1901
|
Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J, Heyman RA, Staels B, Auwerx J. Retinoids increase human apolipoprotein A-11 expression through activation of the retinoid X receptor but not the retinoic acid receptor. Mol Cell Biol 1996; 16:3350-60. [PMID: 8668150 PMCID: PMC231329 DOI: 10.1128/mcb.16.7.3350] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Considering the link between plasma high-density lipoprotein (HDL) cholesterol levels and a protective effect against coronary artery disease as well as the suggested beneficial effects of retinoids on the production of the major HDL apolipoprotein (apo), apo A-I, the goal of this study was to analyze the influence of retinoids on the expression of apo A-II, the other major HDL protein. Retinoic acid (RA) derivatives have a direct effect on hepatic apo A-II production, since all-trans (at) RA induces apo A-II mRNA levels and apo A-II secretion in primary cultures of human hepatocytes. In the HepG2 human hepatoblastoma cell line, both at-RA and 9-cis RA as well as the retinoid X receptor (RXR)-specific agonist LGD 1069, but not the RA receptor (RAR) agonist ethyl-p-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthyl)-l-pro penyl]-benzoic acid (TTNPB), induce apo A-II mRNA levels. Transient-transfection experiments with a reporter construct driven by the human apo A-II gene promoter indicated that 9-cis RA and at-RA, as well as the RXR agonists LGD 1069 and LG 100268, induced apo A-II gene expression at the transcriptional level. Only minimal effects of the RAR agonist TTNPB were observed on the apo A-II promoter reporter construct. Unilateral deletions and site-directed mutagenesis identified the J site of the apo A-II promoter mediating the responsiveness to RA. This element contains two imperfect half-sites spaced by 1 oligonucleotide. Cotransfection assays in combination with the use of RXR or RAR agonists showed that RXR but not RAR transactivates the apo A-II promoter through this element. By contrast, RAR inhibits the inductive effects of RXR on the apo A-II J site in a dose-dependent fashion. Gel retardation assays demonstrated that RXR homodimers bind, although with a lower affinity than RAR-RXR heterodimers, to the AH-RXR response element. In conclusion, retinoids induce hepatic apo A-II production at the transcriptional level via the interaction of RXR with an element in the J site containing two imperfect half-sites spaced by 1 oligonucleotide, thereby demonstrating an important role of RXR in controlling human lipoprotein metabolism. Since the J site also confers responsiveness of the apo A-II gene to fibrates and fatty acids via the activation of peroxisome proliferator-activated receptor-RXR heterodimers, this site can be considered a plurimetabolic response element.
Collapse
Affiliation(s)
- N Vu-Dac
- Unite 325 Institut National de la Santé et de la Recherche Médicale, Département d'Athérosclerose, Institut Pasteur de Lille, France
| | | | | | | | | | | | | |
Collapse
|
1902
|
Tan NS, Lam TJ, Ding JL. The first contiguous estrogen receptor gene from a fish, Oreochromis aureus: evidence for multiple transcripts. Mol Cell Endocrinol 1996; 120:177-92. [PMID: 8832578 DOI: 10.1016/0303-7207(96)03836-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The O. aureus estrogen receptor (OaER) gene of 40.4 kb containing ten exons is the first complete piscine gene to be cloned. There are two extra introns: intron I that divides the 5' UTR into two exons, and intron V that intersperses D and E1 exons. Except for I and V, other introns have identical positions to those of human ER gene. All the donor and acceptor splice sites exhibit consensus sequences. The promoter lacks consensus TATA and CAAT boxes. This region exhibits several putative regulatory elements. A functional imperfect ERE deviating at two bases is located in the leader exon, thus suggesting that this gene is autoregulated. The OaER gene lacks an A region whereas its C and E domains are highly conserved. Within the ER subfamily, OaER exhibits the longest F domain of 77 amino acids. OaER has a long 3'UTR constituting >1/2 of its transcript. Using RT-PCR and SI nuclease mapping, we report for the first time the usage of both alternative transcriptional start sites and polyadenylation signals during estrogen-induced OaER expression. Thus, O. aureus may have four species of ER transcripts differing structurally in their transcriptional start sites and lengths of their 3' UTR.
Collapse
Affiliation(s)
- N S Tan
- Department of Zoology, National University of Singapore, Singapore
| | | | | |
Collapse
|
1903
|
Schulman G, Lindemeyer R, Barman A, Karnik S, Bastl CP. Atrial natriuretic peptide inhibits mineralocorticoid receptor function in rat colonic surface cells. J Clin Invest 1996; 98:157-66. [PMID: 8690788 PMCID: PMC507412 DOI: 10.1172/jci118761] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Atrial natriuretic peptide (ANP) inhibits and aldosterone (ALDO) stimulates Na conductive transport. Therefore, the effects of ANP and its second messenger cGMP on mineralocorticoid receptor (MR) function in rat colon surface and crypt cells were examined. 100 nM 8-Br-cGMP decreased surface [3H]ALDO binding by 42 +/- 4% but increased crypt [3HvALDO binding by 52+/-16%. ANP decreased surface [3H]ALDO binding by approximately 50% after a 2.5-h lag period but had no effect on crypt ALDO binding. ANP and cGMP rapidly (< 15 min) inhibited surface cell ALDO-induced MR nuclear translocation but did not affect crypt MR nuclear translocation. Inhibition of cGMP-dependent protein kinase with KT5823 blocked the inhibitory effects of ANP and 8-Br-cGMP on surface cell ALDO binding and MR nuclear translocation. In crypt, KT5823 increased baseline [3H]ALDO binding but did not inhibit the stimulatory effect of exogenous cGMP. DEAE-cellulose chromatography and gel mobility shift assay showed that ANP did not inhibit surface MR activation. ANP inhibited ALDO stimulated short circuit current in distal colon. These data demonstrate cell-specific regulation of MR function. In surface cells, ANP rapidly inhibits MR nuclear translocation and ALDO-induced short circuit current. ANP inhibition of MR function may be an additional mechanism of ANP antagonism of Na reabsorption.
Collapse
Affiliation(s)
- G Schulman
- Department of Medicine, Temple University Health Sciences Center, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | |
Collapse
|
1904
|
Abstract
A mouse cDNA encoding a putative DNA-binding protein of the zinc-finger type was isolated from an E8.5 mouse embryonic cDNA library. Sequence comparison revealed a high degree of homology between this mouse cDNA and the human and rat orphan receptor Tr2-11 isolated from prostate cDNA libraries. This transcript was detected in early-to-midgestation embryos and was seen to level off during later stages of development. In adult animals, a high level of expression was detected only in the testis, starting at postnatal day 18, a stage when active meiosis begins to occur. A specific antibody was raised, and immunoreactive signal was specifically located in the adlumenal compartment of the seminiferous tubule, where advanced germ cells reside. In mice fed a vitamin A-depleted diet, where the testes were depleted of advanced germ cells, expression of this protein could not be detected, suggesting a biological relation of this orphan receptor and male germ-cell differentiation. Using a retinoic acid response element (RARE)-containing reporter system, it was demonstrated that expression of this protein dramatically repressed both the basal and the retinoic acid (RA)-regulated promoter activities of this reporter. Thus, this orphan receptor could play a role in modulating both the basic transcription machinery and the RA signalling pathway during embryogenesis and male germ-cell differentiation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies/immunology
- Base Sequence
- Blotting, Northern
- COS Cells
- Cloning, Molecular
- DNA, Complementary
- Fluorescent Antibody Technique, Indirect
- Gene Expression
- Genes, Reporter
- Humans
- Luciferases/genetics
- Male
- Mice/embryology
- Molecular Sequence Data
- Nuclear Receptor Subfamily 2, Group C, Member 1
- Receptors, Retinoic Acid/metabolism
- Receptors, Steroid/genetics
- Receptors, Thyroid Hormone/genetics
- Regulatory Sequences, Nucleic Acid
- Sequence Homology, Amino Acid
- Testis/metabolism
- Tissue Distribution
- Vitamin A Deficiency
Collapse
Affiliation(s)
- C H Lee
- Department of Pharmacology, University of Minnesota, Minneapolis 55455, USA
| | | | | |
Collapse
|
1905
|
Katzenellenbogen JA, Katzenellenbogen BS. Nuclear hormone receptors: ligand-activated regulators of transcription and diverse cell responses. CHEMISTRY & BIOLOGY 1996; 3:529-36. [PMID: 8807884 DOI: 10.1016/s1074-5521(96)90143-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Signal transduction via nuclear hormone receptors is unusual in that the hormone ligand forms an integral part of the protein complex involved in DNA binding and transcriptional activation. New structural and biochemical results have begun to unravel how these receptors produce different effects in different cells, and the structural changes involved in transcriptional activation.
Collapse
|
1906
|
Balbín M, López-Otín C. Hormonal regulation of the human pepsinogen C gene in breast cancer cells. Identification of a cis-acting element mediating its induction by androgens, glucocorticoids, and progesterone. J Biol Chem 1996; 271:15175-81. [PMID: 8663058 DOI: 10.1074/jbc.271.25.15175] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Pepsinogen C is an aspartic proteinase mainly involved in the digestion of proteins in the stomach, which is also synthesized by certain human breast tumors. To examine the possibility that extragastric production of this proteolytic enzyme could be mediated by hormonal factors, we have analyzed pepsinogen C gene expression in human breast cancer cells subjected to different hormonal treatments. Northern blot analyses revealed the expression of pepsinogen C gene by T-47D breast cancer cells after induction with dihydrotestosterone, dexamethasone, and progesterone but not with estradiol, retinoic acid, or ethanol. Reverse transcription-polymerase chain reaction analysis in a series of breast cancer cell lines confirmed the amplification of pepsinogen C mRNA after induction with dihydrotestosterone, in those cells expressing the androgen receptor mRNA. The promoter region of the pepsinogen C gene was functionally characterized by transient expression of a vector containing the promoter region cloned in front of the chloramphenicol acetyltransferase (CAT) reporter gene. CAT activity in T-47D cells was stimulated in the presence of dihydrotestosterone, dexamethasone, and progesterone but not by estradiol. By further deletion mapping of the pepsinogen C promoter, a minimal region (AGAACTattTGTTCC) was identified as being responsible for glucocorticoid-, androgen-, and progesterone-regulated gene expression.
