4851
|
Di Matteo G, Salerno M, Guarguaglini G, Di Fiore B, Palitti F, Lavia P. Interactions with single-stranded and double-stranded DNA-binding factors and alternative promoter conformation upon transcriptional activation of the Htf9-a/RanBP1 and Htf9-c genes. J Biol Chem 1998; 273:495-505. [PMID: 9417108 DOI: 10.1074/jbc.273.1.495] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The murine Htf9-a/RanBP1 and Htf9-c genes are divergently transcribed from a shared TATA-less promoter. Transcription of both genes is initiated on complementary DNA strands and is controlled by cell cycle-dependent mechanisms. The bidirectional promoter harbors a genomic footprint flanking the major transcription start site of both genes. Transient promoter assays showed that the footprinted element is important for transcription of both genes. Protein-binding experiments and antibody assays indicated that members of the retinoid X receptor family interact with the double-stranded site. In addition, distinct factors interact with single DNA strands of the element. Double-stranded binding factors were highly expressed in liver cells, in which neither gene is transcribed, while single-stranded binding proteins were abundant in cycling cells, in which transcription of both genes is efficient. In vivo S1 analysis of the promoter depicted an S1-sensitive organization in cells in which transcription of both genes is active; S1 sensitivity was not detected in conditions of transcriptional repression. Thus, the same element is a target for either retinoid X receptor factors, or for single-stranded binding proteins, and form distinct complexes in different cellular conditions depending on the DNA conformation in the binding site.
Collapse
Affiliation(s)
- G Di Matteo
- CNR Centre of Evolutionary Genetics, c/o Department of Genetics and Molecular Biology, University "La Sapienza," Rome 00185, Italy
| | | | | | | | | | | |
Collapse
|
4852
|
Demangel C, Rouyre S, Alzari PM, Nato F, Longacre S, Lafaye P, Mazie JC. Phage-displayed mimotopes elicit monoclonal antibodies specific for a malaria vaccine candidate. Biol Chem 1998; 379:65-70. [PMID: 9504719 DOI: 10.1515/bchm.1998.379.1.65] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The phage-displayed peptide CGRVCLRC (C15) has been isolated from a random library by affinity screening with the D14-3 monoclonal antibody, which was raised to the 42 kDa C-terminal fragment of the major merozoite surface protein 1 of Plasmodium vivax (Pv42). In order to investigate the use of such mimotopes as possible vaccine components, we studied the antibody response in Biozzi mice immunized with C15. High titers of antibodies cross-reacting with Pv42 were generated and the IC50 of all immune sera were in the 5 x 10(-9) M range. Two monoclonal antibodies that specifically bind the Pv42 fragment were isolated. Although these mAbs had a lower affinity for Pv42 when compared to D14-3, they reproduced the cross-reactivity of D14-3 with the equivalent protein in P. cynomolgi, a close relative of P. vivax. DNA sequence analysis showed similarities between the germline genes and the canonical CDR conformations of all three antibodies, but molecular modeling failed to reveal common structural features of their paratopes that could account for their cross-reacting patterns. These data demonstrate that mimotopes selected from random repertoires do not necessarily represent structural equivalents of the original antigen but provide functional images that could replace it for vaccine development.
Collapse
Affiliation(s)
- C Demangel
- Hybridolab, Institut Pasteur, Paris, France
| | | | | | | | | | | | | |
Collapse
|
4853
|
Lu D, Yang H, Shaw G, Raizada MK. Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons. Endocrinology 1998; 139:365-75. [PMID: 9421435 DOI: 10.1210/endo.139.1.5679] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Angiotensin II (Ang II) interaction with the neuronal AT1 receptor results in a chronic stimulation of neuromodulation that involves the expression of norepinephrine transporter (NET) and tyrosine hydroxylase (TH). In view of this unique property and the presence of putative nuclear localization signal (NLS) consensus sequence in the AT1 receptor, this study was conducted to investigate the hypothesis that Ang II would induce nuclear sequestration of this G protein-coupled receptor and that the sequestration may have implications on Ang II-induced expression of NET and TH genes. Incubation of neuronal cultures with Ang II caused a time- and dose-dependent increase in the levels of AT1 receptor immunoreactivity in the nucleus. A 6.7-fold increase was observed with 100 nM Ang II, in 15 min, that was blocked by losartan, an AT1 receptor-specific antagonist. Ang II-induced nuclear sequestration was specific for AT1 receptor, because Ang II failed to produce a similar effect on neuronal AT2 receptors. The presence of the putative NLS sequence in the cytoplasmic tail of the AT1 receptor seems to be the key in nuclear targeting because: 1) nuclear targeting was attenuated by a peptide of the AT1 receptor that contained the putative NLS sequence; and 2) Ang II failed to cause nuclear translocation of the AT2 receptor, which does not contain the putative NLS. Ang II also caused a time- and dose-dependent stimulation of P62 phosphorylation, a glycoprotein of the nuclear pore complex. A 6-fold stimulation of phosphorylation was observed with 100 nM Ang II, in 15 min, that was completely blocked by losartan and not by PD123,319, an AT2 receptor specific antagonist. Preloading of neurons with p62-pep (a peptide containing consenses of mitogen-activated protein kinase in p62) resulted in a loss of Ang II-induced p62 phosphorylation and stimulation of NET and TH messenger RNA levels. In conclusion, these data demonstrate that Ang II induces nuclear sequestration of AT1 receptor involving NLS in the AT1 receptor and p62 of the nuclear pore complex in brain neurons. A possible role of such a nuclear targeting of the AT1 receptor on chronic neuromodulatory actions of Ang II has been discussed.
Collapse
Affiliation(s)
- D Lu
- Department of Physiology, University of Florida, College of Medicine, Gainesville 32610, USA
| | | | | | | |
Collapse
|
4854
|
OHKURA N, MARUYAMA K, TSUKADA T, HOSONO T, YAMAGUCHI K. The NGFI-B Family: Orphan Nuclear Receptors of the Steroid/Thyroid Receptor Superfamily. J Reprod Dev 1998. [DOI: 10.1262/jrd.44.321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Naganari OHKURA
- Growth Factor Division, National Cancer Center Research Institute,5-1-1 Tsukiji,Chuo-ku, Tokyo 104-0045, Japan
| | - Kouji MARUYAMA
- Growth Factor Division, National Cancer Center Research Institute,5-1-1 Tsukiji,Chuo-ku, Tokyo 104-0045, Japan
| | - Toshihiko TSUKADA
- Growth Factor Division, National Cancer Center Research Institute,5-1-1 Tsukiji,Chuo-ku, Tokyo 104-0045, Japan
| | - Tetsuji HOSONO
- Growth Factor Division, National Cancer Center Research Institute,5-1-1 Tsukiji,Chuo-ku, Tokyo 104-0045, Japan
| | - Ken YAMAGUCHI
- Growth Factor Division, National Cancer Center Research Institute,5-1-1 Tsukiji,Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
4855
|
Brüggenwirth HT, Boehmer AL, Lobaccaro JM, Chiche L, Sultan C, Trapman J, Brinkmann AO. Substitution of Ala564 in the first zinc cluster of the deoxyribonucleic acid (DNA)-binding domain of the androgen receptor by Asp, Asn, or Leu exerts differential effects on DNA binding. Endocrinology 1998; 139:103-10. [PMID: 9421404 DOI: 10.1210/endo.139.1.5696] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the androgen receptor of a patient with androgen insensitivity, the alanine residue at position 564 in the first zinc cluster of the DNA-binding domain was substituted by aspartic acid. In other members of the steroid receptor family, either valine or alanine is present at the corresponding position, suggesting the importance of a neutral amino acid residue at this site. The mutant receptor was transcriptionally inactive, which corresponded to the absence of specific DNA binding in gel retardation assays, and its inactivity in a promoter interference assay. Two other receptor mutants with a mutation at this same position were created to study the role of position 564 in the human androgen receptor on DNA binding in more detail. Introduction of asparagine at position 564 resulted in transcription activation of a mouse mammary tumor virus promoter, although at a lower level compared with the wild-type receptor. Transcription activation of an (ARE)2-TATA promoter was low, and binding to different hormone response elements could not be visualized. The receptor with a leucine residue at position 564 was as active as the wild-type receptor on a mouse mammary tumor virus promoter and an (ARE)2-TATA promoter, but interacted differentially with several hormone response elements in a gel retardation assay. The results of the transcription activation and DNA binding studies could partially be predicted from three-dimensional modeling data. The phenotype of the patient was explained by the negative charge, introduced at position 564.
Collapse
Affiliation(s)
- H T Brüggenwirth
- Department of Endocrinology & Reproduction, Erasmus University, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
4856
|
Dussault I, Fawcett D, Matthyssen A, Bader JA, Giguère V. Orphan nuclear receptor ROR alpha-deficient mice display the cerebellar defects of staggerer. Mech Dev 1998; 70:147-53. [PMID: 9510031 DOI: 10.1016/s0925-4773(97)00187-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It has recently been shown that the neurological mutant mouse staggerer (sg) harbors a deletion within the Rora gene that encodes the orphan nuclear receptor ROR alpha. This deletion removes an exon encoding part of the ligand binding domain of the putative receptor, generating an ROR alpha truncated protein (ROR alpha(sg)). It is unknown whether sg acts as a null or highly hypomorphic allele. To address this question, we have generated a null mutation of Rora by targeted disruption of its DNA binding domain in ES cells. The Rora-/- mice are viable but display tremor, body imbalance, small size and die between 3-4 weeks, similar to the sg mouse. Histological examination of the cerebellum of Rora-/- and sg mice showed similar defects, including small size and fewer ectopically localized Purkinje cells. Northern blot analysis of cerebellar RNA showed that ROR alpha transcripts are still expressed in the Rora-/- and sg mutants, although with altered mobilities. However, the cerebellum of the Rora-/- mutant does not express the ROR alpha protein. Attempts to complement the defect of the Rora-/- with sg failed, demonstrating conclusively that the sg defects are caused by the absence of functional ROR alpha.
