to speed up message delivery. Synaptic transactions can no longer be assumed to correspond to a simple process of propagating numbers powered by a factor measuring the presynaptic capacity to influence the postsynaptic electrical activity, but they must be modeled by more powerful formal tools supporting both numerical and symbolic calculations. It is proposed here that formal language theory is the adequate mathematical tool to handle such symbolic processing. The purpose of the present review is therefore: (a) to discuss the relevant and recent literature about trophic factors, signal transduction mechanisms, neuromodulators and neurotransmitters in order (b) to point out the common features of these correlated processes; and (c) to show how they may be organized into a formal model supported by the theory of fuzzy formal languages (d) to model the brain as a distributed intelligent problem solver.
Collapse
Affiliation(s)
- A F Rocha
- RANI-Research on Artificial and Natural Intelligence, UNICAMP Brazil, Jundiaí, Brazil.
| |
Collapse
|
1172
|
Humphreys RC, Lydon JP, O'Malley BW, Rosen JM. Use of PRKO mice to study the role of progesterone in mammary gland development. J Mammary Gland Biol Neoplasia 1997; 2:343-54. [PMID: 10935022 DOI: 10.1023/a:1026343212187] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To better understand the distinct physiological roles played by progesterone and estrogen receptors (PR and ER) as well as to study directly PR function in an in vivo context, a novel mutant mouse strain, the PR knockout (PRKO) mouse, was generated carrying a germline loss of function mutation at the PR locus. Mouse mammary gland development has been examined in PRKO mice using reciprocal transplantation experiments to investigate the effects of the stromal and epithelial PRs on ductal and lobuloalveolar development. The absence of PR in transplanted donor epithelium, but not in recipient stroma, prevented normal lobuloalveolar development in response to estrogen and progesterone treatment. Conversely, the presence of PR in the transplanted donor epithelium, but not in the recipient stroma, revealed that PR in the stroma may be necessary for ductal development. Stimulation of ductal development by the PR may, therefore, be mediated by an unknown secondary signaling molecule, possibly a growth factor. The continued stimulation of the stromal PR appears to be dependent on reciprocal signal(s) from the epithelium. Thus, the combination of gene knockout and reciprocal transplantation technologies has provided some new insights into the role of stromal-epithelial interactions and steroid hormones in mammary gland development.
Collapse
Affiliation(s)
- R C Humphreys
- National Institutes of Health, Developmental Biology Section, Bethesda, Maryland 208922, USA
| | | | | | | |
Collapse
|
1173
|
Groshong SD, Owen GI, Grimison B, Schauer IE, Todd MC, Langan TA, Sclafani RA, Lange CA, Horwitz KB. Biphasic regulation of breast cancer cell growth by progesterone: role of the cyclin-dependent kinase inhibitors, p21 and p27(Kip1). Mol Endocrinol 1997; 11:1593-607. [PMID: 9328342 DOI: 10.1210/mend.11.11.0006] [Citation(s) in RCA: 175] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Depending on the tissue, progesterone is classified as a proliferative or a differentiative hormone. To explain this paradox, and to simplify analysis of its effects, we used a breast cancer cell line (T47D-YB) that constitutively expresses the B isoform of progesterone receptors. These cells are resistant to the proliferative effects of epidermal growth factor (EGF). Progesterone treatment accelerates T47D-YB cells through the first mitotic cell cycle, but arrests them in late G1 of the second cycle. This arrest is accompanied by decreased levels of cyclins D1, D3, and E, disappearance of cyclins A and B, and sequential induction of the cyclin-dependent kinase (cdk) inhibitors p21 and p27(Kip1). The retinoblastoma protein is hypophosphorylated and extensively down-regulated. The activity of the cell cycle-dependent protein kinase, cdk2, is regulated biphasically by progesterone: it increases initially, then decreases. This is consistent with the biphasic proliferative increase followed by arrest produced by one pulse of progesterone. A second treatment with progesterone cannot restart proliferation despite adequate levels of transcriptionally competent PR. Instead, a second progesterone dose delays the fall of p21 and enhances the rise of p27(Kip1), thereby intensifying the progesterone resistance in an autoinhibitory loop. However, during the progesterone-induced arrest, the cell cycling machinery is poised to restart. The first dose of progesterone increases the levels of EGF receptors and transiently sensitizes the cells to the proliferative effects of EGF. We conclude that progesterone is neither inherently proliferative nor antiproliferative, but that it is capable of stimulating or inhibiting cell growth depending on whether treatment is transient or continuous. We also suggest that the G1 arrest after progesterone treatment is accompanied by cellular changes that permit other, possibly tissue-specific, factors to influence the final proliferative or differentiative state.
Collapse
Affiliation(s)
- S D Groshong
- University of Colorado Health Sciences Center, The Molecular Biology Program, Denver 80262, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1174
|
Abstract
The results presented underline the fact that the nature and the concentration of the non-esterified fatty acids (NEFAs) liberated from membrane lipids, particularly the essential ones issued from lipid nutrition, clearly belong to a large group of factors (hormones, retinoids, growth factors, cytokines...) which control the shift between cell multiplication and differentiation. NEFAs act on this shift, per se or after being metabolized, by influencing, as second messengers or modulators, the intertwined mechanisms of action of growth factors and steroid hormones. These results may explain the molecular links which exist between endocrinology, oncology and nutrition.