Collapse
Affiliation(s)
- M Balbín
- Departamento de Biología Funcional, Universidad de Oviedo, 33006 Oviedo, Spain
| | | |
Collapse
|
1907
|
Marcus SL, Capone JP, Rachubinski RA. Identification of COUP-TFII as a peroxisome proliferator response element binding factor using genetic selection in yeast: COUP-TFII activates transcription in yeast but antagonizes PPAR signaling in mammalian cells. Mol Cell Endocrinol 1996; 120:31-9. [PMID: 8809736 DOI: 10.1016/0303-7207(96)03813-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-response elements (PPRE) are cis-acting regulatory elements that confer responsiveness to peroxisome proliferators and various fatty acids by serving as target sites for ligand-activated peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor (RXR) heterodimers. Other cellular factors, including additional nuclear hormone receptors, also interact with PPREs and modulate PPAR function. We have developed a positive selection strategy in yeast to identify mammalian factors that functionally interact with PPREs. Saccharomyces cerevisiae containing an integrated copy of the HIS3 gene under transcriptional control of a minimal CYC1 promoter and two copies of the rat enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase PPRE was constructed and transformed with a rat liver cDNA yeast expression library. Plasmids were isolated from his + transformants. One plasmid contained a cDNA encoding the complete rat chicken ovalbumin upstream promoter transcription factor II (COUP-TFII), an orphan member of the nuclear hormone receptor superfamily. COUP-TFII potently activated PPRE-linked reporter gene expression in yeast, and COUP-TFII synthesized in yeast or in vitro formed specific protein/DNA complexes with this PPRE. Significantly, COUP-TFII did not activate transcription of PPRE-linked reporter genes in mammalian cells but rather strongly inhibited induction mediated by PPAR/RXR. Our findings demonstrate the utility of using genetic screening in yeast to identify sequence-specific DNA binding transcription factors.
Collapse
Affiliation(s)
- S L Marcus
- Department of Anatomy and Cell Biology, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
1908
|
Abstract
Human genital skin fibroblasts contain both the full-length 110 K androgen receptor protein (AR-B, apparent M(r) approximately 110,000) and an 87 K N-terminally truncated AR isoform (AR-A, apparent M(r) approximately 87,000). These two AR species are structurally analogous to the A- and B-isoforms of the progesterone receptor (PR). We examined the distribution pattern of human AR isoforms in a variety of fetal and adult tissues by Western blot analysis. Relative levels of immunoreactive AR proteins in high salt tissue extracts were estimated by densitometry in comparison to a standard normal genital skin fibroblast preparation. High AR levels (AR-A + AR-B = 0.8-7.7) were present in male and female reproductive tissues from mid-trimester fetuses, including penis, prostate, testis, epididymis, scrotal skin, labial skin, uterus/cervix, and ovary. AR-A and AR-B (0.08-0.9) also were found in 14 non-genital fetal tissues (bladder, fat, lung, great vessel, trachea, muscle, scalp skin, kidney, thyroid, intestine, thymus, ureter, stomach and rectum). AR-A accounted for 4-26% of the AR protein detected in these tissues. Ten other fetal tissues had low levels of AR-B (0.02-0.3) and little or no detectable AR-A. AR-B also was the predominant or only immunoreactive AR species found in 17 adult human tissues. AR levels in adult reproductive tissues (prostate, endometrium, ovary, uterus, fallopian tube, testis, seminal vesicle, myometrium, and ejaculatory duct) ranged from 0.1 to 2.2. Immunoreactive AR (0.4-0.8) also was present in specimens of prostate carcinoma, endometrial carcinoma, thyroid carcinoma and kidney. Lower levels of AR (0.03-0.1) were detected in adult breast, colon, lung and adrenal gland specimens. This study demonstrates that immunoreactive AR protein is present in a wide variety of human fetal and adult tissues and that two AR isoforms are expressed in many tissues.
Collapse
Affiliation(s)
- C M Wilson
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas 75235-9041, USA
| | | |
Collapse
|
1909
|
Harbers M, Wahlström GM, Vennström B. Transactivation by the thyroid hormone receptor is dependent on the spacer sequence in hormone response elements containing directly repeated half-sites. Nucleic Acids Res 1996; 24:2252-9. [PMID: 8710493 PMCID: PMC145925 DOI: 10.1093/nar/24.12.2252] [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: 02/01/2023] Open
Abstract
The thyroid hormone receptor (TR) regulates the transcription of its target genes by interacting with specific hormone response elements consisting usually of directly repeated half-sites with the consensus sequence AGGTCA. To investigate the role of the spacer sequences separating the half-sites, heterodimers formed by TRalpha and the retinoid-X receptor (RXR) were used in a PCR based selection and amplification assay. The TRalpha/RXR heterodimer selected for elements with directly repeated half-sites having a spacer of 4 nucleotides (DR4). Preferences for nucleotides in the TR binding half-site motif as well as for the 4 nucleotides separating the two half-sites were found. DNA binding and transfection studies using DR4 elements with different spacer sequences showed the importance of these nucleotides for the activity of the response element: some spacer sequences allowed little or no transactivation from the element, whereas other sequences supported strong transactivation. A pyrimidine nucleotide in position three of the spacer enhanced TRalpha binding and transactivation. Additional experiments showed that heterodimers between RXR and other putative receptors exhibited a similar but distinct specificity for the spacer sequence. Our results thus suggest that the four nucleotides separating the two half-sites in hormone response elements have a major role in determining induction of hormone responsive genes.
Collapse
Affiliation(s)
- M Harbers
- Karolinska Institute, Medical Nobel Institute, Department of Cell and Molecular Biology, Laboratory of Developmental Biology, Stockholm, Sweden
| | | | | |
Collapse
|
1910
|
Bamberger AM, Bamberger CM, Gellersen B, Schulte HM. Modulation of AP-1 activity by the human progesterone receptor in endometrial adenocarcinoma cells. Proc Natl Acad Sci U S A 1996; 93:6169-74. [PMID: 8650238 PMCID: PMC39208 DOI: 10.1073/pnas.93.12.6169] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The composite transcription factor activating protein 1 (AP-1) integrates various mitogenic signals in a large number of cell types, and is therefore a major regulator of cell proliferation. In the normal human endometrium, proliferation and differentiation alternate in a cyclic fashion, with progesterone being largely implicated in the latter process. However, the effects of progesterone and the progesterone receptor (hPR) on AP-1 activity in the human endometrium are not known. To address this issue, HEC-1-B endometrial adenocarcinoma cells, which are devoid of hPR, were transfected with luciferase reporter constructs driven by two different AP-1-dependent promoters. Unexpectedly, cotransfection of hPR caused a marked induction of luciferase activity in the absence of ligand on both promoters. The magnitude of this induction was similar to that observed in response to the phorbol ester TPA. Addition of ligand reversed the stimulating effect of the unliganded hPR on AM activity in these cells. These effects were specific for hPR, and were not observed with either human estrogen receptor or human glucocorticoid receptor. Furthermore, they strictly depended on the presence of AP-1-responsive sequences within target promoters. Finally, the described effects of hPR on AP-1 activity were shown to be cell-type specific, because they could not be demonstrated in SKUT-1-B, JEG-3, and COS-7 cells. To our knowledge this is the first report of an unliganded steroid receptor stimulating AP-1 activity. This effect and its reversal in the presence of ligand suggest a novel mechanism, through which hPR can act as a key regulator of both proliferation and differentiation in the human endometrium.
Collapse
Affiliation(s)
- A M Bamberger
- IHF Institute for Hormone and Fertility Research, University of Hamburg, Germany
| | | | | | | |
Collapse
|
1911
|
Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci U S A 1996; 93:5925-30. [PMID: 8650195 PMCID: PMC39164 DOI: 10.1073/pnas.93.12.5925] [Citation(s) in RCA: 3232] [Impact Index Per Article: 111.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
We have cloned a novel member of the nuclear receptor superfamily. The cDNA of clone 29 was isolated from a rat prostate cDNA library and it encodes a protein of 485 amino acid residues with a calculated molecular weight of 54.2 kDa. Clone 29 protein is unique in that it is highly homologous to the rat estrogen receptor (ER) protein, particularly in the DNA-binding domain (95%) and in the C-terminal ligand-binding domain (55%). Expression of clone 29 in rat tissues was investigated by in situ hybridization and prominent expression was found in prostate and ovary. In the prostate clone 29 is expressed in the epithelial cells of the secretory alveoli, whereas in the ovary the granuloma cells in primary, secondary, and mature follicles showed expression of clone 29. Saturation ligand-binding analysis of in vitro synthesized clone 29 protein revealed a single binding component for 17beta-estradiol (E2) with high affinity (Kd= 0.6 nM). In ligand-competition experiments the binding affinity decreased in the order E2 > diethylstilbestrol > estriol > estrone > 5alpha-androstane-3beta,17beta-diol >> testosterone = progesterone = corticosterone = 5alpha-androstane-3alpha,17beta-diol. In cotransfection experiments of Chinese hamster ovary cells with a clone 29 expression vector and an estrogen-regulated reporter gene, maximal stimulation (about 3-fold) of reporter gene activity was found during incubation with 10 nM of E2. Neither progesterone, testosterone, dexamethasone, thyroid hormone, all-trans-retinoic acid, nor 5alpha-androstane-3alpha,I7beta-diol could stimulate reporter gene activity, whereas estrone and 5alpha-androstane-3beta,17beta-diol did. We conclude that clone 29 cDNA encodes a novel rat ER, which we suggest be named rat ERbeta to distinguish it from the previously cloned ER (ERalpha) from rat uterus.