Collapse
MESH Headings
- Alleles
- Animals
- Cerebellum/metabolism
- Cerebellum/pathology
- Female
- Gene Targeting
- Genetic Complementation Test
- Genetic Vectors
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout/anatomy & histology
- Mice, Knockout/genetics
- Mice, Knockout/metabolism
- Mice, Neurologic Mutants/anatomy & histology
- Mice, Neurologic Mutants/genetics
- Mice, Neurologic Mutants/metabolism
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1
- Phenotype
- Receptors, Cytoplasmic and Nuclear/deficiency
- Receptors, Cytoplasmic and Nuclear/genetics
- Sequence Deletion
- Trans-Activators/deficiency
- Trans-Activators/genetics
Collapse
Affiliation(s)
- I Dussault
- Molecular Oncology Group, Royal Victoria Hospital, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
4857
|
Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998; 391:82-6. [PMID: 9422509 DOI: 10.1038/34184] [Citation(s) in RCA: 1622] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a member of the nuclear receptor family of transcription factors, a large and diverse group of proteins that mediate ligand-dependent transcriptional activation and repression. Expression of PPAR-gamma is an early and pivotal event in the differentiation of adipocytes. Several agents that promote differentiation of fibroblast lines into adipocytes have been shown to be PPAR-gamma agonists, including several prostanoids, of which 15-deoxy-delta-prostaglandin J2 is the most potent, as well as members of a new class of oral antidiabetic agents, the thiazolidinediones, and a variety of non-steroidal anti-inflammatory drugs (NSAIDs). Here we show that PPAR-gamma agonists suppress monocyte elaboration of inflammatory cytokines at agonist concentrations similar to those found to be effective for the promotion of adipogenesis. Inhibition of cytokine production may help to explain the incremental therapeutic benefit of NSAIDs observed in the treatment of rheumatoid arthritis at plasma drug concentrations substantially higher than are required to inhibit prostaglandin G/H synthase (cyclooxygenase).
Collapse
Affiliation(s)
- C Jiang
- Department of Molecular Biology, Massachusetts General Hospital, Boston 02114, USA
| | | | | |
Collapse
|
4858
|
|
4859
|
Koivisto P, Kolmer M, Visakorpi T, Kallioniemi OP. Androgen receptor gene and hormonal therapy failure of prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 152:1-9. [PMID: 9422516 PMCID: PMC1858130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Androgen receptor (AR) is a nuclear transcription factor that binds male sex steroids and mediates the biological effects of these hormones to the target cells, such as the epithelial cells of the prostate gland, by activating transcription of androgen-dependent genes. Withdrawal of androgens or the peripheral blockade of androgen action remain the critical therapeutic options for the treatment of advanced prostate cancer. However, after initial regression, many prostate cancers become hormone refractory and progress further with eventual fatal outcome. Understanding the mechanisms of tumor progression and endocrine therapy failure is an important goal. A large number of different molecular mechanisms may be responsible for development of hormone-refractory recurrent tumors. Many of these involve the AR gene and its complex downstream signaling pathways. The role of AR mutations and altered transactivational properties of the receptor have received the most attention as causative factors for progression. However, other mechanisms, such as AR gene amplification and overexpression or increased local bioconversion of androgens, may contribute to the development of progression by mechanisms that involve androgen-dependent cell growth. Here we review the role of the AR gene and its putative downstream effector pathways during human prostate cancer progression and endocrine therapy failure.
Collapse
Affiliation(s)
- P Koivisto
- Laboratory of Cancer Genetics, Tampere University Hospital and Institute of Medical Technology, University of Tampere, Finland
| | | | | | | |
Collapse
|
4860
|
Schneider-Hirsch S, Bauer UM, Heiermann R, Rentrop M, Maelicke A. Cloning of the human NCNF gene. J Recept Signal Transduct Res 1998; 18:1-13. [PMID: 9493564 DOI: 10.3109/10799899809039161] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have cloned from a cDNA library of human testis tissue the human homologue to the mouse nuclear orphan receptor NCNF (neuronal cell nuclear factor). The open reading frame encodes a protein of 480 amino acids, the sequence of which (EMBL accession no. X99975) is 98.3% identical to the mouse homologue. Northern blot analysis of adult human tissues revealed a broad pattern of tissue expression. Similar to NCNF expression in mouse testis, two transcript forms of the single copy gene are expressed in human tissues. The two transcript forms which differ only in their 3'UTR, result in human from differential polyadenylation, in mouse from alternative splicing. Based on the high level of sequence identity of human and murine NCNF, it is likely that also the human nuclear receptor is involved in the control of neurogenesis and gametogenesis.
Collapse
Affiliation(s)
- S Schneider-Hirsch
- Institute of Physiological Chemistry and Pathobiochemistry, Johannes Gutenberg-University Medical School, Mainz, Germany
| | | | | | | | | |
Collapse
|
4861
|
Yamano K, Miwa S. Differential gene expression of thyroid hormone receptor alpha and beta in fish development. Gen Comp Endocrinol 1998; 109:75-85. [PMID: 9446725 DOI: 10.1006/gcen.1997.7011] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To understand the mechanisms of flounder metamorphosis, which is controlled by thyroid hormone, gene expression of the thyroid hormone receptors (TR alpha A, TR alpha B, TR beta 1, and TR beta 2) was studied in developing flounder larvae, using quantitative reverse-transcriptase polymerase chain reaction and in situ hybridization. TR gene transcripts were found at very low levels in fertilized flounder eggs. Substantial amounts of TR mRNAs were present in premetamorphic larvae, except for TR alpha B mRNA, which was low throughout larval development. TR alpha A gene transcripts increased rapidly in metamorphic climax and decreased rapidly postclimax. The expression level of TR beta s increased in climax, reached its peak postclimax, and remained high in metamorphosed juveniles. In situ hybridization confirmed the decrease in TR alpha transcripts in most tissues postclimax and further revealed the ubiquitous expression of TR genes and distinct tissue specificity of alpha and beta subtypes in the overall fish body. These results suggest that thyroid hormone exerts effects directly on each tissue during fish metamorphosis and that gene expression of TR subtypes is differentially regulated both temporally and regionally. Thus, the results suggest that the development of each tissue of the flounder by thyroid hormone is further controlled at the receptor level by the differential expression of TRs.
Collapse
Affiliation(s)
- K Yamano
- Inland Station, National Research Institute of Aquaculture, Mie, Japan
| | | |
Collapse
|
4862
|
Dhadialla TS, Carlson GR, Le DP. New insecticides with ecdysteroidal and juvenile hormone activity. ANNUAL REVIEW OF ENTOMOLOGY 1998; 43:545-569. [PMID: 9444757 DOI: 10.1146/annurev.ento.43.1.545] [Citation(s) in RCA: 423] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Agrochemical research over the last two decades has resulted in the discovery of chemically novel insecticides that mimic the action of the two insect growth and developmental hormones, the steroidal 20-hydroxyecdysone (20E) and the sesquiterpenoid juvenile hormone (JH). Bisacylhydrazines are non-steroidal agonists of 20E and exhibit their insecticidal activity via interaction with the ecdysteroid receptor proteins. Interestingly, two of the bisacylhydrazine (tebufenozide and RH-2485) insecticides are very selectively toxic to lepidopteran pests. These insecticides are safe to beneficial insects and have a benign ecotoxicological profile. Aromatic non-terpenoidal insecticides (fenoxycarb and pyriproxyfen) mimic the action of JHs. However, like the JHs, their exact mode of action is not well understood. These insecticides are toxic to a broad spectrum of insects during their embryonic, last larval, or reproductive stages. The insecticidal, ecotoxicological properties and the mode of action of the two groups of insecticides are reviewed in this article.
Collapse
Affiliation(s)
- T S Dhadialla
- Rohm and Haas Company, Insecticide Research Laboratories, Spring House, Pennsylvania 19477, USA.
| | | | | |
Collapse
|
4863
|
Paolucci M, Callard IP. Characterization of progesterone-binding moieties in the little skate Raja erinacea. Gen Comp Endocrinol 1998; 109:106-18. [PMID: 9446728 DOI: 10.1006/gcen.1997.7012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this study we report evidence of a [3H]progesterone-binding moiety in the liver and oviduct of the little skate Raja erinacea. It is characterized by high affinity, low capacity and DNA-cellulose-binding activity. Furthermore Western blot analysis revealed that monoclonal antibodies against the chicken progesterone receptor (PR) subunits A and B cross-reacted with a 110-kDa band in the liver and a 80-kDa band in the oviduct. When analyzed by DEAE-Sepharose ion-exchange column chromatography, [3H]progesterone-binding molecules resolved into two peaks, one nonadherent and one adherent to the column. The liver adherent peak eluted in a linear gradient at a NaCl concentration of about 0.07 M and resolved on Western blot as a single band of a 110 kDa. The oviduct adherent peak eluted at about 0.14 M NaCl and resolved on Western blot as a single band of 80 kDa. Competition studies showed that the progesterone-binding moiety in the cytosol was specific for progesterone. On the contrary, the nuclear component is not specific for progesterone; it also binds testosterone and estradiol 17 beta in the oviduct, and progesterone, testosterone, dihydrotestosterone, estradiol 17 beta, mibolerone, and R5020 in the liver. The [3H]progesterone-binding activity was monitored in both liver and oviduct of females in different reproductive stages. Females were separated into three groups; laying, nonlaying, and immature. [3H]Progesterone-binding activity levels were higher in the liver of immature than of nonlaying skates, and it was undetectable in laying skates. [3H]Progesterone binding was higher in the oviduct of laying and nonlaying skates than of immature skates. This PR-binding moiety has many characteristics of a true receptor: high affinity, low capacity, binds to DNA, and cross-reacts with antibodies against chicken PR. However, while the cytosolic form of this progesterone-binding component was quite specific for P, nuclear extracted material was nonspecific. If these progesterone-binding components are homologous with the PR A and PR B forms of other vertebrates, as we believe, it is clear that there are species differences that probably relate to phylogenetic level and physiology of the organism.