Collapse
Affiliation(s)
- E A Nunez
- Laboratoire de Biochimie Endocrinienne, Faculté de Médecine X. Bichat, Université Denis Diderot, Paris, France
| |
Collapse
|
1175
|
Palmer HJ, Paulson KE. Reactive oxygen species and antioxidants in signal transduction and gene expression. Nutr Rev 1997; 55:353-61. [PMID: 9354079 DOI: 10.1111/j.1753-4887.1997.tb01561.x] [Citation(s) in RCA: 156] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) are produced by cellular metabolic reactions, and have been implicated in the pathogenesis of several diseases, including atherosclerosis, cancer, and Alzheimer's disease. Interestingly, clinical and epidemiologic studies have, in some cases, indicated that antioxidant nutrients may be effective in disease prevention. However, the efficacy of specific antioxidants in disease prevention is often both controversial and inconclusive. In an effort to elucidate the role of ROS and antioxidants in disease development and prevention, the chemistries of ROS and antioxidants have been examined extensively. Recently, molecular and cellular approaches have demonstrated that ROS and antioxidants can directly affect the cellular signaling apparatus and, consequently, the control of gene expression. This new research provides the link between ROS and antioxidant chemistries and the mechanisms of disease processes and prevention. This review illustrates how ROS function as potential intracellular and extracellular signaling molecules and how antioxidants can affect this process.
Collapse
Affiliation(s)
- H J Palmer
- Jean Mayer USDA Human Nutrition Research Center, Tufts University, Boston, MA 02111, USA
| | | |
Collapse
|
1176
|
Lin BC, Hong SH, Krig S, Yoh SM, Privalsky ML. A conformational switch in nuclear hormone receptors is involved in coupling hormone binding to corepressor release. Mol Cell Biol 1997; 17:6131-8. [PMID: 9315673 PMCID: PMC232463 DOI: 10.1128/mcb.17.10.6131] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nuclear hormone receptors are ligand-regulated transcription factors that modulate gene expression in response to small, hydrophobic hormones, such as retinoic acid and thyroid hormone. The thyroid hormone and retinoic acid receptors typically repress transcription in the absence of hormone and activate it in the presence of hormone. Transcriptional repression is mediated, in part, through the ability of these receptors to physically associate with ancillary polypeptides called corepressors. We wished to understand the mechanism by which corepressors are recruited to unliganded nuclear hormone receptors and are released on the binding of hormone. We report here that an alpha-helical domain located at the thyroid hormone receptor C terminus appears to undergo a hormone-induced conformational change required for release of corepressor and that amino acid substitutions that abrogate this conformational change can impair or prevent corepressor release. In contrast, retinoid X receptors appear neither to undergo an equivalent conformational alteration in their C termini nor to release corepressor in response to cognate hormone, consistent with the distinct transcriptional regulatory properties displayed by this class of receptors.
Collapse
Affiliation(s)
- B C Lin
- Division of Biological Sciences, University of California at Davis, 95616, USA
| | | | | | | | | |
Collapse
|
1177
|
Choi HS, Chung M, Tzameli I, Simha D, Lee YK, Seol W, Moore DD. Differential transactivation by two isoforms of the orphan nuclear hormone receptor CAR. J Biol Chem 1997; 272:23565-71. [PMID: 9295294 DOI: 10.1074/jbc.272.38.23565] [Citation(s) in RCA: 227] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have identified a new murine orphan member of the nuclear hormone receptor superfamily, termed mCAR, that is closely related to the previously described human orphan MB67, referred to here as hCAR. Like hCAR, mCAR expression is highest in liver. In addition to the most abundant mCAR1 isoform, the mCAR gene expresses a truncated mCAR2 variant that is missing the C-terminal portion of the ligand binding/dimerization domain. The mCAR gene has 8 introns, and this mCAR2 variant is generated by a splicing event that skips the 8th exon. mCAR1, like hCAR, binds as a heterodimer with the retinoid X receptor to the retinoic acid response element from the promoter of the retinoic acid receptor beta2 isoform. Consistent with its lack of a critical heterodimerization interface, the mCAR2 variant does not bind this site. Both mCAR1 and hCAR are apparently constitutive transcriptional activators. This activity is dependent on the presence of the conserved C-terminal AF-2 transcriptional activation motif. As expected from its inability to bind DNA, the mCAR2 variant neither transactivates by itself nor inhibits transactivation by hCAR or mCAR1.