Collapse
Affiliation(s)
- G G Kuiper
- Center for Biotechnology and Department of Medical Nutrition, Karolinska Institute, Huddinge, Sweden
| | | | | | | | | |
Collapse
|
1912
|
Affiliation(s)
- A Robbins
- Population Council Center for Biomedical Research, New York, New York 10021, USA
| | | |
Collapse
|
1913
|
Zhang X, Jeyakumar M, Bagchi MK. Ligand-dependent Cross-talk between Steroid and Thyroid Hormone Receptors. J Biol Chem 1996. [DOI: 10.1074/jbc.271.25.14825] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
1914
|
Bai W, Weigel NL. Phosphorylation of Ser211 in the chicken progesterone receptor modulates its transcriptional activity. J Biol Chem 1996; 271:12801-6. [PMID: 8662804 DOI: 10.1074/jbc.271.22.12801] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The chicken progesterone receptor has been shown to be phosphorylated in vivo at four major sites. Previous studies have shown that mutation of one of the hormone-dependent phosphorylation sites, Ser530, to alanine decreases the transcriptional activity of the receptor under conditions where ligand is limited. Here, we present evidence for the functional significance of another phosphorylation site, Ser211. Mutation of Ser211 to alanine results in a decrease in the transcriptional activity of the receptor and affects the phosphorylation-dependent decrease in mobility of the receptor in SDS-polyacrylamide gel electrophoresis. The degree of reduction in transcriptional activity is dependent on both the cell type and the reporters used in the studies but is independent of hormone concentration, suggesting that phosphorylation at Ser211 regulates the activity of the receptor through a mechanism distinct from Ser530 phosphorylation.
Collapse
Affiliation(s)
- W Bai
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
1915
|
Starr DB, Matsui W, Thomas JR, Yamamoto KR. Intracellular receptors use a common mechanism to interpret signaling information at response elements. Genes Dev 1996; 10:1271-83. [PMID: 8675013 DOI: 10.1101/gad.10.10.1271] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The glucocorticoid receptor (GR) activates transcription in certain glucocorticoid response element (GRE) contexts, and represses or displays no activity in others. We isolated point mutations in one GRE, plfG, at which GR activated transcription under conditions in which the wild-type element was inactive or conferred repression, implying that GREs may carry signals that are interpreted by bound receptors. Consistent with this notion, we identified a mutant rat GR, K461A, which activated transcription in all GRE contexts tested, implying that this residue is important in interpretation of GRE signals. In a yeast screen of 60,000 GR mutants for strong activation from plfG, all 13 mutants isolated contained substitutions at K461. This lysine residue is highly conserved in the zinc-binding region (ZBR) of the intracellular receptor (IR) superfamily; when it was mutated in MR and RARbeta, the resulting receptors similarly activated transcription at response elements that their wild-type counterparts repressed or were inactive. We suggest that IR response elements serve in part as signaling components, and that a critical lysine residue serves as an allosteric "lock" that restricts IRs to inactive or repressing configurations except in response element contexts that signal their conversion to transcriptional activators. Therefore, mutation of this residue produces altered receptors that activate in many or all response element contexts.
Collapse
Affiliation(s)
- D B Starr
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, 94143-0450, USA
| | | | | | | |
Collapse
|
1916
|
Htun H, Barsony J, Renyi I, Gould DL, Hager GL. Visualization of glucocorticoid receptor translocation and intranuclear organization in living cells with a green fluorescent protein chimera. Proc Natl Acad Sci U S A 1996; 93:4845-50. [PMID: 8643491 PMCID: PMC39367 DOI: 10.1073/pnas.93.10.4845] [Citation(s) in RCA: 289] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
A highly fluorescent mutant form of the green fluorescent protein (GFP) has been fused to the rat glucocorticoid receptor (GR). When GFP-GR is expressed in living mouse cells, it is competent for normal transactivation of the GR-responsive mouse mammary tumor virus promoter. The unliganded GFP-GR resides in the cytoplasm and translocates to the nucleus in a hormone-dependent manner with ligand specificity similar to that of the native GR receptor. Due to the resistance of the mutant GFP to photobleaching, the translocation process can be studied by time-lapse video microscopy. Confocal laser scanning microscopy showed nuclear accumulation in a discrete series of foci, excluding nucleoli. Complete receptor translocation is induced with RU486 (a ligand with little agonist activity), although concentration into nuclear foci is not observed. This reproducible pattern of transactivation-competent GR reveals a previously undescribed intranuclear architecture of GR target sites.
Collapse
Affiliation(s)
- H Htun
- Laboratory of Molecular Virology, National Cancer Institute, Bethesda, MD 20892-5055, USA
| | | | | | | | | |
Collapse
|
1917
|
Huss JM, Wang SI, Astrom A, McQuiddy P, Kasper CB. Dexamethasone responsiveness of a major glucocorticoid-inducible CYP3A gene is mediated by elements unrelated to a glucocorticoid receptor binding motif. Proc Natl Acad Sci U S A 1996; 93:4666-70. [PMID: 8643461 PMCID: PMC39336 DOI: 10.1073/pnas.93.10.4666] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Elements responsible for dexamethasone responsiveness of CYP3A23, a major glucocorticoid-inducible member of the CYP3A gene family, have been identified. DNase I footprint analysis of the proximal promoter region revealed three protected sites (sites A, B, and C) within the sequence defined by -167 to -60. Mutational analysis demonstrated that both sites B and C were necessary for maximum glucocorticoid responsiveness and functioned in a cooperative manner. Interestingly, neither site contained a glucocorticoid responsive element. Embedded in site C was an imperfect direct repeat (5'-AACTCAAAGGAGGTCA-3'), showing homology to an AGGTCA steroid receptor motif, typically recognized by the estrogen receptor family, while site B contained an ATGAACT direct repeat; these core sequences were designated dexamethasone response elements 1 and 2 (DexRE-1 and -2), respectively. Neither element has previously been associated with a glucocorticoid-activated transcriptional response. Conversion of the DexRE-1 to either a perfect thyroid hormone or vitamin D3 responsive element further enhanced induction by dexamethasone. Gel-shift analysis demonstrated that glucocorticoid receptor did not associate with either DexRE-1 or -2; hence, glucocorticoid receptor does not directly mediate glucocorticoid induction of CYP3A23. These unusual features suggest an alternate pathway through which glucocorticoids exert their effects.
Collapse
Affiliation(s)
- J M Huss
- Department of Oncology, McArdle Laboratory, University of Wisconsin, Madison, 53706, USA
| | | | | | | | | |
Collapse
|
1918
|
Hong H, Kohli K, Trivedi A, Johnson DL, Stallcup MR. GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proc Natl Acad Sci U S A 1996; 93:4948-52. [PMID: 8643509 PMCID: PMC39385 DOI: 10.1073/pnas.93.10.4948] [Citation(s) in RCA: 510] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The yeast two-hybrid system was used to isolate a clone from a 17-day-old mouse embryo cDNA library that codes for a novel 812-aa long protein fragment, glucocorticoid receptor-interacting protein 1 (GRIP1), that can interact with the hormone binding domain (HBD) of the glucocorticoid receptor. In the yeast two-hybrid system and in vitro, GRIP1 interacted with the HBDs of the glucocorticoid, estrogen, and androgen receptors in a hormone-regulated manner. When fused to the DNA binding domain of a heterologous protein, the GRIP1 fragment activated a reporter gene containing a suitable enhancer site in yeast cells and in mammalian cells, indicating that GRIP1 contains a transcriptional activation domain. Overexpression of the GRIP1 fragment in mammalian cells interfered with hormone-regulated expression of mouse mammary tumor virus-chloramphenicol acetyltransferase gene and constitutive expression of cytomegalovirus-beta-galactosidase reporter gene, but not constitutive expression from a tRNA gene promoter. This selective squelching activity suggests that GRIM can interact with an essential component of the RNA polymerase II transcription machinery. Finally, while a steroid receptor HBD fused with a GAL4 DNA binding domain did not, by itself, activate transcription of a reporter gene in yeast, coexpression of this fusion protein with GRIP1 strongly activated the reporter gene. Thus, in yeast, GRIP1 can serve as a coactivator, potentiating the transactivation functions in steroid receptor HBDs, possibly by acting as a bridge between HBDs of the receptors and the basal transcription machinery.