Collapse
Affiliation(s)
- M Paolucci
- Department of Biology, Boston University, Massachusetts 02215, USA
| | | |
Collapse
|
4864
|
Peet DJ, Doyle DF, Corey DR, Mangelsdorf DJ. Engineering novel specificities for ligand-activated transcription in the nuclear hormone receptor RXR. CHEMISTRY & BIOLOGY 1998; 5:13-21. [PMID: 9479476 DOI: 10.1016/s1074-5521(98)90083-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The retinoid X receptor (RXR) activates transcription of target genes in response to its natural ligand, 9-cis retinoic acid (9cRA), and a number of RXR-specific synthetic ligands. To discover the potential for engineering nuclear receptors for activation of transcription by novel ligands, we used structure-based mutagenesis to change the ligand specificity of RXR. RESULTS By making substitutions at only two positions (Phe313 and Leu436) we engineered two new classes of RXR proteins that had altered ligand specificities. The first class exhibits decreased activation by 9cRA and increased activation by synthetic ligands. The second class continues to be activated by 9cRA but no longer responds to synthetic ligands. The magnitude of the change in specificity that can be accomplished is greater than 280-fold. CONCLUSIONS These results confirm that Phe313 and Leu436 are crucial determinants of ligand specificity for RXR and demonstrate that nuclear receptors are exceptionally promising protein scaffolds for the introduction of novel ligand specificities through structure-based protein engineering.
Collapse
Affiliation(s)
- D J Peet
- Howard Hughes Medical Institute, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas 75235-9050, USA
| | | | | | | |
Collapse
|
4865
|
Takeuchi A, Reddy GS, Kobayashi T, Okano T, Park J, Sharma S. Nuclear Factor of Activated T Cells (NFAT) as a Molecular Target for 1α,25-Dihydroxyvitamin D3-Mediated Effects. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.1.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abstract
The molecular basis of the immunomodulatory properties of 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) remains elusive. We demonstrate here that 1α,25(OH)2D3-mediated suppressive effects on the inducible expression of cytokine genes in human T cells may, in part, be due to diminished activity of the transcription factor NFAT. The vitamin D3 receptor (VDR) and its heterodimeric partner retinoid X receptor α (RXRα) specifically bound to the distal NFAT site in the human IL-2 promoter, and this binding was abolished by mutating unique regions in the NFAT oligonucleotide. In vitro inhibition of NFAT complex formation was noted when VDR-RXRα heterodimers were added to DNA binding reactions containing nuclear extracts from activated B or T cells, whereas in vitro NFκB complex formation was not significantly influenced. Furthermore, 1α,25(OH)2D3 treatment of activated T cells resulted in decreased formation of NFAT complexes detected upon incubation of nuclear extracts from these cells with 32P-labeled probe. Transient expression of both VDR and RXRα, but not of a single component, was capable of inhibiting expression of a NFAT-driven reporter gene in stimulated Jurkat cells in a ligand-dependent manner. These results suggest that NFAT plays a crucial role in 1α,25(OH)2D3-mediated immunosuppressive activity.
Collapse
Affiliation(s)
- Atsuko Takeuchi
- *Section of Experimental Pathology, Department of Pathology, Roger Williams Medical Center, Brown University, Providence, RI 02908
| | - G. Satyanarayana Reddy
- †Department of Pediatrics, Women and Infants Hospital, Brown University, Providence, RI 02903; and
| | - Tadashi Kobayashi
- ‡Department of Hygienic Sciences, Kobe Pharmaceutical University, Kobe, Japan
| | - Toshio Okano
- ‡Department of Hygienic Sciences, Kobe Pharmaceutical University, Kobe, Japan
| | - Jungchan Park
- *Section of Experimental Pathology, Department of Pathology, Roger Williams Medical Center, Brown University, Providence, RI 02908
| | - Surendra Sharma
- *Section of Experimental Pathology, Department of Pathology, Roger Williams Medical Center, Brown University, Providence, RI 02908
| |
Collapse
|
4866
|
Coventry S, Kapur RP, Siebert JR. Cyclopamine-induced holoprosencephaly and associated craniofacial malformations in the golden hamster: anatomic and molecular events. Pediatr Dev Pathol 1998; 1:29-41. [PMID: 10463269 DOI: 10.1007/s100249900004] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Holoprosencephaly is a complex congenital malformation of the brain and is often associated with a spectrum of facial anomalies ranging from normocephaly or nondiagnostic changes to cleft lip/palate (premaxillary dysgenesis), cebocephaly, ethmocephaly, and cyclopia. The primary insult is thought to occur during gastrulation, when prechordal mesenchyme and overlying anterior neural plate undergo complex developmental interactions. Exposure to cyclopamine, a steroid isolated from the desert plant Veratrum californicum, causes holoprosencephaly in mammalian embryos. We have begun to study the pathogenesis of cyclopamine-induced holoprosencephaly and associated craniofacial anomalies in Syrian golden hamsters (Mesocricetus auratus). Embryos were exposed to a single maternal dose of cyclopamine during gastrulation on embryonic day (E) 7.0. By E13.0, 62% of fetuses showed craniofacial malformations, including premaxillary dysgenesis, ocular hypotelorism, and cebocephaly. Facial anomalies were associated with absence of the premaxilla and abnormalities of the midline cranial base, particularly the ethmoid and sphenoid bones. Histological sections from cyclopamine-treated embryos at earlier stages showed marked deficiency of cranial mesenchyme derived from the rostral neural crest. Expression of two transcription factors, HNF-3 beta and Hox-b5, which have been implicated in specification of rostral and caudal neural crest cells, respectively, were examined immunohistochemically. Treatment with cyclopamine caused a transient loss of HNF-3 beta immunoreactivity in prechordal mesenchyme, but had no effect on Hox-b5 expression. The findings suggest that an early event in the pathogenesis of cyclopamine-induced holoprosencephaly may be altered expression of selected proteins in the prechordal mesenchyme and floor plate with secondary impaired development of the adjacent neural plate and cranial neural crest.
Collapse
Affiliation(s)
- S Coventry
- Department of Laboratories (CH-37), Children's Hospital and Medical Center, Seattle, WA 98105, USA
| | | | | |
Collapse
|
4867
|
SHALIN L, PERSSON E, MASIRONI B, RODRIGUEZ-MARTINEZ H, ERIKSSON H. Expression of the Estrogen and Progesterone Receptors and Insulin-like Growth Factor-I Genes in the Porcine Myometrium during Early Pregnancy. J Reprod Dev 1998. [DOI: 10.1262/jrd.44.229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Lena SHALIN
- Division for Reproductive Endocrinology, Karolinska Hospital, S-171 76 , Stockholm, Sweden
| | - Elisabeth PERSSON
- Departments of Anatomy and Histology, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, S-750 07, Uppsala, Sweden
| | - Britt MASIRONI
- Division for Reproductive Endocrinology, Karolinska Hospital, S-171 76 , Stockholm, Sweden
| | - Heriberto RODRIGUEZ-MARTINEZ
- Departments of Obstetrics and Gynaecology, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, S-750 07, Uppsala, Sweden
| | - Håkan ERIKSSON
- Division for Reproductive Endocrinology, Karolinska Hospital, S-171 76 , Stockholm, Sweden
| |
Collapse
|
4868
|
|
4869
|
Esquenet M, Swinnen JV, Van Veldhoven PP, Denef C, Heyns W, Verhoeven G. Retinoids stimulate lipid synthesis and accumulation in LNCaP prostatic adenocarcinoma cells. Mol Cell Endocrinol 1997; 136:37-46. [PMID: 9510066 DOI: 10.1016/s0303-7207(97)00210-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In a previous report we demonstrated that androgens markedly stimulate accumulation of lipid droplets in LNCaP cells. The effects were already evident at low concentrations of androgens optimal for proliferation but became much more pronounced at high concentrations optimal for differentiation. In the present report we explored whether other agonists acting by nuclear receptors and modulating LNCaP growth and differentiation also affect lipid accumulation. The agonists investigated were 1alpha,25-dihydroxycholecalciferol (VD3), all-trans-retinoic acid (atRA), and triiodothyronine (T3). Lipid accumulation was evaluated by Oil Red O staining followed by image analysis of Oil Red O-stained cells or by extraction and measurement of absorbency. Only marginal effects were noted for VD3 and T3. The atRA, on the contrary, increased lipid staining 5-12-fold. This effect required high concentrations of retinoids (10[-6] M) and was accompanied by growth stimulation. Lipid accumulation was less pronounced than that observed with maximally effective concentrations of androgens (10[-3] M R1881). Thin layer chromatography (TLC) and enzymatic determination of the various lipid fractions demonstrated that retinoids increase triacylglycerides and an unidentified lipid fraction with a slightly higher mobility. In contrast with androgens, however, they did not stimulate the accumulation of cholesterol esters. Incorporation studies with [2-14C]acetate revealed that the increased accumulation of the mentioned lipids is related both to increased synthesis and to decreased secretion. Retinoid-induced lipid accumulation is accompanied by increased steady-state levels of the mRNA encoding fatty acid synthase (FAS), a key enzyme involved in lipid synthesis, while the expression of HMG-CoA-reductase, an enzyme controlling cholesterol synthesis is only marginally affected. It is concluded that retinoids share the ability of androgens to increase lipid accumulation in LNCaP cells. The nature of the lipids affected by both agonists, however, differs at least in part suggesting that the underlying mechanisms may also be different. For the studied compounds (androgens, VD3, atRA, and T3) no simple and consistent relationship could be observed between their ability to decrease proliferation and increase differentiation on the one hand and their ability to promote lipid accumulation on the other hand.