Collapse
Affiliation(s)
- H S Choi
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
1178
|
Takeyama K, Kitanaka S, Sato T, Kobori M, Yanagisawa J, Kato S. 25-Hydroxyvitamin D3 1alpha-hydroxylase and vitamin D synthesis. Science 1997; 277:1827-30. [PMID: 9295274 DOI: 10.1126/science.277.5333.1827] [Citation(s) in RCA: 350] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Renal 25-hydroxyvitamin D3 1alpha-hydroxylase [1alpha(OH)ase] catalyzes metabolic activation of 25-hydroxyvitamin D3 into 1alpha, 25-dihydroxyvitamin D3 [1alpha,25(OH)2D3], an active form of vitamin D, and is inhibited by 1alpha,25(OH)2D3. 1alpha(OH)ase, which was cloned from the kidney of mice lacking the vitamin D receptor (VDR-/- mice), is a member of the P450 family of enzymes (P450VD1alpha). Expression of 1alpha(OH)ase was suppressed by 1alpha, 25(OH)2D3 in VDR+/+ and VDR+/- mice but not in VDR-/- mice. These results indicate that the negative feedback regulation of active vitamin D synthesis is mediated by 1alpha(OH)ase through liganded VDR.
Collapse
Affiliation(s)
- K Takeyama
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Yayoi, Bunkyo-ku, Tokyo 113, Japan
| | | | | | | | | | | |
Collapse
|
1179
|
Affiliation(s)
- M B Rogers
- Department of Biology, University of South Florida Tampa 33620-5150, USA
| |
Collapse
|
1180
|
Abstract
FTZ-F1 is a member of the orphan nuclear receptors, which belongs to the steroid hormone receptor superfamily, and plays a role in the blastoderm and nervous system development in Drosophila. Recently, several FTZ-F1 family genes have been cloned in several species. SF-1/Ad4BPs have been identified as master regulators controlling steroidogenic P-450 genes in mammals and are considered to be the mammalian homologues of FTZ-F1. Moreover, SF-1/Ad4BP plays a critical role in the sexual differentiation of gonads in mammals. In vertebrates, except for mammals, the functional homologue of SF-1/Ad4BP has not been identified before. Herein, we cloned two chicken cDNAs (OR2.0 and OR2.1), which encode putative FTZ-F1 family receptors, by reverse transcriptase-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends (RACE). OR2.1 consists of 3255 bp, is expressed in the adrenal glands and gonads, and is considered to be the chicken counterpart of mammalian SF-1/Ad4BP. However, OR2.0 consists of 2945 bp, is expressed in the livers and the adrenal glands, and is considered to be the chicken counterpart of mouse LRH-1, which is a member of the FTZ-F1 family in mammals.
Collapse
Affiliation(s)
- T Kudo
- Central Research Institute, Itoham Foods Inc., Ibaraki, Japan
| | | |
Collapse
|
1181
|
Abstract
The question of how sequence-specific transcription factors access their cognate sites in nucleosomally organized DNA is discussed on the basis of genomic footprinting data and chromatin reconstitution experiments. A classification of factors into two categories is proposed: (i) initiator factors which are able to bind their target sequences within regular nucleosomes and initiate events leading to chromatin remodelling and transactivation; (ii) effector factors which are unable to bind regular nucleosomes and depend on initiator factors or on a pre-set nucleosomal structure for accessing their target sequences in chromatin. Studies with the MMTV promoter suggest that the extent and number of protein-DNA contacts determine whether a factor belongs to one or the other category. Initiator factors have only a few DNA contacts clustered on one side of the double helix, whereas effector factors have extensive contacts distributed throughout the whole circumference of the DNA helix. Thus, the nature of DNA recognition confers to sequence-specific factors their specific place in the sequential hierarchy of gene regulatory events.
Collapse
Affiliation(s)
- M Beato
- Institut für Molekularbiologie und Tumorforschung, Philipps Universität, E.-Mannkopff-Strasse 2, 35037 Marburg, Germany.
| | | |
Collapse
|
1182
|
Vivat V, Zechel C, Wurtz JM, Bourguet W, Kagechika H, Umemiya H, Shudo K, Moras D, Gronemeyer H, Chambon P. A mutation mimicking ligand-induced conformational change yields a constitutive RXR that senses allosteric effects in heterodimers. EMBO J 1997; 16:5697-709. [PMID: 9312028 PMCID: PMC1170201 DOI: 10.1093/emboj/16.18.5697] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mutations of a single residue in the retinoid X receptor alpha (RXRalpha) ligand-binding pocket (LBP) generate constitutive, ligand-binding-competent mutants with structural and functional characteristics similar to those of agonist-bound wild-type RXR. Modelling of the mouse RXRalphaF318A LBP suggests that, like agonist binding, the mutation disrupts a cluster of van der Waals interactions that maintains helix H11 in the apo-receptor location, thereby shifting the thermodynamic equilibrium to the holo form. Heterodimerization with some apo-receptors (retinoic acid, thyroid hormone and vitamin D3 receptors) results in 'silencing' of RXRalphaF318A constitutive activity, which, on the other hand, efficiently contributes to synergistic transactivation within NGFI-B-RXR heterodimers. RAR mutants disabled for corepressor binding and/or lacking a functional AF-2 activation domain, do not relieve RXR 'silencing'. Not only RAR agonists, but also the RAR antagonist BMS614 induce conformational changes allowing RXR to exert constitutive (RXRalphaF318A) or agonist-induced (wild-type RXR) activity in heterodimers. Interestingly, the RXRalphaF318A constitutive activity generated within heterodimers in the presence of BMS614 requires the integrity of both RXR and RAR AF-2 domains. These observations suggest that, within RXR-RAR heterodimers, RAR can adopt a structure distinct from that of the active holo-RAR, thus allowing RXR to become transcriptionally responsive to agonists.