Collapse
Affiliation(s)
- H Hong
- Department of Pathology, University of Southern California, Los Angeles, 90033, USA
| | | | | | | | | |
Collapse
|
1919
|
Wallimann P, Seiler P, Diederich F. Dendrophanes: Novel Steroid-Recognizing Dendritic Receptors. Preliminary Communication. Helv Chim Acta 1996. [DOI: 10.1002/hlca.19960790320] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
1920
|
Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, Lin SC, Heyman RA, Rose DW, Glass CK, Rosenfeld MG. A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors. Cell 1996; 85:403-14. [PMID: 8616895 DOI: 10.1016/s0092-8674(00)81118-6] [Citation(s) in RCA: 1733] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nuclear receptors regulate gene expression by direct activation of target genes and inhibition of AP-1. Here we report that, unexpectedly, activation by nuclear receptors requires the actions of CREB-binding protein (CBP) and that inhibition of AP-1 activity is the apparent result of competition for limiting amounts of CBP/p300 in cells. Utilizing distinct domains, CBP directly interacts with the ligand-binding domain of multiple nuclear receptors and with the p160 nuclear receptor coactivators, which upon cloning have proven to be variants of the SRC-1 protein. Because CBP represents a common factor, required in addition to distinct coactivators for function of nuclear receptors, CREB, and AP-1, we suggest that CBP/p300 serves as an integrator of multiple signal transduction pathways within the nucleus.
Collapse
Affiliation(s)
- Y Kamei
- Howard Hughes Medical Institute, School of Medicine, University of California, San Diego, La Jolla, 92093-0648, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1921
|
Shi YB, Wong J, Puzianowska-Kuznicka M, Stolow MA. Tadpole competence and tissue-specific temporal regulation of amphibian metamorphosis: roles of thyroid hormone and its receptors. Bioessays 1996; 18:391-9. [PMID: 8639162 DOI: 10.1002/bies.950180509] [Citation(s) in RCA: 124] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Amphibian metamorphosis is a post-embryonic process that systematically transforms different tissues in a tadpole. Thyroid hormone plays a causative role in this complex process by inducing a cascade of gene regulation. While natural metamorphosis does not occur until endogenous thyroid hormone has been synthesized, tadpoles are competent to respond to exogenous thyroid hormone shortly after hatching. In addition, even though the metamorphic transitions of individual organs are all controlled by thyroid hormone, each occurs at distinct developmental stages. Recent molecular studies suggest that this competence of premetamorphic tadpoles to respond to the hormone and the developmental stage-dependent regulation of tissue-specific transformations are determined in part by the levels of thyroid hormone receptors and the concentrations of cellular free thyroid hormone. In addition, at least two genes, encoding a cytosolic thyroid hormone binding protein and a 5-deiodinase, respectively, are likely to be critical players in regulating cellular free thyroid hormone concentrations. This review discusses how all of these molecular components coordinate to induce amphibian metamorphosis in a correct spatial and temporal manner. These studies provide us with general clues as to how and why tissues become competent to respond to hormonal signals.
Collapse
Affiliation(s)
- Y B Shi
- Laboratory of Molecular Embryology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
1922
|
Trainor CD, Omichinski JG, Vandergon TL, Gronenborn AM, Clore GM, Felsenfeld G. A palindromic regulatory site within vertebrate GATA-1 promoters requires both zinc fingers of the GATA-1 DNA-binding domain for high-affinity interaction. Mol Cell Biol 1996; 16:2238-47. [PMID: 8628290 PMCID: PMC231211 DOI: 10.1128/mcb.16.5.2238] [Citation(s) in RCA: 186] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
GATA-1, a transcription factor essential for the development of the erythroid lineage, contains two adjacent highly conserved zinc finger motifs. The carboxy-terminal finger is necessary and sufficient for specific binding to the consensus GATA recognition sequence: mutant proteins containing only the amino-terminal finger do not bind. Here we identify a DNA sequence (GATApal) for which the GATA-1 amino-terminal finger makes a critical contribution to the strength of binding. The site occurs in the GATA-1 gene promoters of chickens, mice, and humans but occurs very infrequently in other vertebrate genes known to be regulated by GATA proteins. GATApal is a palindromic site composed of one complete [(A/T)GATA(A/G)] and one partial (GAT) canonical motif. Deletion of the partial motif changes the site to a normal GATA site and also reduces by as much as eightfold the activity of the GATA-1 promoter in an erythroid precursor cell. We propose that GATApal is important for positive regulation of GATA-1 expression in erythroid cells.
Collapse
Affiliation(s)
- C D Trainor
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
1923
|
Vilar J, Gilbert T, Moreau E, Merlet-Bénichou C. Metanephros organogenesis is highly stimulated by vitamin A derivatives in organ culture. Kidney Int 1996; 49:1478-87. [PMID: 8731117 DOI: 10.1038/ki.1996.208] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Vitamin A and its metabolic derivatives are known to be key signalling molecules in regulating morphogenetic events in vertebrate development. Here we investigated their possible involvement during mammalian kidney development using paired rat metanephros organ culture. Metanephroi were explanted from 14-day-old embryos and cultured for six days in a chemically defined medium containing a retinoid at a dose of 10(-11) to 10(-4) M. Retinol, all-trans and 9-cis retinoic acid were able to promote metanephros growth and differentiation in vitro. A significant increase in the number of nephrons was observed from 10(-8) M of retinol and 10(-10) M of all-trans retinoic acid, before any change in growth parameters. A threefold increase in the number of nephrons was obtained at a dose of 10(-6) M. At low retinoid concentrations, there was a modulating effect of triiodothyronine on retinoid-stimulated nephrogenesis since the absence of triiodothyronine in the medium enhanced the nephrogenic stimulation. Exposure of metanephroi from 13-day-old embryos to all-trans retinoic acid (10(-7) M) led to a sixfold increase of nephron formation. Finally, we analyzed the branching pattern of the ureteric bud and showed that within 48 hours of culture, it was significantly more developed upon retinoid exposure. In conclusion, this study demonstrates that retinoic acid is a key regulator of renal organogenesis in controlling nephrogenic induction processes and ureteric bud patterning, and that the younger the metanephros, the greater the effect.
Collapse
Affiliation(s)
- J Vilar
- INSERM U319, Développement Normal et Pathologique des Fonctions Epithéliales, Université Denis Diderot, Paris, France
| | | | | | | |
Collapse
|
1924
|
Leng X, Cooney AJ, Tsai SY, Tsai MJ. Molecular mechanisms of COUP-TF-mediated transcriptional repression: evidence for transrepression and active repression. Mol Cell Biol 1996; 16:2332-40. [PMID: 8628300 PMCID: PMC231221 DOI: 10.1128/mcb.16.5.2332] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
COUP-TF, an orphan member of the nuclear receptor superfamily, has been proposed to play a key role in regulating organogenesis, neurogenesis, and cellular differentiation during embryonic development. Since heterodimierization is a common theme within the nuclear receptor superfamily and has been demonstrated to modulate transcriptional properties of heterodimeric partners via allosteric interactions, we have devised a strategy to examine the silencing function of COUP-TF in a heterodimeric context. We find that the intrinsic active repression function of COUP-TF is not affected by heterodimerization. Moreover, COUP-TF can transrepress the ligand-dependent activation of its heterodimeric partners without its own DNA binding site. Using receptor deletion mutants in transfection assays, we show that the region necessary for COUP-TF silencing function is not sufficient for its transrepression activity. Furthermore, our studies indicate that in addition to its active repression function, COUP-TF can repress several different types of activator-dependent transactivation. However, this active repression function of COUP-TF may be differentially regulated by some other activator(s). These studies provide new insights into the molecular mechanism(s) of COUP-TF-mediated repression.
Collapse
Affiliation(s)
- X Leng
- Department of Cell Biology, Baylor College of Medicine, Hoston, Texas 77030, USA
| | | | | | | |
Collapse
|
1925
|
Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression. J Lipid Res 1996. [DOI: 10.1016/s0022-2275(20)42003-6] [Citation(s) in RCA: 899] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
1926
|
Yu RT, Umesono K. Transcriptional control underlying head morphogenesis during vertebrate embryonic development. J Biosci 1996. [DOI: 10.1007/bf02703093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1927
|
Delort JP, Capecchi MR. TAXI/UAS: A molecular switch to control expression of genes in vivo. Hum Gene Ther 1996; 7:809-20. [PMID: 8860833 DOI: 10.1089/hum.1996.7.7-809] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Numerous therapies and biological questions could be addressed in mammals by the application of a molecular switch that would allow physicians and/or investigators to turn individual genes on or off during the lifetime of the organism. We have constructed such a switch, composed of three elements: (i) an inducible promoter that is normally absent from mammalian genomes; (ii) a receptor that, when it is bound to an inducer drug, specifically activates transcription from the inducible promoter; and (iii) inducer drugs, such as RU486, whose pharmacological properties in humans and several mammalian species including mouse have been well studied. The molecular switch is functional in transiently and stably transfected cells. Importantly, both the total output and the induction levels of the reporter gene can be finely tuned, with induction levels of over 100-fold being readily attained. Finally, we demonstrate that the molecular switch can be used to regulate a mouse transgene using a gene therapy paradigm. The specificity of the system suggests that it should be useful in the analysis of gene function in transgenic animals and in the design of strategies for human gene therapy.