Collapse
Affiliation(s)
- M Esquenet
- Laboratory for Experimental Medicine and Endocrinology, Onderwijs en Navorsing, Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
4870
|
Bamberger CM, Else T, Bamberger AM, Beil FU, Schulte HM. Regulation of the human interleukin-2 gene by the alpha and beta isoforms of the glucocorticoid receptor. Mol Cell Endocrinol 1997; 136:23-8. [PMID: 9510064 DOI: 10.1016/s0303-7207(97)00209-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The immunosuppressive effects of glucocorticoids are largely due to transcriptional inhibition of immunologically relevant genes, such as the interleukin-2 (IL-2) gene. These effects are mediated by the intracellular glucocorticoid receptor (GR). In humans, alternative splicing of the GR precursor mRNA gives rise to two receptor isoforms, termed GRalpha and GRbeta. We previously demonstrated that GRbeta could antagonize GRalpha-mediated transactivation of a glucocorticoid-responsive element (GRE)-driven reporter gene in COS-7 cells. The present study was designed to analyze the roles of the two GR isoforms on glucocorticoid-mediated transrepression of the IL-2 gene. Using a recently developed transfection technique, we demonstrate that in primary human lymphocytes, stimulation of a 548 bp IL-2 promoter-luciferase reporter construct by phorbol ester and calcium ionophore is reversed by dexamethasone to a similar extent as in Jurkat T lymphoma cells transfected with a GRalpha expression vector. Transfection of a GRbeta expression vector alone did not result in IL-2 promoter repression in response to glucocorticoids. Furthermore, GRbeta did not antagonize the repressive effects of GRalpha on IL-2 promoter activity. Surprisingly, overexpression of GRbeta in Jurkat cells did not cause significant inhibition of GRalpha-induced transactivation of a GRE-dependent luciferase reporter gene either. We conclude that the transrepressive effect of glucocorticoids on IL-2 gene transcription is exclusively mediated by GRalpha. GRbeta can neither antagonize GRalpha-mediated transactivation nor transrepression in Jurkat cells, indicating a cell type-specific pattern of GRbeta-mediated antiglucocorticoid activity.
Collapse
Affiliation(s)
- C M Bamberger
- IHF Institute for Hormone and Fertility Research, University of Hamburg, Germany.
| | | | | | | | | |
Collapse
|
4871
|
Yoshida E, Aratani S, Itou H, Miyagishi M, Takiguchi M, Osumu T, Murakami K, Fukamizu A. Functional association between CBP and HNF4 in trans-activation. Biochem Biophys Res Commun 1997; 241:664-9. [PMID: 9434765 DOI: 10.1006/bbrc.1997.7871] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CBP functions as a key transcriptional coactivator for a variety of transcription factors. We show here that the hepatocyte nuclear factor 4 (HNF4), a transcription factor in the nuclear receptor superfamily with no defined ligand, is cloned by yeast two-hybrid system using CBP as a bait. GST-pull down assay with nuclear extracts or in vitro translation products revealed that CBP and HNF4 interact with each other at the middle portion (aa 119-375) of HNF4 and two distinct regions (aa 271-451 and 1626-2259) of CBP, respectively, in the ligand-independent manner. Co-transfection experiments indicated that CBP is capable of activating HNF4 site-mediated transcription. These results suggested a functional association between CBP and HNF4 in trans-activation.
Collapse
Affiliation(s)
- E Yoshida
- Institute of Applied Biochemistry, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
4872
|
Abstract
Twenty-five years ago, then President Nixon "declared war" on cancer. In this personal commentary, the war is reviewed. There have been obvious triumphs, for instance in cure of acute lymphocytic leukemia and other forms of childhood cancer, Hodgkin's disease, and testicular cancer. However, substantial advances in molecular oncology have yet to impinge on mortality statistics. Too many adults still die from common epithelial cancers. Failure to appreciate that local invasion and distant metastasis rather then cell proliferation itself are lethal, obsession with cure of advanced disease rather than prevention of early disease, and neglect of the need to arrest preneoplastic lesions, may all have served to make victory elusive.
Collapse
Affiliation(s)
- M B Sporn
- Department of Pharmacology, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| |
Collapse
|
4873
|
Dowell P, Ishmael JE, Avram D, Peterson VJ, Nevrivy DJ, Leid M. p300 functions as a coactivator for the peroxisome proliferator-activated receptor alpha. J Biol Chem 1997; 272:33435-43. [PMID: 9407140 DOI: 10.1074/jbc.272.52.33435] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The integrator protein, p300, was demonstrated to interact with mouse peroxisome proliferator-activated receptor alpha in a ligand-enhanced manner. The PPARalpha-interacting domain of p300 was mapped to amino acids 39-117 which interacted strongly with PPARalpha but did not interact with retinoic acid receptor-gamma or retinoid X receptor-alpha. Amino acids within the carboxyl terminus of PPARalpha as well as residues within the hinge region were required for ligand-dependent interaction with p300. p300 enhanced the transcriptional activation properties of PPARalpha and, therefore, can be considered a bona fide coactivator for this nuclear receptor. These observations extend the group of p300-interacting proteins to include mPPARalpha and further characterize the molecular mechanisms of PPARalpha-mediated transcriptional regulation.
Collapse
Affiliation(s)
- P Dowell
- Program in Molecular and Cellular Biology, College of Pharmacy, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | | | | | | | |
Collapse
|
4874
|
Samad TA, Krezel W, Chambon P, Borrelli E. Regulation of dopaminergic pathways by retinoids: activation of the D2 receptor promoter by members of the retinoic acid receptor-retinoid X receptor family. Proc Natl Acad Sci U S A 1997; 94:14349-54. [PMID: 9405615 PMCID: PMC24972 DOI: 10.1073/pnas.94.26.14349] [Citation(s) in RCA: 164] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dopamine is a neuromodulator involved in the control of key physiological functions. Dopamine-dependent signal transduction is activated through the interaction with membrane receptors of the seven-transmembrane domain G protein-coupled family. Among them, dopamine D2 receptor is highly expressed in the striatum and the pituitary gland as well as by mesencephalic dopaminergic neurons. Lack of D2 receptors in mice leads to a locomotor parkinsonian-like phenotype and to pituitary tumors. The D2 receptor promoter has characteristics of a housekeeping gene. However, the restricted expression of this gene to particular neurons and cells points to a strict regulation of its expression by cell-specific transcription factors. We demonstrate here that the D2 receptor promoter contains a functional retinoic acid response element. Furthermore, analysis of retinoic acid receptor-null mice supports our finding and shows that in these animals D2 receptor expression is reduced. This finding assigns to retinoids an important role in the control of gene expression in the central nervous system.
Collapse
Affiliation(s)
- T A Samad
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/ULP, Collège de France, BP 163, 67404 Illkirch Cedex, Strasbourg, France
| | | | | | | |
Collapse
|
4875
|
Su Y, Shi Y, Stolow MA, Shi YB. Thyroid hormone induces apoptosis in primary cell cultures of tadpole intestine: cell type specificity and effects of extracellular matrix. J Cell Biol 1997; 139:1533-43. [PMID: 9396758 PMCID: PMC2132612 DOI: 10.1083/jcb.139.6.1533] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Thyroid hormone (T3 or 3,5,3'-triiodothyronine) plays a causative role during amphibian metamorphosis. To investigate how T3 induces some cells to die and others to proliferate and differentiate during this process, we have chosen the model system of intestinal remodeling, which involves apoptotic degeneration of larval epithelial cells and proliferation and differentiation of other cells, such as the fibroblasts and adult epithelial cells, to form the adult intestine. We have established in vitro culture conditions for intestinal epithelial cells and fibroblasts. With this system, we show that T3 can enhance the proliferation of both cell types. However, T3 also concurrently induces larval epithelial apoptosis, which can be inhibited by the extracellular matrix (ECM). Our studies with known inhibitors of mammalian cell death reveal both similarities and differences between amphibian and mammalian cell death. These, together with gene expression analysis, reveal that T3 appears to simultaneously induce different pathways that lead to specific gene regulation, proliferation, and apoptotic degeneration of the epithelial cells. Thus, our data provide an important molecular and cellular basis for the differential responses of different cell types to the endogenous T3 during metamorphosis and support a role of ECM during frog metamorphosis.
Collapse
Affiliation(s)
- Y Su
- Laboratory of Molecular Embryology, National Institute of Child Health and Human Development, Bethesda, Maryland 20892-5431, USA
| | | | | | | |
Collapse
|
4876
|
Caelles C, González-Sancho JM, Muñoz A. Nuclear hormone receptor antagonism with AP-1 by inhibition of the JNK pathway. Genes Dev 1997; 11:3351-64. [PMID: 9407028 PMCID: PMC316827 DOI: 10.1101/gad.11.24.3351] [Citation(s) in RCA: 262] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The activity of c-Jun, the major component of the transcription factor AP-1, is potentiated by amino-terminal phosphorylation on serines 63 and 73 (Ser-63/73). This phosphorylation is mediated by the Jun amino-terminal kinase (JNK) and required to recruit the transcriptional coactivator CREB-binding protein (CBP). AP-1 function is antagonized by activated members of the steroid/thyroid hormone receptor superfamily. Recently, a competition for CBP has been proposed as a mechanism for this antagonism. Here we present evidence that hormone-activated nuclear receptors prevent c-Jun phosphorylation on Ser-63/73 and, consequently, AP-1 activation, by blocking the induction of the JNK signaling cascade. Consistently, nuclear receptors also antagonize other JNK-activated transcription factors such as Elk-1 and ATF-2. Interference with the JNK signaling pathway represents a novel mechanism by which nuclear hormone receptors antagonize AP-1. This mechanism is based on the blockade of the AP-1 activation step, which is a requisite to interact with CBP. In addition to acting directly on gene transcription, regulation of the JNK cascade activity constitutes an alternative mode whereby steroids and retinoids may control cell fate and conduct their pharmacological actions as immunosupressive, anti-inflammatory, and antineoplastic agents.