Collapse
MESH Headings
- Allosteric Regulation
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Binding Sites
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/metabolism
- Dimerization
- Humans
- Ligands
- Mice
- Models, Structural
- Mutagenesis, Site-Directed
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Protein Conformation/drug effects
- Protein Multimerization
- Receptors, Cytoplasmic and Nuclear
- Receptors, Retinoic Acid/antagonists & inhibitors
- Receptors, Retinoic Acid/chemistry
- Receptors, Retinoic Acid/metabolism
- Receptors, Steroid/chemistry
- Receptors, Steroid/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Retinoid X Receptors
- Retinoids/pharmacology
- Transcription Factors/chemistry
- Transcription Factors/metabolism
- Transfection
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- V Vivat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS/INSERM/ULP/Collège de France, BP 163, 67404 Illkrich Cedex, CU de Strasbourg, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1183
|
Miyoshi Y, Nakao K. A mutant thyroid hormone receptor beta 1 identified in a patient with resistance to thyroid hormone inhibits the activities of not only the wild-type TRs, but also other nuclear receptors. Biochem Biophys Res Commun 1997; 238:39-43. [PMID: 9299447 DOI: 10.1006/bbrc.1997.7184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although mutations of human thyroid hormone receptor beta (hTR beta) have been associated with resistance to thyroid hormone (RTH), the molecular basis by which the mutant TRs cause the various clinical symptoms is unknown. We show here that a mutant TR beta [corrected] identified in a patient with RTH inhibited the transcriptional activities of, not only the wild-type TR beta, but also other nuclear receptors including retinoid X receptor alpha (RXR alpha), vitamin D3 receptor (VDR) and retinoic acid receptor (RAR alpha). We provide evidence that these inhibitions by the mutant TR beta [corrected] occur by different mechanisms. Namely, the mutant TR beta interferes with VDR and RAR alpha by competition for binding to the corresponding response elements, but the pathway through RXR alpha is mainly inhibited by squelching of RXR alpha in solution. These findings suggest that in patients with RTH, not only the T3 responsive genes but also other responsive genes are inhibited by the mutant TRs, which might explain the variety of clinical symptoms in RTH.
Collapse
Affiliation(s)
- Y Miyoshi
- Department of Medicine and Clinical Science, Kyoto University School of Medicine, Japan.
| | | |
Collapse
|
1184
|
Abstract
Brassinosteroids are a class of growth-promoting regulators that play a key role throughout plant development. Despite their importance, nothing is known of the mechanism of action of these steroid hormones. We describe the identification of 18 Arabidopsis dwarf mutants that are unable to respond to exogenously added brassinosteroid, a phenotype that might be expected for brassinosteroid signaling mutants. All 18 mutations define alleles of a single previously described gene, BRI1. We cloned BRI1 and examined its expression pattern. It encodes a ubiquitously expressed putative receptor kinase. The extracellular domain contains 25 tandem leucine-rich repeats that resemble repeats found in animal hormone receptors, plant disease resistance genes, and genes involved in unknown signaling pathways controlling plant development.
Collapse
Affiliation(s)
- J Li
- Plant Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | |
Collapse
|
1185
|
Gong W, Chávez S, Beato M. Point mutation in the ligand-binding domain of the progesterone receptor generates a transdominant negative phenotype. Mol Endocrinol 1997; 11:1476-85. [PMID: 9280063 DOI: 10.1210/mend.11.10.9991] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A short conserved alpha-helix in the carboxyl-terminal activation function of the ligand-binding domain of steroid hormone receptors, called AF2, is important for ligand-dependent transactivation of inducible genes. We have generated two AF2 mutants of the B isoform of human progesterone receptor (PRB): a point mutant, PRBE911A, and a short deletion, PRB delta907-913R. The two mutants are expressed at levels comparable to the wild type receptor in transfected cells. The PRBE911A mutant showed similar hormone- and DNA- binding affinities as the wild type receptor, whereas the PRB delta907-913R mutant was defective in hormone and DNA binding. Both mutants were inactive when transiently transfected in CV-1 cells, which do not express endogenous PR. However, the point mutant, but not the deletion mutant, inhibited transactivation by cotransfected wild type PRB in a hormone-dependent fashion. The activity of endogenous PR in T47D cells or of endogenous glucocorticoid receptor in HeLa cells was also inhibited by the PRBE911A, but not by the deletion mutant. The point mutant was less active when introduced into an N-terminal truncated form of PR, where it gave rise to proteins that formed homodimers with poor affinity for DNA, but were able to form heterodimers with PRB. The negative dominant phenotype of the PRBE911A mutant likely originates from competition with wild type receptors for binding to DNA and will be useful for mechanistic studies of receptor function.