Collapse
Affiliation(s)
- J P Delort
- Howard Hughes Medical Institute, University of Utah, Salt Lake City 84112, USA
| | | |
Collapse
|
1928
|
Rowan BG, Ip MM. Differential binding of mutant glucocorticoid receptors to the glucocorticoid response element of the tyrosine aminotransferase gene. J Steroid Biochem Mol Biol 1996; 58:147-62. [PMID: 8809196 DOI: 10.1016/0960-0760(96)00026-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Glucocorticoid receptors (GCRs) in sublines of the mouse P1798 lymphosarcoma that are sensitive (S) or resistant (R) to glucocorticoid-induced cell lysis were examined for their ability to bind to a single glucocorticoid responsive element (GRE). Mobility shift assays detected two specific complexes that were identical in both S and R cellular extracts. Antibodies against the GCR N-terminus supershifted complexes, suggesting that the 97 kDa wild-type GCR (WT-GCR) in S cells, and the variant, 97 kDa non-steroid-binding GCR (NSB-GCR) in R cells were components of both complexes. Sephacryl S300 gel filtration column fractions containing the WT-GCR and NSB-GCR formed complexes with the GRE, while fractions containing a second GCR variant in R cells, the 45 kDa steroid-binding truncated GCR (TR-GCR), did not. Southwestern blotting detected a GRE-binding, 97 kDa protein band in both S and R extracts. A 45 kDa band was not detected. UV crosslinking of protein to DNA revealed protein in the range of 92-120 kDa crosslinked to the GRE in both S and R extracts. No crosslinking was detected at 45 kDa. Strong interaction of the NSB-GCR with GREs and lack of binding of the TR-GCR to single GREs illustrate a complex receptor system in the P1798 lymphosarcoma.
Collapse
Affiliation(s)
- B G Rowan
- Department of Experimental Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | |
Collapse
|
1929
|
Trapp T, Holsboer F. Nuclear orphan receptor as a repressor of glucocorticoid receptor transcriptional activity. J Biol Chem 1996; 271:9879-82. [PMID: 8626619 DOI: 10.1074/jbc.271.17.9879] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Nuclear orphan receptors belong to the superfamily of ligand-activated transcription factors that show a close structural relationship and sequence homology. Ligands and functions of most of the orphan receptors have not yet been identified. The first nuclear orphan receptors that were cloned displayed a high degree of amino acid identity with the human estrogen receptor and were termed estrogen receptor-related (ERR) 1 and 2. In the present study, we show that ERR2 functions as a potent repressor of transcriptional activity mediated by the glucocorticoid receptor (GR). Transient transfection of different cell lines with a steroid-responsive reporter plasmid and receptor expression plasmids revealed that transcriptional activity mediated by GR in response to agonists was strongly suppressed by coexpression of ERR2. The orphan receptor displayed no promoter activity when expressed without GR. The inhibitory activity of ERR2 is cell-specific and also receptor-specific because transactivation mediated by the progesterone receptor is unaffected by ERR2. Our observations provide evidence that the nuclear orphan receptor ERR2 acts as an endogenous modulator of GR transcriptional activity.
Collapse
Affiliation(s)
- T Trapp
- Department of Neuroendocrinology, Max Planck Institute of Psychiatry, Clinical Institute, 80804 Munich, Germany
| | | |
Collapse
|
1930
|
Miyata KS, McCaw SE, Patel HV, Rachubinski RA, Capone JP. The orphan nuclear hormone receptor LXR alpha interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling. J Biol Chem 1996; 271:9189-92. [PMID: 8621574 DOI: 10.1074/jbc.271.16.9189] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The yeast two-hybrid system was used to isolate novel cellular factors that interact with the mouse peroxisome proliferator-activated receptor alpha (PPARalpha). One of the interacting clones isolated encoded LXRalpha, a recently described human orphan nuclear hormone receptor. LXRalpha bound directly to PPARalpha, as well as to the common heterodimerization partner 9-cis-retinoic acid receptor (RXRalpha). LXRalpha did not form a DNA binding complex with PPARalpha on synthetic hormone response elements composed of direct repeats of the TGACCT consensus half-site or on naturally occurring peroxisome proliferator response elements (PPREs) or LXRalpha response elements. However, LXRalpha inhibited binding of PPARalpha/RXRalpha heterodimers to PPREs, and coexpression of LXRalpha in mammalian cells antagonized peroxisome proliferator signaling mediated by PPARalpha/RXRalpha in vivo. These findings identify a novel partner for PPARalpha and suggest that LXRalpha plays a role in modulating PPAR-signaling pathways in the cell.
Collapse
Affiliation(s)
- K S Miyata
- Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
1931
|
Couette B, Fagart J, Jalaguier S, Lombes M, Souque A, Rafestin-Oblin ME. Ligand-induced conformational change in the human mineralocorticoid receptor occurs within its hetero-oligomeric structure. Biochem J 1996; 315 ( Pt 2):421-7. [PMID: 8615809 PMCID: PMC1217212 DOI: 10.1042/bj3150421] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To determine the first steps involved in the mechanism of action of aldosterone and its antagonists, we analysed the ligand-induced structural changes of the human mineralocorticoid receptor (hMR) translated in vitro. Limited chymotrypsin digestion of the receptor generated a 30 kDa fragment. Following binding of a ligand to hMR, the 30 kDa fragment became resistant to chymotrypsin proteolysis, indicating a change in the receptor conformation. Differences in sensitivity to chymotrypsin of the 30 kDa fragment were observed after binding of agonists and antagonists to hMR, suggesting that these two classes of ligands induced different hMR conformations. Several lines of evidence allowed us to identify the 30 kDa fragment as the subregion encompassing the C-terminal part of the hinge region and the ligand-binding domain (LBD) or hMR (hMR 711-984). (1) The 30 kDa fragment is not recognized by FD4, an antibody directed against the N-terminal region of hMR. (2) Aldosterone remains associated with the 30 kDa fragment after chymotrypsin proteolysis of the aldosterone-hMR complex. (3) A truncated hMR, lacking the last 40 C-terminal amino acids (hMR 1-944), yields a 26 kDa proteolytic fragment. In addition, we showed that the unbound and the aldosterone-bound 30 kDa fragment were both associated with heat-shock protein (hsp) 90, indicating that the ligand-induced conformational change takes place within the hetero-oligomeric structure and that the 711-984 region is sufficient for hsp90-MR interaction. We conclude that the ligand-induced conformational change of the receptor is a crucial step in mineralocorticoid action. It occurs within the LBD, precedes the release of hsp90 from the receptor and is dependent upon the agonist/antagonist nature of the ligand.
Collapse
Affiliation(s)
- B Couette
- INSERM U246, Faculté de Médecine Xavier Bichat, Paris, France
| | | | | | | | | | | |
Collapse
|
1932
|
Malik S, Karathanasis SK. TFIIB-directed transcriptional activation by the orphan nuclear receptor hepatocyte nuclear factor 4. Mol Cell Biol 1996; 16:1824-31. [PMID: 8657158 PMCID: PMC231169 DOI: 10.1128/mcb.16.4.1824] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The orphan nuclear receptor hepatocyte nuclear factor 4 (HNF-4) is required for development and maintenance of the liver phenotype. HNF-4 activates several hepatocyte-specific genes, including the gene encoding apolipoprotein AI (apoAI), the major protein component of plasma high-density lipoprotein. The apoAI gene is activated by HNF-4 through a nuclear receptor binding element (site A) located in its liver-specific enhancer. To decipher the mechanism whereby HNF-4 enhances apoAI gene transcription, we have reconstituted its activity in a cell-free system. Functional HNF-4 was purified to homogeneity from a bacterial expression system. In in vitro transcription assays employing nuclear extract from HeLa cells, which do not contain HNF-4, recombinant HNF-4 stimulated transcription from basal promoters linked to site A. Activation by HNF-4 did not exhibit a ligand requirement, but phosphorylation of HNF-4 in the in vitro transcription system was observed. The activation function of HNF-4 was localized to a domain displaying strong homology to the conserved AF-2 region of nuclear receptors. Dissection of the transcription cycle revealed that HNF-4 activated transcription by facilitating assembly of a preinitiation complex intermediate consisting of TBP, the TATA box-binding protein component of TFIID and TFIID, via direct physical interactions with TFIIB. However, recruitment of TFIIB by HNF-4 was not sufficient for activation, since HNF-4 deletion derivatives lacking AF-2 bound TFIIB. On the basis of these results, HNF-4 appears to activate transcription at two distinct levels. The first step involves AF-2-independent recruitment of TFIIB to the promoter complex; the second step is AF-2 dependent and entails entry of preinitiation complex components acting downstream of TFIIB.
Collapse
Affiliation(s)
- S Malik
- Department of Cardiovascular Molecular Biology, Lederle Laboratories, Pearl River, New York 10965, USA
| | | |
Collapse
|
1933
|
Vanacker JM, Corbau R, Adelmant G, Perros M, Laudet V, Rommelaere J. Transactivation of a cellular promoter by the NS1 protein of the parvovirus minute virus of mice through a putative hormone-responsive element. J Virol 1996; 70:2369-77. [PMID: 8642664 PMCID: PMC190079 DOI: 10.1128/jvi.70.4.2369-2377.1996] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The promoter of the thyroid hormone receptor alpha gene (c-erbA-1) is activated by the nonstructural protein 1 (NS1) of parvovirus minute virus of mice (prototype strain [MVMp]) in ras-transformed FREJ4 cells that are permissive for lytic MVMp replication. This stimulation may be related to the sensitivity of host cells to MVMp, as it does not take place in parental FR3T3 cells, which are resistant to the parvovirus killing effect. The analysis of a series of deletion and point mutants of the c-erbA-1 promoter led to the identification of an upstream region that is necessary for NS1-driven transactivation. This sequence harbors a putative hormone-responsive element and is sufficient to render a minimal promoter NS1 inducible in FREJ4 but not in FR3T3 cells, and it is involved in distinct interactions with proteins from the respective cell lines. The NS1-responsive element of the c-erbA-1 promoter bears no homology with sequences that were previously reported to be necessary for NS1 DNA binding and transactivation. Altogether, our data point to a novel, cell-specific mechanism of promoter activation by NS1.