Collapse
Affiliation(s)
- C Caelles
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas, E-28029 Madrid, Spain
| | | | | |
Collapse
|
4877
|
Tanaka T, Yoshida N, Kishimoto T, Akira S. Defective adipocyte differentiation in mice lacking the C/EBPbeta and/or C/EBPdelta gene. EMBO J 1997; 16:7432-43. [PMID: 9405372 PMCID: PMC1170343 DOI: 10.1093/emboj/16.24.7432] [Citation(s) in RCA: 612] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To investigate the role of C/EBP family members during adipocyte differentiation in vivo, we have generated mice lacking the C/EBPbeta and/or C/EBPdelta by gene targeting. Approximately 85% of C/EBPbeta(-/-).delta(-/-) mice died at the early neonatal stage. By 20 h after birth, brown adipose tissue of the interscapular region in wild-type mice contained many lipid droplets, whereas C/EBPbeta(-/-).delta(-/-) mice did not accumulate droplets. In addition, the epidydimal fat pad weight of surviving adult C/EBPbeta(-/-).delta(-/-) mice was significantly reduced compared with wild-type mice. However, these adipose tissues in C/EBPbeta(-/-).delta(-/-) mice exhibit normal expression of C/EBPalpha and PPARgamma, despite impaired adipogenesis. These results demonstrated that C/EBPbeta and C/EBPdelta have a synergistic role in terminal adipocyte differentiation in vivo. The induction of C/EBPalpha and PPARgamma does not always require C/EBPbeta and C/EBPdelta, but co-expression of C/EBPalpha and PPARgamma is not sufficient for complete adipocyte differentiation in the absence of C/EBPbeta and C/EBPdelta.
Collapse
Affiliation(s)
- T Tanaka
- Department of Biochemistry, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663, Japan
| | | | | | | |
Collapse
|
4878
|
Abstract
Vertebrates appear about 500 million years ago in the fossil record. This is only 25-50 million years after the great explosion of multicellular invertebrate body plans in the early Cambrian. On a geological time scale, this interval is a 'blink of an eye', suggesting that the evolution of regulatory genes is likely to be important in the origins of vertebrates. Here we present evidence for a role of steroid receptors in this process based on a phylogenetic analysis suggesting that receptors for androgens, glucocorticoids, mineralocorticoids, and progesterone evolved from an ancestral steroid receptor gene by two successive duplications over a brief time that could have coincided with the origins of vertebrates. Moreover, the duplications of these steroid receptors may be additional evidence for the two duplications on a genome-scale that have been proposed to be important in the evolution of vertebrates. The two successive duplications of steroid receptor genes and their subsequent sequence divergence leading to steroid-specific receptors that regulate growth, development, reproduction and homeostasis in vertebrates may have been one of the events important in vertebrate survival after the Cambrian during global extinctions that occurred about 440 and 370 million years ago.
Collapse
Affiliation(s)
- M E Baker
- Department of Medicine, University of California, San Diego, La Jolla 92093-0623, USA.
| |
Collapse
|
4879
|
Abstract
This review focuses on known genes whose expression may be perturbed by teratogens during early embryogenesis (preorganogenesis). Teratogens may disrupt embryogenesis by modifying positional information. Genes controlling positional information include those specifying the primary body axes: anterior-posterior, dorsal-ventral, or left-right. These genes often encode transcription factors, whose regulation or activation can stimulate aberrant tissue differentiation and morphogenesis. Alternatively, teratogens may directly affect cell differentiation, proliferation, or apoptosis. Hydrophilic signalling molecules such as growth factors and hydrophobic molecules such as retinoids regulate these processes. The signalling pathways activated often induce the coordinate regulation of tissue specific gene expression. In addition to modifying individual signalling pathways, teratogens can synergize with or antagonize the effects of other teratogens through inappropriate interactions between signal transduction pathways. Since teratogens may often directly or indirectly perturb the expression of known or as yet undescribed developmentally critical genes, this review also provides a short description of techniques to identify genes whose expression is altered by teratogens.
Collapse
Affiliation(s)
- M B Rogers
- Department of Biology, University of South Florida, Tampa 33620-5150, USA.
| | | | | |
Collapse
|
4880
|
Klinge CM, Silver BF, Driscoll MD, Sathya G, Bambara RA, Hilf R. Chicken ovalbumin upstream promoter-transcription factor interacts with estrogen receptor, binds to estrogen response elements and half-sites, and inhibits estrogen-induced gene expression. J Biol Chem 1997; 272:31465-74. [PMID: 9395481 DOI: 10.1074/jbc.272.50.31465] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chicken ovalbumin upstream promoter-transcription factor (COUP-TF) was identified as a low abundance protein in bovine uterus that co-purified with estrogen receptor (ER) in a ligand-independent manner and was separated from the ER by its lower retention on estrogen response element (ERE)-Sepharose. In gel mobility shift assays, COUP-TF bound as an apparent dimer to ERE and ERE half-sites. COUP-TF bound to an ERE half-site with high affinity, Kd = 1.24 nM. In contrast, ER did not bind a single ERE half-site. None of the class II nuclear receptors analyzed, i.e. retinoic acid receptor, retinoid X receptor, thyroid receptor, peroxisome proliferator-activated receptor, or vitamin D receptor, were constituents of the COUP-TF.DNA binding complex detected in gel mobility shift assays. Direct interaction of COUP-TF with ER was indicated by GST "pull-down" and co-immunoprecipitation assays. The nature of the ER ligand influenced COUP-TF-ERE half-site binding. When ER was liganded by the antiestrogen 4-hydroxytamoxifen (4-OHT), COUP-TF-half-site interaction decreased. Conversely, COUP-TF transcribed and translated in vitro enhanced the ERE binding of purified estradiol (E2)-liganded ER but not 4-OHT-liganded ER. Co-transfection of ER-expressing MCF-7 human breast cancer cells with an expression vector for COUP-TFI resulted in a dose-dependent inhibition of E2-induced expression of a luciferase reporter gene under the control of three tandem copies of EREc38. The ability of COUP-TF to bind specifically to EREs and half-sites, to interact with ER, and to inhibit E2-induced gene expression suggests COUP-TF regulates ER action by both direct DNA binding competition and through protein-protein interactions.
Collapse
Affiliation(s)
- C M Klinge
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky 40292, USA.
| | | | | | | | | | | |
Collapse
|
4881
|
Bender M, Imam FB, Talbot WS, Ganetzky B, Hogness DS. Drosophila ecdysone receptor mutations reveal functional differences among receptor isoforms. Cell 1997; 91:777-88. [PMID: 9413987 DOI: 10.1016/s0092-8674(00)80466-3] [Citation(s) in RCA: 204] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The steroid hormone ecdysone directs Drosophila metamorphosis via three heterodimeric receptors that differ according to which of three ecdysone receptor isoforms encoded by the EcR gene (EcR-A, EcR-B1, or EcR-B2) is activated by the orphan nuclear receptor USP. We have identified and molecularly mapped two classes of EcR mutations: those specific to EcR-B1 that uncouple metamorphosis, and embryonic-lethal mutations that map to common sequences encoding the DNA- and ligand-binding domains. In the larval salivary gland, loss of EcR-B1 results in loss of activation of ecdysone-induced genes. Comparable transgenic expression of EcR-B1, EcR-B2, and EcR-A in these mutant glands results, respectively, in full, partial, and no repair of that loss.
Collapse
Affiliation(s)
- M Bender
- Department of Developmental Biology, Stanford University Medical Center, California 94305, USA
| | | | | | | | | |
Collapse
|
4882
|
Katagiri Y, Hirata Y, Milbrandt J, Guroff G. Differential regulation of the transcriptional activity of the orphan nuclear receptor NGFI-B by membrane depolarization and nerve growth factor. J Biol Chem 1997; 272:31278-84. [PMID: 9395454 DOI: 10.1074/jbc.272.50.31278] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The immediate-early gene NGFI-B (also called nur77) encodes an orphan nuclear receptor that activates transcription through a unique response element (NBRE). NGFI-B is rapidly induced and modified via phosphorylation by a variety of stimuli that induce cells to differentiate or to proliferate. We have shown that the in vitro phosphorylation of Ser350 located within the "A-box," a motif necessary for DNA binding by NGFI-B, results in a decrease in the binding of NGFI-B to its response element (Hirata, Y., Kiuchi, K., Chen, H.-C., Milbrandt, J., and Guroff, G. (1993) J. Biol. Chem. 268, 24808-24812). We show here that nerve growth factor (NGF)-induced changes in the in vivo phosphorylation of Ser350 accompany transcriptional deactivation of NGFI-B in PC12 cells, that membrane depolarization and NGF treatment cause differential phosphorylation of NGFI-B, and that the transcriptional activation caused by exogenous expression of NGFI-B or membrane depolarization can be inhibited by NGF treatment. In addition, the mutation of Ser350 to Ala abolished the inhibitory effect of NGF on the transcriptional activation of NGFI-B in PC12 cells. These data could provide new insights into the regulation of transcriptional activity required for some neurons to switch from activity-dependent survival to neurotrophin-dependent survival during development.