Collapse
Affiliation(s)
- W Gong
- Institut für Molekularbiologie und Tumorforschung, Philipps Universität, Marburg, Germany
| | | | | |
Collapse
|
1186
|
Heckert LL, Wilson EM, Nilson JH. Transcriptional repression of the alpha-subunit gene by androgen receptor occurs independently of DNA binding but requires the DNA-binding and ligand-binding domains of the receptor. Mol Endocrinol 1997; 11:1497-506. [PMID: 9280065 PMCID: PMC1502414 DOI: 10.1210/mend.11.10.9996] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The pituitary glycoprotein hormones LH and FSH regulate the reproductive cycle and are sensitive to feedback by gonadal steroids. The common alpha-subunit shared by these hormones is transcriptionally repressed by androgen receptor (AR) in the presence of its ligand dihydrotestosterone. This identifies at least one mechanism that contributes to AR-dependent suppression of gonadotropin synthesis. Repression of alpha-subunit transcription by AR requires only the sequences within the first 480 bp of the promoter. While this region contains a high-affinity binding site for AR, this element does not mediate the suppressive effects of androgens. Instead, two other elements within the promoter-regulatory region (alpha-basal element and cAMP-regulatory element), which are important for expression of the alpha-subunit gene in gonadotropes, mediate the effects of AR. This suggests that AR inhibits activity of the alpha-subunit promoter by interfering with the transcriptional properties of the proteins that bind to alpha-basal element and the cAMP-regulatory elements. Furthermore, transfection analysis of various mutant ARs identified both the DNA-binding and ligand-binding domains of the receptor as critical for repression. Comparisons with the MMTV promoter revealed distinct structural requirements that underlie the transactivation and transrepression properties of AR.
Collapse
Affiliation(s)
- L L Heckert
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City 66160, USA
| | | | | |
Collapse
|
1187
|
Spiegelman VS, Budunova IV, Carbajal S, Slaga TJ. Resistance of transformed mouse keratinocytes to growth inhibition by glucocorticoids. Mol Carcinog 1997. [DOI: 10.1002/(sici)1098-2744(199709)20:1<99::aid-mc11>3.0.co;2-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
1188
|
Wissink S, van Heerde EC, Schmitz ML, Kalkhoven E, van der Burg B, Baeuerle PA, van der Saag PT. Distinct domains of the RelA NF-kappaB subunit are required for negative cross-talk and direct interaction with the glucocorticoid receptor. J Biol Chem 1997; 272:22278-84. [PMID: 9268377 DOI: 10.1074/jbc.272.35.22278] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The RelA subunit of NF-kappaB and the glucocorticoid receptor mutually repress each others transcriptional activity, thus providing a mechanism for immunosuppression. Deletion analysis of the glucocorticoid receptor has shown that the DNA binding domain and the ligand binding domain are essential components for repression. Here, we show by deletions and point mutations that both the Rel homology domain and the transactivation domains of RelA are required for repression of the transcriptional activity of the glucocorticoid receptor in intact cells. However, only the Rel homology domain of RelA was found to associate with the glucocorticoid receptor in vitro. RelA mutants, not able to repress glucocorticoid receptor activity, but still able to dimerize, behaved as transdominant inhibitors of the repressive activity of wild type RelA. Furthermore, we show that the 13 S E1A protein is able to interfere with the transrepressive activity of RelA. We propose that negative cross-talk between the glucocorticoid receptor and RelA is due to direct interaction via the Rel homology domain of RelA and the DNA binding domain of the glucocorticoid receptor in combination with interference by the transactivation domains of RelA with the transcriptional activity of the glucocorticoid receptor.
Collapse
Affiliation(s)
- S Wissink
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
1189
|
Hong SH, David G, Wong CW, Dejean A, Privalsky ML. SMRT corepressor interacts with PLZF and with the PML-retinoic acid receptor alpha (RARalpha) and PLZF-RARalpha oncoproteins associated with acute promyelocytic leukemia. Proc Natl Acad Sci U S A 1997; 94:9028-33. [PMID: 9256429 PMCID: PMC23013 DOI: 10.1073/pnas.94.17.9028] [Citation(s) in RCA: 295] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Retinoic acid receptors (RARs) are hormone-regulated transcription factors that control key aspects of normal differentiation. Aberrant RAR activity may be a causal factor in neoplasia. Human acute promyelocytic leukemia, for example, is tightly linked to chromosomal translocations that fuse novel amino acid sequences (denoted PML, PLZF, and NPM) to the DNA-binding and hormone-binding domains of RARalpha. The resulting chimeric receptors have unique transcriptional properties that may contribute to leukemogenesis. Normal RARs repress gene transcription by associating with ancillary factors denoted corepressors (also referred to as SMRT, N-CoR, TRAC, or RIP13). We report here that the PML-RARalpha and PLZF-RARalpha oncoproteins retain the ability of RARalpha to associate with corepressors, and that this corepressor association correlates with certain aspects of the leukemic phenotype. Unexpectedly, the PLZF moiety itself can interact with SMRT corepressor. This interaction with corepressor is mediated, in part, by a POZ motif within PLZF. Given the presence of POZ motifs in a number of known transcriptional repressors, similar interactions with SMRT may play a role in transcriptional silencing by a variety of both receptor and nonreceptor transcription factors.