Collapse
Affiliation(s)
- J M Vanacker
- Tumor Virology Unit, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
1934
|
Gleizes-Escala C, Lesca P, Larrieu G, Dupuy J, Pineau T, Galtier P. Effect of exposure of rabbit hepatocytes to sulfur-containing anthelmintics (oxfendazole and fenbendazole) on cytochrome P4501A1 expression. Toxicol In Vitro 1996; 10:129-39. [DOI: 10.1016/0887-2333(95)00117-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/1995] [Indexed: 10/16/2022]
|
1935
|
Wade PA, Jaehning JA. Transcriptional corepression in vitro: a Mot1p-associated form of TATA-binding protein is required for repression by Leu3p. Mol Cell Biol 1996; 16:1641-8. [PMID: 8657139 PMCID: PMC231150 DOI: 10.1128/mcb.16.4.1641] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Signals from transcriptional activators to the general mRNA transcription apparatus are communicated by factors associated with RNA polymerase II or the TATA-binding protein (TBP). Currently, little is known about how gene-specific transcription repressors communicate with RNA polymerase II. We have analyzed the requirements for repression by the saccharomyces cerevisiae Leu3 protein (Leu3p) in a reconstituted transcription system. We have identified a complex form of TBP which is required for communication of the repressing signal. This TFIID-like complex contains a known TBP-associated protein, Mot1p, which has been implicated in the repression of a subset of yeast genes by genetic analysis. Leu3p-dependent repression can be reconstituted with purified Mot1p and recombinant TBP. In addition, a mutation in the Mot1 gene leads to partial derepression of the Leu3p-dependent LEU2 promoter. These in vivo and in vitro observations define a role for Mot1p as a transcriptional corepressor.
Collapse
Affiliation(s)
- P A Wade
- Department of Biochemistry, Biophysics and Genetics, University of Colorado Health Sciences Center, Denver, 80262, USA
| | | |
Collapse
|
1936
|
Trapp T, Holsboer F. Heterodimerization between mineralocorticoid and glucocorticoid receptors increases the functional diversity of corticosteroid action. Trends Pharmacol Sci 1996; 17:145-9. [PMID: 8984741 DOI: 10.1016/0165-6147(96)81590-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Gene regulation by steroids is mediated by the binding of the endogenous or pharmacological ligand to the corresponding nuclear receptor. Ligand-activated steroid receptors usually regulate the expression of responsive genes by binding to common response elements on DNA as homodimers. However, recent findings indicate that mineralocorticoid and glucocorticoid receptors are able to interact by forming heterodimers. In tissues coexpressing both of these corticosteroid receptors, heterodimerization between them may be a hitherto unrecognized modality for the transcriptional regulation of corticosteroid-responsive genes. In this review, Thorsten Trapp and Florian Holsboer discuss the potential impact of this heterodimerization on corticosteriod physiology and pharmacology.
Collapse
Affiliation(s)
- T Trapp
- Clinical Institute, Max Planck Institute of Psychiatry, Munich, Germany
| | | |
Collapse
|
1937
|
Abstract
Glucocorticoids modulate brain function and behaviour through different mechanisms. Although classical effects are mediated through intracellular receptors that modulate gene transcription, recent evidence supports the existence of rapid, nongenomic steroid effects through the neuronal membrane. In this study, we explored possible rapid behavioural effects of corticosterone in the rat, which could provide a model to characterize further the mechanisms involved in rapid corticosteroid nongenomic actions. We found that a corticosterone injection, at doses (2.5 or 5 mg/kg) that mimic plasma concentrations produced by substantial stress, rapidly increases (within 7.5 min of its systemic administration) the locomotor response displayed by rats in a novel environment (activity cage). A lower dose of 1 mg/kg failed to induce this effect. In addition, corticosterone failed to increase locomotion when administered to rats that had been previously exposed to the activity cage. Corticosterone-induced increased locomotion in a novelty situation was not counteracted by either the intracerebroventricular administration of the protein synthesis inhibitor cycloheximide, or by the intracerebroventricular administration of specific antagonists for each type of intracellular corticosteroid receptor, i.e. RU28318, a mineralocorticoid receptor antagonist and RU38486, a glucocorticoid receptor antagonist. Further studies supported the viability of the receptor antagonists to display an anti-corticosteroid action interfering, as previously reported, with the behavioural &winning test. Therefore, the rapid actions of corticosterone in locomotor activity described here, which appear to be nongenomic, might provide a model for future research on the elucidation of the mechanisms involved in steroid-membrane interactions.
Collapse
Affiliation(s)
- C Sandi
- Psychobiology Research Group, Cajal Institute, CSIC, Madrid, Spain
| | | | | |
Collapse
|
1938
|
Brinkmann A, Jenster G, Ris-Stalpers C, van der Korput H, Brüggenwirth H, Boehmer A, Trapman J. Molecular basis of androgen insensitivity. Steroids 1996; 61:172-5. [PMID: 8732995 DOI: 10.1016/0039-128x(96)00008-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Male sexual differentiation and development proceed under direct control of androgens. Androgen action is mediated by the intracellular androgen receptor, which belongs to the superfamily of ligand-dependent transcription factors. In the X-linked androgen insensitivity syndrome, defects in the androgen receptor gene have prevented the normal development of both internal and external male structures in 46, XY individuals. The complete form of androgen insensitivity syndrome is characterized by 46, XY karyotype, external female phenotype, intra-abdominal testes, absence of uterus and ovaries, blindly ending vagina, and gynecomastia. There is also a group of disorders of androgen action that result from partial impairment of androgen receptor function. Clinical indications can be abnormal sexual development of individuals with a predominant male phenotype with severe hypospadias and micropenis or of individuals with a predominantly female phenotype with cliteromegaly, ambiguous genitalia, and gynecomastia. Complete or gross deletions of the androgen receptor gene have not been frequently found in persons with the complete androgen insensitivity syndrome, whereas point mutations at several different sites in exons 2-8 encoding the DNA- and androgen-binding domain have been reported in both partial and complete forms of androgen insensitivity, with a relatively high number of mutations in two clusters in exons 5 and 7. The number of mutations in exon 1 is extremely low, and no mutations have been reported in the hinge region, located between the DNA-binding domain and the ligand-binding domain. The X-linked condition of spinal and bulbar muscle atrophy (Kennedy's disease) is characterized by a progressive motor neuron degeneration associated with signs of androgen insensitivity and infertility. The molecular cause of spinal and bulbar muscle atrophy is an expanded length (> 40 residues) of one of the polyglutamine stretches in the N-terminal domain of the androgen receptor.
Collapse
Affiliation(s)
- A Brinkmann
- Department of Endocrinology and Reproduction, Erasmus University Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
1939
|
Souque A, Fagart J, Couette B, Rafestin-Oblin ME. Sulfhydryl groups are involved in the binding of agonists and antagonists to the human mineralocorticoid receptor. J Steroid Biochem Mol Biol 1996; 57:315-21. [PMID: 8639467 DOI: 10.1016/0960-0760(95)00278-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To investigate the role of sulfhydryl groups in the interaction of agonists and antagonists with the human mineralocorticoid receptor (hMR) the effect of methyl methanethiosulfonate (MMTS) on free and liganded-hMR was examined. hMR was expressed in insect cells (Sf9) using the baculovirus system. Treatment of cytosol with MMTS at 4 degrees C inhibited the binding to hMR of both [3H]aldosterone and [3H]RU26752 (a synthetic aldosterone antagonist). At 4 degrees C, the sensitivity to MMTS of the liganded-hMR complexes was dependent upon the nature of the ligands: agonists (aldosterone, corticosterone and cortisol) rendered the hMR resistant to MMTS, whereas antagonists (progesterone and RU26752) did not protect the receptor against MMTS inactivation. Analysis of the dose- and time-dependent effects of MMTS revealed that the free hMR and the RU26752-hMR complexes displayed a similar sensitivity to MMTS and that MMTS increased the dissociation of RU26752 from the hMR. At 4 degrees C the aldosterone-hMR complexes were not affected by MMTS treatment, whereas at 20 degrees C MMTS increased the dissociation of aldosterone from hMR. This effect was unrelated to the dissociation of hsp90 from hMR, because the sensitivity of the aldosterone-hmR complexes to MMTS remained unchanged after covalent linkage between hsp90 and the receptor. Our results suggest that agonists and antagonists modify the receptor conformation in distinct ways that render cysteine residues of the ligand binding domain more or less accessible to the MMTS action.
Collapse
Affiliation(s)
- A Souque
- INSERM U246, Faculté de Médecine Xavier Bichat, Paris, France
| | | | | | | |
Collapse
|
1940
|
Bush SM, Folta S, Lannigan DA. Use of the yeast one-hybrid system to screen for mutations in the ligand-binding domain of the estrogen receptor. Steroids 1996; 61:102-9. [PMID: 8852826 DOI: 10.1016/0039-128x(95)00200-a] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This report describes a novel yeast one-hybrid system which easily allows for the detection of mutations in the ligand-binding domain of the estrogen receptor. This screen is based on the observation that a fusion protein consisting of the GAL4 DNA-binding domain and the estrogen receptor can interact with a GAL4 upstream activating sequence and induce the expression of an integrated GAL1-lacZ gene only in the presence of estradiol. Various deletion mutants of the estrogen receptor were tested in this assay and activating function 1 which is present in the N-terminus of the estrogen receptor was found to be responsible for the transactivation produced in the assay. To test if the screen could be used to detect random mutants in the ligand-binding domain of the estrogen receptor the region of the human receptor between amino acids 381 to 403 was mutated by oligonucleotide saturation mutagenesis. Two of the mutants generated by this mutagenesis were characterized to demonstrate that the results obtained from the screen in the yeast screen are relevant to mammalian systems. One of the mutants which has a valine at position number 388 instead of a glycine was able to transactivate in both the yeast and a mammalian system. This mutant was a more potent activator of transcription and also appeared to have a higher affinity for [3H]estradiol in vivo than the wild type receptor. The other mutant which was characterized has five amino acid changes from amino acids 390 through 400. This mutant was nonfunctional in the yeast and mammalian transcription assays and did not bind [3H]estradiol in vivo or in vitro.