Collapse
Affiliation(s)
- Y Katagiri
- Section on Growth Factors, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
4883
|
Quignon F, Chen Z, de Thé H. Retinoic acid and arsenic: towards oncogene-targeted treatments of acute promyelocytic leukaemia. BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1333:M53-61. [PMID: 9426202 DOI: 10.1016/s0304-419x(97)00025-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- F Quignon
- CNRS UPR 9051, Hopital St Louis 1, Paris, France
| | | | | |
Collapse
|
4884
|
Monaghan AP, Bock D, Gass P, Schwäger A, Wolfer DP, Lipp HP, Schütz G. Defective limbic system in mice lacking the tailless gene. Nature 1997; 390:515-7. [PMID: 9394001 DOI: 10.1038/37364] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The gene tailless is a member of the superfamily of genes that encode transcription factors of the ligand-activated nuclear receptor type, and is expressed in the invertebrate and vertebrate brain. In mice, its transcripts are restricted to the periventricular zone of the forebrain, the site of origin of neurons and glia. Here we use homologous recombination to generate mice that lack a functional tailless protein. Homozygous mutant mice are viable at birth, indicating that tailless is not required for prenatal survival; however, adult mutant mice show a reduction in the size of rhinencephalic and limbic structures, including the olfactory, infrarhinal and entorhinal cortex, amygdala and dentate gyrus. Both male and female mice are more aggressive than usual and females lack normal maternal instincts. These animals therefore enable a molecular approach to be taken towards understanding the genetic architecture and morphogenesis of the forebrain.
Collapse
Affiliation(s)
- A P Monaghan
- Division of Molecular Biology of the Cell I, German Cancer Research Centre, Heidelberg
| | | | | | | | | | | | | |
Collapse
|
4885
|
Brown PJ, Smith-Oliver TA, Charifson PS, Tomkinson NC, Fivush AM, Sternbach DD, Wade LE, Orband-Miller L, Parks DJ, Blanchard SG, Kliewer SA, Lehmann JM, Willson TM. Identification of peroxisome proliferator-activated receptor ligands from a biased chemical library. CHEMISTRY & BIOLOGY 1997; 4:909-18. [PMID: 9427656 DOI: 10.1016/s1074-5521(97)90299-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The peroxisome proliferator-activated receptors (PPARs) were cloned as orphan members of the nuclear receptor superfamily of transcription factors. The identification of subtype-selective ligands for PPARalpha and PPARgamma has led to the discovery of their roles in the regulation of lipid metabolism and glucose homeostasis. No subtype-selective PPARdelta ligands are available and the function of this subtype is currently unknown. RESULTS A three-component library was designed in which one of the monomers was biased towards the PPARs and the other two monomers were chosen to add chemical diversity. Synthesis and screening of the library resulted in the identification of pools with activity on each of the PPAR subtypes. Deconvolution of the pools with the highest activity on PPARdelta led to the identification of GW 2433 as the first high-affinity PPARdelta ligand. [3H]GW 2433 is an effective radioligand for use in PPARdelta competition-binding assays. CONCLUSIONS The synthesis of biased chemical libraries is an efficient approach to the identification of lead molecules for members of sequence-related receptor families. This approach is well suited to the discovery of small-molecule ligands for orphan receptors.
Collapse
Affiliation(s)
- P J Brown
- Department of Medicinal Chemistry, Glaxo Wellcome Research & Development, PO Box 13398, 5 Moore Drive, Research Triangle Park, NC 27709-3398, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4886
|
Abstract
The past year has witnessed increased confusion as to the role of gonadal hormones in the development of neuroeffectors for sexually differentiated vocalizations in several species. Are sex differences in levels of circulating gonadal hormones robust enough to account for the full spectrum of male/female differences? Understanding how vocal behaviors are generated has improved, permitting greater insights into how differences in cell number and type contribute to male- and female-specific songs in frogs and birds.
Collapse
Affiliation(s)
- D B Kelley
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA.
| |
Collapse
|
4887
|
Qi JS, Desai-Yajnik V, Yuan Y, Samuels HH. Constitutive activation of gene expression by thyroid hormone receptor results from reversal of p53-mediated repression. Mol Cell Biol 1997; 17:7195-207. [PMID: 9372952 PMCID: PMC232577 DOI: 10.1128/mcb.17.12.7195] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Thyroid hormone receptor (T3R) is a member of the steroid hormone receptor gene family of nuclear hormone receptors. In most cells T3R activates gene expression only in the presence of its ligand, L-triiodothyronine (T3). However, in certain cell types (e.g., GH4C1 cells) expression of T3R leads to hormone-independent constitutive activation. This activation by unliganded T3R occurs with a variety of gene promoters and appears to be independent of the binding of T3R to specific thyroid hormone response elements (TREs). Previous studies indicate that this constitutive activation results from the titration of an inhibitor of transcription. Since the tumor suppresser p53 is capable of repressing a wide variety of gene promoters, we considered the possibility that the inhibitor is p53. Evidence to support this comes from studies indicating that expression of p53 blocks T3R-mediated constitutive activation in GH4C1 cells. In contrast with hormone-independent activation by T3R, p53 had little or no effect on T3-dependent stimulation which requires TREs. In addition, p53 mutants which oligomerize with wild-type p53 and interfere with its function also increase promoter activity. This enhancement is of similar magnitude to but is not additive with the stimulation mediated by unliganded T3R, suggesting that they target the same factor. Since p53 mutants are known to target wild-type p53 in the cell, this suggests that T3R also interacts with p53 in vivo and that endogenous levels of p53 act to suppress promoter activity. Evidence supporting both functional and physical interactions of T3R and p53 in the cell is presented. The DNA binding domain (DBD) of T3R is important in mediating constitutive activation, and the receptor DBD appears to functionally interact with the N terminus of p53 in the cell. In vitro binding studies indicate that the T3R DBD is important for interaction of T3R with p53 and that this interaction is reduced by T3. These findings are consistent with the in vivo studies indicating that p53 blocks constitutive activation but not ligand-dependent stimulation. These studies provide insight into mechanisms by which unliganded nuclear hormone receptors can modulate gene expression and may provide an explanation for the mechanism of action of the v-erbA oncoprotein, a retroviral homolog of chicken T3R alpha.
Collapse
Affiliation(s)
- J S Qi
- Department of Pharmacology, New York University Medical Center, New York 10016, USA
| | | | | | | |
Collapse
|
4888
|
Liang VC, Sedgwick T, Shi YB. Characterization of the Xenopus homolog of an immediate early gene associated with cell activation: sequence analysis and regulation of its expression by thyroid hormone during amphibian metamorphosis. Cell Res 1997; 7:179-93. [PMID: 9444397 DOI: 10.1038/cr.1997.19] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The complex transformation of a tadpole to a frog during amphibian development is under the control of thyroid hormone (T3). T3 is known to regulate gene transcription through its nuclear receptors. We have previously isolated many genes which are up-regulated by T3 in the intestine of Xenopus laevis tadpoles. We have now cloned a full-length cDNA for one such gene (IU12). Sequence analysis shows that the IU12 cDNA encodes a plasma membrane protein with 12 transmembrane domains and homologous to a mammalian gene associated with cell activation and organ development. Similarly, we have found that IU12 is activated during intestinal remodeling when both cell death and proliferation take place. Furthermore, IU12 is an early T3-response gene and its expression in the intestine during T3-induced metamorphosis mimics that during normal development. These results argue for a role of IU12 in the signal transduction pathways leading to intestinal metamorphosis.
Collapse
Affiliation(s)
- V C Liang
- Laboratory of Molecular Embryology NICHD/NIH, Bethesda, MD 20892-5431, USA
| | | | | |
Collapse
|
4889
|
Wirtanen L, Huard V, Séguin C. Molecular cloning from neurulating Ambystoma mexicanum embryos of the cDNA encoding an orphan nuclear receptor (aDOR1) closely related to TR2-11. Differentiation 1997; 62:159-70. [PMID: 9503600 DOI: 10.1046/j.1432-0436.1998.6240159.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have isolated a cDNA encoding a novel orphan nuclear receptor, aDOR1, closely related to testicular receptor-2 (TR2) orphan receptor family members, from neurulating Ambystoma mexicanum embryos. The cDNA sequence predicts a protein primary sequence of 416 amino acids with a calculated molecular weight of 45.8 kDa. While the DNA-binding domains of aDOR1 and hTR2-11 share 96% identity, considerable divergence is observed at both extremities of the peptides. At the N-terminus, aDOR1 is 66% identical to hTR2-11 and longer by 37 amino acids. At the C-terminus, despite a greater similarity (69%), aDOR1 is much shorter than the hTR2 isoforms and seems to encode a distinct ligand-binding domain. Expression of aDOR1 was studied by the reverse transcription polymerase chain reaction assay (RT-PCR). High mRNA levels were detected during oogenesis, they remained high during the cleavage stage, and decreased at the mid-blastula transition (MBT). Transcripts increased again at the end of gastrulation, reached a peak level during neurulation, and leveled off after closure of the neural tube. In neurulas dissected along the anteroposterior axis, aDOR1 mRNA was enriched at both extremities of the embryo, while no particular distribution was favored along the dorsoventral axis. Retinoic acid (RA) treatments at the beginning of gastrulation did not affect overall mRNA levels in the neurula nor its distribution along both axes. In the adult, expression was predominant in the brain; lower levels (about 15%) were detectable in all germ layer derivatives, except muscle. These results suggest that aDOR1 may be required for the early determination events occurring during the cleavage stages of development, and may be involved in embryogenesis and in brain function.