Collapse
Affiliation(s)
- S H Hong
- Section of Microbiology, Division of Biological Sciences, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
1190
|
Chang KH, Chen Y, Chen TT, Chou WH, Chen PL, Ma YY, Yang-Feng TL, Leng X, Tsai MJ, O'Malley BW, Lee WH. A thyroid hormone receptor coactivator negatively regulated by the retinoblastoma protein. Proc Natl Acad Sci U S A 1997; 94:9040-5. [PMID: 9256431 PMCID: PMC23019 DOI: 10.1073/pnas.94.17.9040] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The retinoblastoma protein (Rb) plays a critical role in cell proliferation, differentiation, and development. To decipher the mechanism of Rb function at the molecular level, we have systematically characterized a number of Rb-interacting proteins, among which is the clone C5 described here, which encodes a protein of 1,978 amino acids with an estimated molecular mass of 230 kDa. The corresponding gene was assigned to chromosome 14q31, the same region where genetic alterations have been associated with several abnormalities of thyroid hormone response. The protein uses two distinct regions to bind Rb and thyroid hormone receptor (TR), respectively, and thus was named Trip230. Trip230 binds to Rb independently of thyroid hormone while it forms a complex with TR in a thyroid hormone-dependent manner. Ectopic expression of the protein Trip230 in cells, but not a mutant form that does not bind to TR, enhances specifically TR-dependent transcriptional activity. Coexpression of wild-type Rb, but not mutant Rb that fails to bind to Trip230, inhibits such activity. These results not only identify a coactivator molecule that modulates TR activity, but also uncover a role for Rb in a pathway that responds to thyroid hormone.
Collapse
Affiliation(s)
- K H Chang
- Department of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, 15355 Lambda Drive, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1191
|
Sun Z, Pan J, Balk SP. Androgen receptor-associated protein complex binds upstream of the androgen-responsive elements in the promoters of human prostate-specific antigen and kallikrein 2 genes. Nucleic Acids Res 1997; 25:3318-25. [PMID: 9241247 PMCID: PMC146870 DOI: 10.1093/nar/25.16.3318] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
An increasing number of proteins which bind to hormone-dependent nuclear receptors and mediate their effects on gene expression are being identified. The human prostate-specific antigen (PSA) and kallikrein 2 (KLK2) genes are regulated by the androgen receptor (AR). Using electrophoresis mobility shift assays (EMSA), a common nuclear protein(s) which binds upstream of the androgen-responsive elements (AREs) in the PSA and KLK2 promoters was identified. Binding occurred between bp -539 and -399 and bp -349 and -224 in the PSA and KLK2 promoters respectively, which were shown previously to be necessary for AR-mediated transactivation. Glutathione S-transferase (GST)-AR fusion proteins were constructed to determine whether the AR interacted directly with this protein or protein complex. Specific interactions were observed with AR fusion proteins containing the DNA binding domain. EMSA supershift experiments and GST-AR pull-down experiments followed by Western blotting identified a Fos-related protein(s) of approximately 40 kDa as part of this complex. Competition experiments with a double-stranded oligonucleotide containing an AP-1 binding site demonstrated that DNA binding was not mediated by AP-1. These results indicate that a Fos-containing protein complex distinct from AP-1 binds upstream of the AREs in the PSA and KLK2 promoters, interacts with the AR and may participate in regulation of these two androgen-responsive genes.
Collapse
Affiliation(s)
- Z Sun
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
1192
|
Dorssers LC, Veldscholte J. Identification of a novel breast-cancer-anti-estrogen-resistance (BCAR2) locus by cell-fusion-mediated gene transfer in human breast-cancer cells. Int J Cancer 1997; 72:700-5. [PMID: 9259413 DOI: 10.1002/(sici)1097-0215(19970807)72:4<700::aid-ijc24>3.0.co;2-d] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Development of anti-estrogen resistance limits the benefit of endocrine therapy of breast cancer. The mechanistic basis for resistance to the anti-estrogen tamoxifen may involve (epi)genetic alterations within tumor cells. We have initiated a random search for genes allowing estrogen-dependent ZR-75-1 human breast-cancer cells to proliferate in the presence of tamoxifen. The strategy was based on insertion mutagenesis of ZR-75-1 cells using defective retrovirus and subsequent identification of common integration sites. As an alternative approach to identify integration loci involved in anti-estrogen resistance, we have applied cell fusion. Integration regions from lethally irradiated, tamoxifen-resistant cells were transferred to hygromycin B-resistant ZR-75-1 cells. Somatic cell hybrids were established by selection for resistance to G418 (encoded by the integrated virus) and hygromycin B. Individual integration loci were thus separated among different cell hybrids and tested for their role in anti-estrogen resistance. Analysis of a panel of 29 somatic-cell hybrids revealed that tamoxifen resistance co-segregated with only 1 of the 2 integration loci present in the tamoxifen-resistant donor cell line. This locus was further identified as a common integration site in our panel of tamoxifen-resistant cell clones. Our results designate this integration site as the second breast-cancer-anti-estrogen-resistance locus (BCAR2), which most likely contains a gene responsible for the anti-estrogen-resistant phenotype in close proximity to the integrated virus. Our data also imply that individual genes can alter the estrogen dependency of human breast-cancer cells in a dominant manner in vitro.