Collapse
Affiliation(s)
- S M Bush
- Department of Pharmacology, University of Vermont, Burlington 05405, USA
| | | | | |
Collapse
|
1941
|
Power SC, Cereghini S. Positive regulation of the vHNF1 promoter by the orphan receptors COUP-TF1/Ear3 and COUP-TFII/Arp1. Mol Cell Biol 1996; 16:778-91. [PMID: 8622679 PMCID: PMC231058 DOI: 10.1128/mcb.16.3.778] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
vHNF1 (also termed HNF1 beta) is a member of the hepatocyte nuclear fa ctor 1 (HNF1; also termed HNF1 alpha) family of homeodomain-containing transcription factors that interact with a sequence motif found in the regulatory regions of a large number of genes expressed mainly in the liver. It has been suggested that vHNF1 plays a role in early differentiation of specialized epithelia of several endoderm- and mesoderm-derived organs, with HNF1 playing a role in later stages. In support of this idea, expression of vHNF1 but not HNF1 is induced upon treatment of the embryonal carcinoma cell line F9 with retinoic acid. We have cloned and analyzed the vHNF1 promoter to gain a better understanding of the regulation of vHNF1 expression and how it relates to the expression of HNF1. We have identified five sites of DNA-protein interaction within the first 260 bp upstream of the transcription start site, which involve at least three different families of transcription factors. Two sites, a distal DR-1 motif and a proximal octamer motif, are the most important for promoter activity. The DR-1 motif interacts with several members of the steroid hormone receptor superfamily including HNF4, COUP-TFI/Ear3, COUP-TFII/Arp1, and RAR alpha/RXR alpha heterodimers. The vHNF1 promoter is transactivated by COUP-TFI/Ear3 and COUP-TFII/Arp1 and, unlike the HNF1 promoter, is virtually unaffected by HNF4. Interestingly, the proximal octamer site and not the DR-1 site is required for COUP-TFI/Ear3 and COUP-TFII/Arp1 transactivation of the vHNF1 promoter. COUP-TFI/Ear3 does not bind directly to this proximal octamer site. We present evidence of an interaction between COUP-TFI/Ear3 and the octamer-binding proteins in vitro and in the cell, suggesting that COUP-TFI and COUP-TFII activate the vHNF1 promoter via an indirect mechanism.
Collapse
Affiliation(s)
- S C Power
- URA Centre National de la Recherche Scientifique, Institut Pasteur, Paris, France
| | | |
Collapse
|
1942
|
Bevan CL, Brown BB, Davies HR, Evans BA, Hughes IA, Patterson MN. Functional analysis of six androgen receptor mutations identified in patients with partial androgen insensitivity syndrome. Hum Mol Genet 1996; 5:265-73. [PMID: 8824883 DOI: 10.1093/hmg/5.2.265] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Partial androgen insensitivity syndrome (PAIS) is caused by defects in the androgen receptor gene and presents with a wide range of undervirilization phenotypes. We studied the consequences of six androgen receptor ligand-binding domain mutations on receptor function in transfected cells. The mutations, Met742Ile, Met780Ile, Gln798Glu, Arg840Cys, Arg855His and Ile869Met, were identified in PAIS patients with phenotypes representing the full spectrum seen in this condition. In all cases the androgen receptor was found to be defective, suggesting that the mutation is the cause of the clinical phenotype. The Gln798Glu mutation is exceptional in that it did not cause an androgen-binding defect in our system, although the mutant receptor was defective in transactivation assays. This mutation may affect an aspect of binding not tested, or may be part of a functional subdomain of the ligand-binding domain involved in transactivation. Overall we found milder mutations to be associated with milder clinical phenotypes. There is also clear evidence that phenotype is not solely dependent on androgen receptor function. Some of the mutant receptors were able to respond to high doses of androgen in vitro, suggesting that patients carrying these mutations may be the best candidates for androgen therapy. One such mutation is Ile869Met. A patient carrying this mutation has virilized spontaneously at puberty, so in vivo evidence agrees with the experimental result. Thus a more complete understanding of the functional consequences of androgen receptor mutations may provide a more rational basis for gender assignment in PAIS.
Collapse
Affiliation(s)
- C L Bevan
- University Department of Paediatrics, University of Cambridge, UK
| | | | | | | | | | | |
Collapse
|
1943
|
Borrás M, Laios I, el Khissiin A, Seo HS, Lempereur F, Legros N, Leclercq G. Estrogenic and antiestrogenic regulation of the half-life of covalently labeled estrogen receptor in MCF-7 breast cancer cells. J Steroid Biochem Mol Biol 1996; 57:203-13. [PMID: 8645630 DOI: 10.1016/0960-0760(95)00272-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Effect of estrogens and antiestrogens (AEs) on estrogen receptor (ER) half-life was analyzed in MCF-7 cells by assessing its progressive disappearance after covalent labeling in situ with [3H]tamoxifen aziridine ([3H]TAZ). Cells were incubated for 1 h with 20 nM [3H]TAZ either in the absence or presence of a 500-fold excess of unlabeled estradiol (E2) (non-specific binding). The entire ER population was labeled by this method as established by subsequent incubation of the cells with [125I]E2. [3H]TAZ labeled cells were maintained in culture for additional 5 h in the absence (control) or presence of increasing amounts (0.1 nM - 1 microM) of either a given estrogen (E2, estrone, diethylstilbestrol, bisphenol), a pure AE (RU 58 668, ICI 164 384) or an AE with residual estrogenic activity (RU 39 411, 4-hydroxytamoxifen, keoxifene). The progressive disappearance of nuclear and cytosolic [3H]TAZ-ER complex during 5 h incubation were assessed by their immunoprecipitation with anti-ER monoclonal antibody (H 222) followed by scintillation counting or SDS-PAGE and fluorography. Fading of labeled receptors was extremely slow (approximately 10% loss after 6 h) in absence of any hormone/antihormone indicating a long half-life of the [3H]TAZ-ER complex. Addition of estrogens as well as pure AEs led to a dramatic reduction of the half-life while AEs with residual estrogenic activity were extremely less efficient in this regard providing an explanation for the ability of latter compounds to up-regulate the receptor since they do not affect ER mRNA synthesis and stability. Receptor disappearance induced by estrogens was closely related to their binding affinity for ER. Newly synthesized ER emerged during the treatment with hormones or antihormones seems to be implicated in the phenomenon since [3H]TAZ was covalently bound and could, therefore, not be displaced by these compounds. Induction of synthesis of a short half-life peptide(s) with degradative activity was demonstrated by addition of cycloheximide or puromycine (both at 50 microM) which completely blocked ER disappearance. The fact that no cleavage products of ER were detected by SDS-PAGE suggested a lysosomial hydrolysis. Hence, hormonal modulation of only a part of ERs may down-regulate their total population until it reaches the steady-state level.
Collapse
Affiliation(s)
- M Borrás
- Laboratoire J-C. Heuson de Cancérologie Mammaire, Service de Médecine Interne, Institut Jules Bordet, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
1944
|
Liu M, Lee MH, Cohen M, Bommakanti M, Freedman LP. Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the induced differentiation of the myelomonocytic cell line U937. Genes Dev 1996; 10:142-53. [PMID: 8566748 DOI: 10.1101/gad.10.2.142] [Citation(s) in RCA: 714] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The hormonal form of vitamin D, 1,25-dihydroxyvitamin D3, acting through its cognate nuclear receptor (vitamin D3 receptor, VDR) will induce myeloid leukemic cell lines to terminally differentiate into monocytes/macrophages. Because VDR acts by transcriptionally regulating responsive genes in a ligand-dependent manner, we sought target genes of the receptor that initiate, the differentiation process in response to ligand. We screened a cDNA library prepared from the myelomonocytic U937 cell line with probes generated from either 1,25-dihydroxyvitamin D3-treated or untreated cells. We report here that a candidate clone that hybridized differentially is the Cdk inhibitor p21WAF1, CIP1. Furthermore, we show that p21 is transcriptionally induced by 1,25-dihydroxyvitamin D3 in a VDR-dependent, but not p53-dependent, manner, and we identify a functional vitamin D response element in the p21 promoter. Transient overexpression of p21 and/or the related Cdk inhibitor p27 in U937 cells in the absence of 1,25-dihydroxyvitamin D3 results in the cell-surface expression of monocyte/macrophage-specific markers, suggesting that ligand-modulated transcriptional induction of the p21 gene facilitates the induced differentiation of this monoblastic cell line. We believe that this is the first report demonstrating that the ectopic overexpression of a Cdk inhibitor such as p21 or p27 directly leads to a terminal differentiation program.