Collapse
MESH Headings
- Ambystoma/embryology
- Amino Acid Sequence
- Animals
- Base Sequence
- Blastocyst
- Blotting, Southern
- Brain/growth & development
- Brain/metabolism
- Cloning, Molecular
- DNA-Binding Proteins
- Embryo, Nonmammalian/drug effects
- Embryo, Nonmammalian/physiology
- Gene Expression Regulation, Developmental
- Humans
- Mice
- Molecular Sequence Data
- Nervous System/embryology
- Nuclear Receptor Subfamily 2, Group C, Member 1
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Thyroid Hormone/genetics
- Sequence Analysis
- Tissue Distribution
- Transcription, Genetic
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- L Wirtanen
- Centre de Recherche en Cancérologie de l'Université Laval, Centre Hospitalier Universitaire de Québec, Canada
| | | | | |
Collapse
|
4890
|
Edinger RS, Mambo E, Evans MI. Estrogen-dependent transcriptional activation and vitellogenin gene memory. Mol Endocrinol 1997; 11:1985-93. [PMID: 9415402 DOI: 10.1210/mend.11.13.0037] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The concept of hepatic memory suggests that a gene responds more rapidly to a second exposure of an inducer than it does during the initial activation. To determine how soon estrogen-dependent DNA/protein interactions occur during the primary response, in vivo dimethylsulfate footprinting was carried out using genomic DNA amplified by ligation-mediated PCR. When estrogen was added to disrupted cells from a hormone-naive liver, changes within and around the estrogen response elements occurred within seconds, indicating a direct and rapid effect on this estrogen-responsive promoter that had never before been activated. Because this effect was so rapid relative to the delayed onset of mRNA accumulation during the primary response, run-on transcription assays were used to determine the transcription profiles for four of the yolk protein genes during the primary and secondary responses to estrogen. As with the accumulation of mRNA, the onset of transcription was delayed for all of these genes after a primary exposure to estrogen. Interestingly, after the secondary exposure to estrogen, the vitellogenin I, vitellogenin II, and very low density apolipoprotein II genes displayed a more rapid onset of transcription, whereas the primary and secondary profiles of apolipoprotein B transcription in response to estrogen were identical. Because the apoB gene is constitutively expressed in the absence of estrogen, and the vitellogenins are quiescent before the administration of the hormone, hepatic memory most likely represents a relatively stable event in the transition to an active state of a gene that is committed for tissue-specific expression.
Collapse
Affiliation(s)
- R S Edinger
- Department of Biochemistry, School of Medicine, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown 26506-9142, USA
| | | | | |
Collapse
|
4891
|
Yu X, Mertz JE. Differential regulation of the pre-C and pregenomic promoters of human hepatitis B virus by members of the nuclear receptor superfamily. J Virol 1997; 71:9366-74. [PMID: 9371596 PMCID: PMC230240 DOI: 10.1128/jvi.71.12.9366-9374.1997] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Synthesis of the pre-C and pregenomic RNAs of human hepatitis B virus (HBV) is directed by two overlapping yet separate promoters (X. Yu and J. E. Mertz, J. Virol. 70:8719-8726, 1996). Previously, we reported the identification of a binding site for the nuclear receptor hepatocyte nuclear factor 4 (HNF4) spanning the TATA box-like sequence of the pre-C promoter. This HNF4-binding site consists of an imperfect direct repeat of the consensus half-site sequence 5'-AGGTCA-3' separated by one nucleotide; i.e., it is a DR1 hormone response element (HRE). We show here that other receptors, including chicken ovalbumin upstream promoter transcription factor 1 (COUP-TF1), human testicular receptor 2 (TR2), and peroxisome proliferator-activated receptors (PPARs) as heterodimers with retinoid X receptors (RXRs), can also specifically bind this DR1 HRE. Synthesis of the pre-C and pregenomic RNAs was affected both in transfected hepatoma cells and in a cell-free transcription system by the binding of factors to this DR1 HRE. Interestingly, whereas some members of the hormone receptor superfamily differentially repressed synthesis of the pre-C RNA (e.g., HNF4 and TR2) or activated synthesis of the pregenomic RNA (e.g., PPARgamma-RXRalpha), other members (e.g., COUP-TF1) coordinately repressed synthesis of both the pre-C and pregenomic RNAs. Thus, HBV likely regulates its expression and replication in part via this DR1 HRE. These findings indicate that appropriate ligands to nuclear receptors may be useful in the treatment of HBV infection.
Collapse
MESH Headings
- Base Sequence
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
- COUP Transcription Factor I
- Cell-Free System
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/pharmacology
- Gene Expression Regulation, Viral
- Genome, Viral
- Hepatitis B Core Antigens/biosynthesis
- Hepatitis B Core Antigens/genetics
- Hepatitis B Core Antigens/metabolism
- Hepatitis B virus/genetics
- Hepatocyte Nuclear Factor 4
- Humans
- Molecular Sequence Data
- Nuclear Proteins/metabolism
- Nuclear Proteins/pharmacology
- Nuclear Receptor Subfamily 2, Group C, Member 1
- Phosphoproteins/metabolism
- Phosphoproteins/pharmacology
- Promoter Regions, Genetic
- Protein Precursors/biosynthesis
- Protein Precursors/genetics
- Protein Precursors/metabolism
- RNA Precursors/biosynthesis
- RNA, Viral/biosynthesis
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Retinoic Acid/metabolism
- Receptors, Thyroid Hormone/metabolism
- Repressor Proteins/metabolism
- Repressor Proteins/pharmacology
- Retinoid X Receptors
- Transcription Factors/metabolism
- Transcription Factors/pharmacology
- Transcription, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- X Yu
- McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison 53706-1599, USA
| | | |
Collapse
|
4892
|
Savoldi G, Fenaroli A, Ferrari F, Rigaud G, Albertini A, Di Lorenzo D. The glucocorticoid receptor regulates the binding of C/EPBbeta on the alpha-1-acid glycoprotein promoter in vivo. DNA Cell Biol 1997; 16:1467-76. [PMID: 9428795 DOI: 10.1089/dna.1997.16.1467] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A complex interaction between the Glucocorticoid Receptor (GR), C/EBPbeta, and other transcription factors activate the Alpha-1 Acid Glycoprotein (AGP) promoter in HTC(JZ-1) rat hepatoma culture cells. This effect is mediated by the so-called Steroid Responsive Unit (SRU) of the AGP promoter that contains several binding sites for C/EBP transcription factors, some of which overlap with the Glucocorticoid Responsive Element (GRE). Our in vivo footprinting experiments revealed that the GRE- and the C/EBP-binding sites were already occupied glucocorticoid dependently in HTC(JZ-1) cells 10 min after dexamethasone administration (10(-6) M). Furthermore, local changes in the chromatine structure shown by the appearance of DNAse I hypersensitive sites (HS sites) also took place. These changes were probably dependent on a tissue-specific organization of the chromatine at the SRU because they were not detectable in a different glucocorticoid-responsive cell line (PC12) that did not express AGP. Here, we have also shown that withdrawal of dexamethasone or addition of the anti-glucocorticoid RU486 were able to revert the pattern induced by dexamethasone in vivo. The disappearance of the protected region and the hypersensitive sites, typical of the hormone activated promoter, confirmed the necessity of the GR to be bound by the agonist and the inability of the GR-antagonist complex to bind the DNA. By functional assays, we showed that the occupancy of the SRU by these transcriptional proteins in vivo correlated with the activation of the AGP gene transcription. With these results, we have shown that one of the functions of the GR to activate transcription of the AGP gene is to recruit C/EBPbeta and to maintain it bound at its target DNA sequences (SRU). This process was not accomplished by RU486.
Collapse
Affiliation(s)
- G Savoldi
- Laboratory of Hormonology and Toxicology, Civic Hospital of Brescia, Italy
| | | | | | | | | | | |
Collapse
|
4893
|
Li H, Leo C, Schroen DJ, Chen JD. Characterization of receptor interaction and transcriptional repression by the corepressor SMRT. Mol Endocrinol 1997; 11:2025-37. [PMID: 9415406 DOI: 10.1210/mend.11.13.0028] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) and N-CoR (nuclear receptor corepressor) are two related transcriptional corepressors that contain separable domains capable of interacting with unliganded nuclear receptors and repressing basal transcription. To decipher the mechanisms of receptor interaction and transcriptional repression by SMRT/N-CoR, we have characterized protein-protein interacting surfaces between SMRT and nuclear receptors and defined transcriptional repression domains of both SMRT and N-CoR. Deletional analysis reveals two individual nuclear receptor domains necessary for stable association with SMRT and a C-terminal helix essential for corepressor dissociation. Coordinately, two SMRT domains are found to interact independently with the receptors. Functional analysis reveals that SMRT contains two distinct repression domains, and the corresponding regions in N-CoR also repress basal transcription. Both repression domains in SMRT and N-CoR interact weakly with mSin3A, which in turn associates with a histone deacetylase HDAC1 in a mammalian two-hybrid assay. Far-Western analysis demonstrates a direct protein-protein interaction between two N-CoR repression domains with mSin3A. Finally we demonstrate that overexpression of full-length SMRT further represses basal transcription from natural promoters. Together, these results support a role of SMRT/N-CoR in corepression through the utilization of multiple mechanisms for receptor interactions and transcriptional repression.
Collapse
Affiliation(s)
- H Li
- Department of Pharmacology and Molecular Toxicology, University of Massachusetts Medical School, Worcester 01655-0126, USA
| | | | | | | |
Collapse
|
4894
|
Latchman DS. How can we use our growing understanding of gene transcription to discover effective new medicines? Curr Opin Biotechnol 1997; 8:713-7. [PMID: 9425662 DOI: 10.1016/s0958-1669(97)80125-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although drugs that target gene transcription are in wide therapeutic use, they were all identified on the basis of their effect on a specific biological process, such as inflammation or hormone responses, and were only subsequently shown to target transcription. The recent progress in understanding the mechanism of action of these drugs, and the mechanisms of transcriptional regulation in general, offers hope for a new generation of drugs isolated on the basis of their ability to modulate either the synthesis of transcription factors, the regulation of their activity by ligands or phosphorylation events, their protein-protein interactions or their binding to DNA.