Collapse
Affiliation(s)
- L C Dorssers
- Department of Molecular Biology, Dr. Daniel den Hoed Cancer Center/University Hospital Rotterdam, The Netherlands.
| | | |
Collapse
|
1193
|
Li H, Gomes PJ, Chen JD. RAC3, a steroid/nuclear receptor-associated coactivator that is related to SRC-1 and TIF2. Proc Natl Acad Sci U S A 1997; 94:8479-84. [PMID: 9238002 PMCID: PMC22964 DOI: 10.1073/pnas.94.16.8479] [Citation(s) in RCA: 433] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Steroids, thyroid hormones, vitamin D3, and retinoids are lipophilic small molecules that regulate diverse biological effects such as cell differentiation, development, and homeostasis. The actions of these hormones are mediated by steroid/nuclear receptors which function as ligand-dependent transcriptional regulators. Transcriptional activation by ligand-bound receptors is a complex process requiring dissociation and recruitment of several additional cofactors. We report here the cloning and characterization of receptor-associated coactivator 3 (RAC3), a human transcriptional coactivator for steroid/nuclear receptors. RAC3 interacts with several liganded receptors through a mechanism which requires their respective ligand-dependent activation domains. RAC3 can activate transcription when tethered to a heterologous DNA-binding domain. Overexpression of RAC3 enhances the ligand-dependent transcriptional activation by the receptors in mammalian cells. Sequence analysis reveals that RAC3 is related to steroid receptor coactivator 1 (SRC-1) and transcriptional intermediate factor 2 (TIF2), two of the most potent coactivators for steroid/nuclear receptors. Thus, RAC3 is a member of a growing coactivator network that should be useful as a tool for understanding hormone action and as a target for developing new therapeutic agents that can block hormone-dependent neoplasia.
Collapse
Affiliation(s)
- H Li
- Department of Pharmacology and Molecular Toxicology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655-0126, USA
| | | | | |
Collapse
|
1194
|
Wallimann P, Marti T, Fürer A, Diederich F. Steroids in Molecular Recognition. Chem Rev 1997; 97:1567-1608. [PMID: 11851459 DOI: 10.1021/cr960373b] [Citation(s) in RCA: 185] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Peter Wallimann
- Laboratorium für Organische Chemie, Universitätstrasse 16, ETH-Zentrum, 8092-Zürich, Switzerland
| | | | | | | |
Collapse
|
1195
|
Abstract
There is a recognized need for eukaryotic molecular gene switches that are tightly regulated by the administration of small molecule drugs. The modular nature of intracellular receptor proteins has allowed the recent development of chimeric receptors that fulfill this need. These switches will help dissect the roles that specific proteins play in signaling pathways, cell differentiation and development. Some of these switches will also be used in gene therapy to regulate therapeutic gene expression.
Collapse
Affiliation(s)
- V E Allgood
- GeneMedicine, Inc., The Woodlands, TX 77381, USA.
| | | |
Collapse
|
1196
|
Miller CW, Morosetti R, Campbell MJ, Mendoza S, Koeffler HP. Integrity of the 1,25-dihydroxyvitamin D3 receptor in bone, lung, and other cancers. Mol Carcinog 1997; 19:254-7. [PMID: 9290702 DOI: 10.1002/(sici)1098-2744(199708)19:4<254::aid-mc6>3.0.co;2-c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Differentiation and proliferation can be regulated in diverse cell types by 1,25-dihydroxyvitamin D3. These effects derive from modulation of gene expression mediated by the interaction of 1,25-dihydroxyvitamin D3 with the vitamin D receptor (VDR). The VDR is one of the nuclear hormone receptors. Because these transcription factors play a key role in growth control, some nuclear hormone receptors, such as the retinoic acid receptor alpha, can be disrupted in cancer. With these alterations in mind, we looked for alterations of the VDR gene in a variety of cancers, including 68 osteosarcomas, 23 other sarcomas, 34 non-small cell lung cancers, and 44 cell lines representing many tumor types. Gross integrity of the VDR gene was examined on Southern blots probed with the coding region of the VDR cDNA. The presence of point mutations targeting VDR exons 2-7 was assessed by polymerase chain reaction-single-strand conformation polymorphism analysis and direct DNA sequencing. Two alterations were detected; direct DNA sequencing of these samples revealed one silent mutation in codon 79 and a base change in intron 3. These results suggest that mutations and rearrangement of the VDR do not play a role in the cancers studied.