Collapse
Affiliation(s)
- M Liu
- Cell Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, Cornell University, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
1945
|
Woo PL, Cha HH, Singer KL, Firestone GL. Antagonistic regulation of tight junction dynamics by glucocorticoids and transforming growth factor-beta in mouse mammary epithelial cells. J Biol Chem 1996; 271:404-12. [PMID: 8550596 DOI: 10.1074/jbc.271.1.404] [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: 01/31/2023] Open
Abstract
The synthetic glucocorticoid, dexamethasone, stimulated the transepithelial electrical resistance and suppressed the DNA synthesis of 31EG4 nontransformed mouse mammary epithelial cells. The addition of transforming growth factor-beta 1 (TGF-beta) to mammary cells simultaneously with or up to 24 h after dexamethasone treatment prevented the steroid induction of transepithelial electrical resistance and stimulated the incorporation of [3H]thymidine. However, the TGF-beta inhibition of tight junction formation did not require de novo DNA synthesis. Confocal microscopy revealed that the organized immunostaining pattern of the tight junction protein, ZO-1, and F-actin at the cell periphery was disrupted by TGF-beta, resulting in disorganized and diffuse staining patterns throughout the cell. Western blot analysis demonstrated that TGF-beta did not alter the protein levels of ZO-1. In contrast to cells not treated or pretreated with steroid for up to 24 h, TGF-beta had no effect on cells pretreated with dexamethasone for 48 h. Transfection of chimeric reporter genes containing promoters responsive to either glucocorticoid or TGF-beta demonstrated that the mutual antagonism of tight junction dynamics by dexamethasone and TGF-beta occurs in the presence of intact signaling pathways. Taken together, our results establish for the first time that glucocorticoids and TGF-beta can antagonistically regulate tight junction formation in a nontransformed mammary cell line.
Collapse
Affiliation(s)
- P L Woo
- Department of Molecular and Cell Biology, University of California at Berkeley 94720, USA
| | | | | | | |
Collapse
|
1946
|
P450 Cytochromes and Hormonal Regulation: An Overview. ADVANCES IN MOLECULAR AND CELL BIOLOGY 1996. [DOI: 10.1016/s1569-2558(08)60338-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
1947
|
Pfeffer U, Fecarotta E, Arena G, Forlani A, Vidali G. Alternative splicing of the estrogen receptor primary transcript normally occurs in estrogen receptor positive tissues and cell lines. J Steroid Biochem Mol Biol 1996; 56:99-105. [PMID: 8603053 DOI: 10.1016/0960-0760(95)00227-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Several laboratories have described estrogen receptor mRNA variants created by skipping internal exons. Some of the putative proteins encoded for by these variants have been functionally characterized by transfection analyses. The variant lacking exon 5 would lead, if translated, to a truncated receptor which shows dominant positive transactivation activity in the absence of hormone. It has been postulated that the variant could account for anti-estrogen resistant tumor growth and for expression of the progesterone receptor in estrogen negative tumors. In order to understand the possible role this and other variants may have in the tumorigenesis of mammary tissue we have carried out a thorough analysis of variants expressed in a tumor cell line (MCF-7), in a tumor sample and in a sample of normal breast tissue derived from mammary reduction surgery. We performed rt-PCR analyses followed by hybridization with exon specific oligonucleotide probes. By these means we have detected nine different variants co-expressed in MCF-7 cells and at least the major variants were equally expressed in normal and neoplastic breast tissue. The same is true for the variant lacking exon 5 which, however, resulted to be a variant of low expression in the three samples analyzed. Variant formation appeared to be restricted to the estrogen receptor messenger since several other members of the superfamily of nuclear receptors did not show variant formation. We also have analyzed the effect of the most abundantly expressed variant, the exon 4 lacking variant, on normal estrogen receptor function, on the growth and on the response to estradiol and to tamoxifen of MCF-7 cells. Although over-expressed at high levels this variant has, if any, only marginal effects on the expression of endogenous estrogen regulated genes and on growth and response to the hormone and its antagonist. Although the lack of function of this variant cannot be extrapolated to other variants, their involvement in tumor formation appears rather unlikely since they are also expressed in normal tissue and the single variant is expressed in addition to many others, some of which might have opposing effects. Variant formation is, however, specific for the estrogen receptor and apparently regulated with tissue specificity as our expression analysis in normal mouse tissues shows. Therefore the variants probably have a physiological significance yet to be discovered.
Collapse
Affiliation(s)
- U Pfeffer
- Laboratory of Molecular Biology, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | |
Collapse
|
1948
|
Epstein E, Silver J, Almogi G, Livni N, Naveh-Many T. Parathyroid hormone mRNA levels are increased by progestins and vary during rat estrous cycle. THE AMERICAN JOURNAL OF PHYSIOLOGY 1996; 270:E158-63. [PMID: 8772488 DOI: 10.1152/ajpendo.1996.270.1.e158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Estrogen increases parathyroid hormone (PTH) mRNA levels in vivo in ovariectomized rats. We now show that the 19-norprogestin R-5020 given to weanling rats or mature ovariectomized rats led to a twofold increase in thyroparathyroid PTH mRNA levels. This increase in PTH mRNA occurred at 24 and 48 h after progesterone but not at 72 h. There were no changes in serum calcium. In vitro, in primary cultures of bovine parathyroid cells, progesterone increased PTH mRNA levels threefold at 10(-8) M and twofold at 10(-9) M after 24 h. Progesterone receptor (PR) mRNA was demonstrated in rat parathyroid tissue by in situ hybridization and in human parathyroid adenoma by immunohisto-chemistry. Changes in PTH mRNA levels during the rat estrous cycle were also studied. At proestrus and estrus PTH mRNA levels were increased significantly by three- and fourfold compared with diestrus. Our results confirm that the parathyroid gland is a target organ for the ovarian sex steroids estrogen and progesterone and are of physiological relevance as shown by the changes during estrus.
Collapse
Affiliation(s)
- E Epstein
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
1949
|
Graham JD, Yeates C, Balleine RL, Harvey SS, Milliken JS, Bilous AM, Clarke CL. Progesterone receptor A and B protein expression in human breast cancer. J Steroid Biochem Mol Biol 1996; 56:93-8. [PMID: 8603052 DOI: 10.1016/0960-0760(95)00226-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The human progesterone receptor (PR) is a ligand-activated nuclear transcription factor which mediates progesterone action in target tissues. Two PR proteins, PR A (81-83 kDa) and PR B (116-120 kDa), have been described and different physiological activities ascribed to each on the basis of in vitro studies, suggesting that their ratio of expression may control progesterone responsiveness in target cells. Presence of PR in breast tumors is an important indicator of likely responsiveness to endocrine agents. However, the relative expression of PR A and B in breast cancer has not been described and its clinical significance has not been addressed. We have examined the expression of PR A and B in PR-positive breast tumors and found that while in most tumors PR A and B were expressed in similar amounts there was a broad overall distribution of PR A:B ratio which deviated significantly from a normal log distribution with tumors containing a PR A:B ration greater than 4 being over-represented in the group. Linear regression analysis revealed that high PR A:B ratios, in general, derived from a low concentration of PR B rather than high expression of PR A. PR A:B protein ratios were not correlated with the age of the patient or with total PR concentration. A third PR protein band (PR 78 kDa) was detected which comprised greater than 20% of total PR protein in a quarter of the tumor samples examined. The characteristics of tumors containing PR 78 kDa were not different from the overall group. In summary, in PR-positive breast tumors the ratio of expression of PR A and B proteins is close to unity as is seen in a number of other progestin target tissues. However, a significant proportion of tumors expressed very low levels of PR B and a consequently high PR A:B ration. Although the clinical consequence of this observation is not known, the in vitro findings that PR A may act as a repressor for PR B suggests that tumors containing primarily PR A may identify a subset of patients with low or aberrant response to endocrine agents.
Collapse
Affiliation(s)
- J D Graham
- Department of Medical Oncology, University of Sydney Westmead Hospital, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
1950
|
Lydon JP, DeMayo FJ, Conneely OM, O'Malley BW. Reproductive phenotpes of the progesterone receptor null mutant mouse. J Steroid Biochem Mol Biol 1996; 56:67-77. [PMID: 8603049 DOI: 10.1016/0960-0760(95)00254-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although progesterone has been traditionally associated with the establishment and maintenance of mammalian pregnancy, a number of studies have implicated physiological roles of this steroid hormone in other reproductive events. At present most of the downstream molecular and cellular mechanisms by which progesterone exerts its effects are unclear; however, the progesterone signal is known to be mediated initially by the progesterone receptor (PR), a member of the nuclear receptor superfamily of transcription factors. In most tissues studied, the PR is induced by ovarian estrogen via the estrogen receptor (ER), thereby implying that many of the observed reproductive physiological responses attributed to PR could conceivably be due to the combined effects of progesterone and estrogen. Therefore, to define clearly the distinct roles of progesterone and estrogen in vivo and to understand better progesterone function in a physiological context, we recently have generated a novel mouse strain in which both forms of the PR were ablated using gene targeting/embryonic stem cell techniques. Surprisingly, both male and female embryos, homozygous for the PR null mutation, developed to adulthood at the normal Mendelian frequency with no deviation in the sex ratio. Although developmental defects have yet to be detected in the adult male PR homozygote, extensive reproductive abnormalities were observed in the female. The reproductive phenotypes consisted of an inability to ovulate, uterine hyperplasia and inflammation, severely limited mammary gland development and an impairment in the induction of a sexual behavioral response. Collectively, these results provide direct in vivo evidence for progesterone's role as a pleiotropic coordinator of diverse reproductive events that together ensure female fertility. Finally, we believe that this animal model will be an invaluable tool in exploring the effects of progesterone in physiological systems other than reproduction and may, in the future, help to redefine progesterone not just as a sex steroid hormone but also as a key regulator of diverse physiological processes.
Collapse
Affiliation(s)
- J P Lydon
- Departement of Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|