Collapse
Affiliation(s)
- D S Latchman
- Department of Molecular Pathology, Windeyer Institute of Medical Science, University College London Medical School, UK.
| |
Collapse
|
4895
|
Grandien K, Berkenstam A, Gustafsson JA. The estrogen receptor gene: promoter organization and expression. Int J Biochem Cell Biol 1997; 29:1343-69. [PMID: 9570132 DOI: 10.1016/s1357-2725(97)89967-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The estrogen receptor (ER) is a ligand-activated transcription factor and a member of a large family of nuclear hormone receptors. As a mediator of estrogen hormone action, the ER is involved in many important physiological processes. ER gene expression has been demonstrated to be restricted to certain tissues and under complex hormonal control. However, the molecular mechanisms involved have remained largely unknown. Due to this lack of knowledge an investigation was undertaken to characterize the promoter organization of ER gene and investigate its expression. Approximately 3 kb of the 5' flanking region of the human ER (hER) gene was isolated and sequenced. By performing RT-PCR and RACE experiments it was shown that the hER gene is transcribed from three different promoters. Transcription of the hER gene from these promoters yields three different mRNA isoforms with unique 5' untranslated regions (5'UTRs), but identical coding regions. The expression pattern of the hER mRNA isoforms was investigated by RT-PCR. Both the A- and B-mRNA isoforms were found to be expressed in breast and uterus, whereas expression of the C-transcript was predominantly detected in liver. In bone cells only expression of the B-mRNA could be detected. The steady-state levels of the A- and B-transcripts in normal breast and uterus were quantified and compared with the hER mRNA levels in established cancer cell lines derived from the same tissues. This demonstrated approximately equal levels of the two transcripts in normal tissues whereas the A-mRNA was the most abundant isoform in the cancer cell lines investigated. Approximately 4.5 kb of the 5' flanking region of the rat ER (rER) gene were sequenced. Sequence analysis and PCR experiments suggested that the promoter organization of the rat and human ER genes is only partially conserved which might indicate species-specific differences in the regulation of ER expression. In conclusion, this work suggests tissue-specific alternative promoter usage as a mechanism in the regulation of human and rat ER gene expression.
Collapse
Affiliation(s)
- K Grandien
- Department of Medical Nutrition, Karolinska Institute, NOVUM, Huddinge, Sweden
| | | | | |
Collapse
|
4896
|
Tenbaum S, Baniahmad A. Nuclear receptors: structure, function and involvement in disease. Int J Biochem Cell Biol 1997; 29:1325-41. [PMID: 9570131 DOI: 10.1016/s1357-2725(97)00087-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear hormone receptors are acting as transcription factors in the cell nucleus. They regulate gene expression of hormonal regulated target genes. The role of hormone in the transcriptional process is to modulate and change the nuclear receptor functionality. Receptors contain a DNA binding domain that enables them to bind to hormone response elements of target genes. Nuclear hormone receptors bind to lipophilic hormones produced by the organisms' endocrine system, which links the secretion of hormones directly to regulation of gene expression of responsive tissues. In recent years increasing numbers of naturally occurring mutations of a variety of nuclear hormone receptor genes were identified in patients showing abnormalities in hormonal response. Here, we present an overview of nuclear receptors and their mutant forms which cause human syndromes or are associated with cancer progression. The major scope of this article is to give an overview on the structural-functional relationship and based on that, to understand the effects of naturally occurring receptor mutants on the molecular level. Thereby, functional aberrations of naturally occurring receptors for androgen, glucocorticoids, mineralocorticoid, estrogen, vitamin D3, retinoic acid, and thyroid hormone as well as the orphan receptor DAX1 are discussed.
Collapse
Affiliation(s)
- S Tenbaum
- Genetisches Institut der Justus-Liebig Universität, Giessen, Germany
| | | |
Collapse
|
4897
|
Polly P, Carlberg C, Eisman JA, Morrison NA. 1 alpha,25-dihydroxyvitamin D3 receptor as a mediator of transrepression of retinoid signaling. J Cell Biochem 1997; 67:287-96. [PMID: 9361184 DOI: 10.1002/(sici)1097-4644(19971201)67:3<287::aid-jcb1>3.0.co;2-s] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The receptors for retinoic acid (RA) and for 1 alpha,25-dihydroxyvitamin D3 (VD), RAR, RXR, and VDR are ligand-inducible members of the nuclear receptor superfamily. These receptors mediate their regulatory effects by binding as dimeric complexes to response elements located in regulatory regions of hormone target genes. Sequence scanning of the tumor necrosis factor-alpha type 1 receptor (TNF alpha RI) gene identified a 3' enhancer region composed of two directly repeated hexameric core motifs spaced by 2 nucleotides (DR2). On this novel DR2-type sequence, but not on a DR5-type RA response element, VD was shown to act through its receptor, the vitamin D receptor (VDR), as a repressor of retinoid signalling. The repression appears to be mediated by competitive protein-protein interactions between VDR, RAR, RXR, and possibly their cofactors. This VDR-mediated transrepression of retinoid signaling suggests a novel mechanism for the complex regulatory interaction between retinoids and VD.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Antigens, CD/genetics
- Binding, Competitive
- Calcitriol/metabolism
- Cholecalciferol
- DNA, Recombinant
- Humans
- Mice
- Osteocalcin/genetics
- Promoter Regions, Genetic/genetics
- Receptors, Calcitriol/genetics
- Receptors, Retinoic Acid/genetics
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Retinoic Acid Receptor alpha
- Retinoid X Receptors
- Signal Transduction/genetics
- Transcription Factors/genetics
- Transcription, Genetic/genetics
- Tretinoin/metabolism
Collapse
Affiliation(s)
- P Polly
- Bone and Mineral Program, Garvan Institute of Medical Research, Sydney, Australia
| | | | | | | |
Collapse
|
4898
|
Seol W, Chung M, Moore DD. Novel receptor interaction and repression domains in the orphan receptor SHP. Mol Cell Biol 1997; 17:7126-31. [PMID: 9372944 PMCID: PMC232569 DOI: 10.1128/mcb.17.12.7126] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
SHP (short heterodimer partner) is a novel orphan receptor that lacks a conventional DNA binding domain and interacts with other members of the nuclear hormone receptor superfamily. We have characterized the SHP sequences required for interaction with other superfamily members, and have defined an SHP repressor domain. In the mammalian two-hybrid system, a fusion of full-length SHP to the GAL4 DNA binding domain shows 9-cis-retinoic acid-dependent interaction with a VP16-retinoid X receptor alpha (RXR alpha) fusion. By deletion analysis, sequences required for this RXR interaction map to the central portion of SHP (amino acids 92 to 148). The same region is required for interaction with RXR in vitro and in the yeast two-hybrid system, and results from the yeast system suggest that the same SHP sequences are required for interaction with other members of the nuclear hormone receptor superfamily such as thyroid hormone receptor and retinoic acid receptor. In mammalian cells, a GAL4-SHP fusion protein shows about 10-fold-decreased transcriptional activation relative to GAL4 alone, and fusion of SHP to the C terminus of a GAL4-VP16 fusion to generate a triple chimera also results in a strong decrease in transactivation activity. Sequences required for this repressor function were mapped to the C terminus of SHP. This region is distinct from that required for corepressor interaction by other members of the nuclear hormone receptor superfamily, and SHP did not interact with N-CoR in either the yeast or mammalian two-hybrid system. Together, these results identify novel receptor interaction and repressor domains in SHP and suggest two distinct mechanisms for inhibition of receptor signaling pathways by SHP.
Collapse
Affiliation(s)
- W Seol
- Department of Molecular Biology, Massachusetts General Hospital, Boston 02114, USA
| | | | | |
Collapse
|
4899
|
Abstract
It is well known that thyroid hormone plays a crucial role in the development and maturation of the nervous system. However, little is known about the role of thyroid hormone in the adult brain. In this short review we have dwelt on this point, with regard to the role of thyroid hormone on neuropeptide gene expression regulation in the paraventricular nucleus of the hypothalamus and in extrahypothalamic brain areas, on neurotrophin and neurotrophin receptor expression in the hippocampus and basal forebrain in basal conditions, and after neurotoxic challenges. Effects of hypothyroidism are discussed in view of a possible role of thyroid status in brain aging quality.
Collapse
Affiliation(s)
- L Calzà
- Institute of Human Physiology, University of Cagliari, Italy
| | | | | |
Collapse
|
4900
|
Kim J, Kemper B. Phenobarbital alters protein binding to the CYP2B1/2 phenobarbital-responsive unit in native chromatin. J Biol Chem 1997; 272:29423-5. [PMID: 9367997 DOI: 10.1074/jbc.272.47.29423] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Phenobarbital is a classical inducer of the drug metabolizing cytochrome P450 genes, but the molecular mechanism of induction has not been elucidated. Functional analyses have identified a phenobarbital-responsive unit in the rat CYP2B1/2 and mouse Cyp2b10 genes about -2.3 kilobase pairs from the transcriptional start site, but little or no changes in protein binding to this region were observed in vitro. To examine the role of chromatin structure, protein binding to the phenobarbital-responsive unit assessed by in vitro DNase I footprinting was compared with that assessed by DNase I in vivo footprints in native chromatin. A region centered on a putative nuclear factor-1 site was the major protected region in in vitro footprints, and there were no detectable differences in binding between extracts from control and phenobarbital-treated animals. In contrast, phenobarbital treatment dramatically altered the protection pattern in native chromatin. In control samples a core region of about 25 base pairs (bp) centered on the nuclear factor-1 site was protected. However, after phenobarbital treatment, the protection of this core region was increased, and more dramatically the region of protection was extended 20 bp to either side so that a total of about 60 bp were protected. These results provide the first evidence that phenobarbital treatment alters the composition or architecture of proteins binding to the phenobarbital-responsive unit region and indicate that chromatin structure is important in this process. Because proteins are bound to the region in the untreated animal, the mechanism of induction involves the activation of proteins bound to the region and possibly recruitment of additional regulatory proteins rather than conversion of a closed chromatin structure to an open one that can bind regulatory factors.
Collapse
Affiliation(s)
- J Kim
- Department of Molecular and Integrative Physiology and the College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | |
Collapse
|