Collapse
Affiliation(s)
- C W Miller
- Division of Hematology/Oncology, Cedars-Sinai Research Institute, University of California at Los Angeles School of Medicine 90048, USA
| | | | | | | | | |
Collapse
|
1197
|
Baniahmad A, Thormeyer D, Renkawitz R. tau4/tau c/AF-2 of the thyroid hormone receptor relieves silencing of the retinoic acid receptor silencer core independent of both tau4 activation function and full dissociation of corepressors. Mol Cell Biol 1997; 17:4259-71. [PMID: 9234683 PMCID: PMC232279 DOI: 10.1128/mcb.17.8.4259] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Members of the thyroid hormone (TR)-retinoic acid receptor (RAR) subfamily of nuclear hormone receptors silence gene expression in the absence of hormone. Addition of cognate ligands leads to dissociation of corepressors, association of coactivators, and transcriptional activation. Here, we used the hRAR alpha silencer core, which encompasses the ligand binding domain, including receptor regions D and E of RAR alpha without the activation function called tau4/tau c/AF-2 and without the F region, to analyze the mechanisms by which transcriptional silencing is relieved. Although the RAR silencer core is able to bind ligand, it acts as a constitutive transcriptional silencer. We have fused various small activation domains to the C terminus of the silencer core and analyzed hormone-dependent changes in receptor function. We show that nine amino acids derived from the hTRbeta are sufficient to transform the RAR silencer core into a hormone-dependent activator. Lengthening the linker between the silencer core and these nine amino acids is not critical for mediating ligand-induced relief of silencing and activation. In addition, we show that a transactivation function at the C terminus is not required for relief of silencing by the hormone, but it is required for transcriptional activation. Furthermore, we created functional silencer fusions which lose their repressive function upon addition of hormone, although the corepressors SMRT and N-CoR remain attached to the receptor.
Collapse
Affiliation(s)
- A Baniahmad
- Genetisches Institut der Justus-Liebig Universität, Giessen, Germany.
| | | | | |
Collapse
|
1198
|
Moilanen A, Rouleau N, Ikonen T, Palvimo JJ, Jänne OA. The presence of a transcription activation function in the hormone-binding domain of androgen receptor is revealed by studies in yeast cells. FEBS Lett 1997; 412:355-8. [PMID: 9256251 DOI: 10.1016/s0014-5793(97)00791-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To assess the importance of various regions of the androgen receptor (AR) in transcriptional regulation, we have compared its activation functions (AFs) in yeast and mammalian cells. The receptor's amino-terminal region contains a major transcriptional activator (AF-1) in both cell types, whereas AF-2 in the ligand-binding domain (LBD) is very weak in mammalian cells but clearly functional in the yeast. Hormone-binding ability of LBD is mandatory for AF-2 to operate, as illustrated by mutated LBD constructs. The activity of AF-2 in yeast is severely attenuated when the hinge region is attached to LBD, suggesting that the former region modulates AF-2 in vivo, probably by presenting an interface for interacting proteins.
Collapse
Affiliation(s)
- A Moilanen
- Department of Physiology, Institute of Biomedicine, University of Helsinki, Finland
| | | | | | | | | |
Collapse
|
1199
|
Batistuzzo de Medeiros SR, Krey G, Hihi AK, Wahli W. Functional interactions between the estrogen receptor and the transcription activator Sp1 regulate the estrogen-dependent transcriptional activity of the vitellogenin A1 io promoter. J Biol Chem 1997; 272:18250-60. [PMID: 9218463 DOI: 10.1074/jbc.272.29.18250] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Two distinct, TATA box-containing promoters regulate the transcriptional activity of the Xenopus vitellogenin A1 gene. These two promoters are of different strength and are separated by 1.8 kilobase pairs of untranslated sequence. Estrogen receptor (ER) and its ligand, 17beta-estradiol, induce the activity of both promoters. The estrogen response elements (EREs) are located proximal to the downstream i promoter while no ERE-like sequences have been identified in the vicinity of the upstream io promoter. We show here, that transcriptional activity of the upstream io promoter is Sp1-dependent. Moreover, we demonstrate that estrogen inducibility of the io promoter results from functional interactions between the io bound Sp1 and the ER bound at the proximity of i. Functional interactions between Sp1 and ER do not require the presence of a TATA box for transcriptional activation, as is demonstrated using the acyl-CoA oxidase promoter. The relative positions that ER and Sp1 occupy with respect to the initiation site determines whether these two transcription activators can synergize for transcription initiation.
Collapse
|
1200
|
Burmester T, Scheller K. Developmentally controlled cleavage of the Calliphora arylphorin receptor and posttranslational action of the steroid hormone 20-hydroxyecdysone. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 247:695-702. [PMID: 9266715 DOI: 10.1111/j.1432-1033.1997.00695.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In response to a rise in ecdysteroid titre, fat body cells of insect larvae take up storage proteins from the haemolymph by receptor-mediated endocytosis. Here we show that the receptor responsible for incorporation of the major haemolymph protein arylphorin of the blowfly, Calliphora vicina, is subject to an unusual posttranslational processing that involves three distinct cleavage steps. After the removal of a 17-amino-acid signal peptide, a receptor precursor of 141 kDa is released. Before reaching the cell surface, the precursor is cleaved a second time, giving rise to the active 92-kDa arylphorin receptor, plus a 48-kDa peptide. The function of this 48-kDa peptide may be the prevention of premature ligand-receptor interaction in the endoplasmic reticulum. 20-Hydroxyecdysone initiates a third cleavage step of the arylphorin receptor, which results in a 62-kDa arylphorin binding protein and a 30-kDa peptide. Contrary to the standard model of steroid hormone action, the process which give rise to receptor cleavage can be induced by 20-hydroxyecdysone in vivo and in vitro even in absence of protein biosynthesis.
Collapse
Affiliation(s)
- T Burmester
- Institut Jacques-Monod, Département Biologie du Développement, CNRS et Université Paris 7, France.
| | | |
Collapse
|