Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Science ; 184(4142): 1188-91, 1974 Jun 14.
Article in English | MEDLINE | ID: mdl-4364362

ABSTRACT

Saturable binding activities for triiodothyronine were demonstrated in vitro with isolated nuclei and soluble nuclear extracts of rat liver, kidney, and cultured GH(1) cells. The binding activity can be extracted from nuclei in soluble form with no significant change in hormone affinity and has properties of a nonhistone protein.


Subject(s)
Cell Nucleus/metabolism , Receptors, Cell Surface/drug effects , Triiodothyronine/metabolism , Animals , Binding Sites/drug effects , Cell Fractionation , Cell Line , In Vitro Techniques , Iodine Radioisotopes , Kidney , Liver , Pituitary Neoplasms , Rats , Rats, Inbred WF
2.
Science ; 181(4106): 1253-6, 1973 Sep 28.
Article in English | MEDLINE | ID: mdl-4737760

ABSTRACT

Cells from a rat pituitary tumor cell line will respond in vitro to physiological concentrations of L-thyroxine and L-triiodothyronine. The cells are grown in a cultutre medium that contains serum from a hypothyroid calf. Dose-response relationships of a vacriety of thyronine derivatives indicate that this system has a specificity of response which is similar to that observed in vitro.


Subject(s)
Glucose/metabolism , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Animals , Cell Division/drug effects , Clone Cells , Dose-Response Relationship, Drug , Pituitary Neoplasms , Rats
3.
Oncogene ; 26(2): 277-83, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-16832349

ABSTRACT

A subset of papillary renal cell carcinomas (RCC) is characterized by the expression of a TFE3 fusion protein, where the fusion partner can be any of the several proteins identified so far such as PSF (PTB associated splicing factor), NonO, PRCC, CLTC and ASPL. These proteins result from chromosomal translocations involving the TFE3 gene located on the X chromosome. Our present study documents the central role of PSF-TFE3 in oncogenic transformation. We show that the inhibition of PSF-TFE3 expression through siRNA or shRNA leads to impaired growth, proliferation, invasion potential and long-term survival of UOK-145 papillary renal carcinoma-derived cells, which endogenously express PSF-TFE3. The oncogenic potential of PSF-TFE3 became evident by stable expression of PSF-TFE3 in NIH-3T3 mouse fibroblast cells, which leads to the acquisition of anchorage-independent growth as revealed by soft agar assay. In addition, the expression of PSF-TFE3 in normal renal proximal tubular epithelial cells from where such tumors originate leads to dedifferentiation and loss of some key functional proteins, which may reflect an initial step in the multistep process of tumor development. This suggests that the expression of PSF-TFE3 in renal epithelial cells plays an important role in the initiation and maintenance of oncogenic phenotype in papillary RCC.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Carcinoma, Papillary/etiology , Carcinoma, Renal Cell/etiology , Kidney Neoplasms/etiology , Oncogene Proteins, Fusion/physiology , RNA-Binding Proteins/physiology , Animals , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Adhesion , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Cells, Cultured , Epithelial Cells/metabolism , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mice , NIH 3T3 Cells , Neoplasm Invasiveness , Neovascularization, Pathologic , PTB-Associated Splicing Factor , RNA, Small Interfering/pharmacology
4.
J Clin Invest ; 63(6): 1229-40, 1979 Jun.
Article in English | MEDLINE | ID: mdl-221536

ABSTRACT

We have previously demonstrated that L-triiodothyronine (L-T3) induces an increase in growth hormone synthesis and messenger RNA in cultured GH1 cells, a rat pituitary cell line. In addition to regulating the growth hormone response, L-T3 elicits a time- and dose-dependent reduction in the level of its nuclear receptor, which is a direct function of the occupancy of the receptor binding site. In this study we have compared the relative affinity of L-T3, triiodothyroacetic acid, D-triiodothyronine (D-T3), and L-thyroxine (L-T4) for the receptor with the induction of the growth hormone synthesis and the ability of these compounds to elicit a reduction in thyroid hormone nuclear receptor levels. Triiodothyroacetic acid and D-T3 were specifically examined because the biologic effect of these compounds in the intact rat is significantly lower than predicted by their affinity for the receptor using isolated rat liver nuclei in vitro. In intact cells each compound demonstrated an excellent relationship between the relative receptor affinity, the induction of growth hormone production, and the concentration-dependent reduction in nuclear receptor levels. With the exception of D-T3, the relative affinity of iodothyronine was identical for the receptor using intact cells in serum-free media, or isolated GH1 cell nuclei in vitro. The apparent receptor affinity of D-T3 with intact cells was 5.5-fold lower than with isolated nuclei, which suggests a decrease in cell entry of D-T3 relative to the other iodothyronines. Quantitation of the [125I]iodothyronine associated with the receptor in GH1 cells after a 36-h incubation with L-125I-T4 was 90% L-T4 and 10% L-T3, which indicates that the major effect of L-T4 in GH1 cells is a result of intrinsic L-T4 activity. Studies with dispersed rat anterior pituitary cells demonstrated that L-T3 induces growth hormone synthesis and elicits a reduction in nuclear receptor levels in the same fashion as GH1 cells. The observation that thyroid hormone influences dispersed rat pituitary cells in a fashion qualitatively similar to GH1 cells may have implications for the growth hormone response of the somatotroph cell in vivo to different thyroidal states.


Subject(s)
Growth Hormone/biosynthesis , Receptors, Cell Surface/metabolism , Thyroxine/pharmacology , Triiodothyronine/analogs & derivatives , Triiodothyronine/pharmacology , Animals , Cell Line , Cell Nucleus/metabolism , Rats , Thyroxine/metabolism , Triiodothyronine/metabolism
5.
J Clin Invest ; 53(2): 656-9, 1974 Feb.
Article in English | MEDLINE | ID: mdl-11344581

ABSTRACT

High-affinity, limited-capacity nuclear binding activities, putative receptors for triiodothyronine, were detected after incubation of hormone with intact rat pituitary GH1 cells in culture, isolated GH1 cell nuclei, or rat liver nuclei. The total number of triiodothyronine binding sites per nucleus was similar in each case (approximately 8,000). The estimated equilibrium dissociation constants were virtually identical in isolated GH1 cell nuclei and rat liver nuclei, and both values were similar to that determined in intact GH2 cells. These results suggest that mechanisms of thyroid hormone action defined in cell culture could apply to thyroid hormone regulatory effects in vivo.


Subject(s)
Liver/metabolism , Pituitary Gland/metabolism , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Pituitary Gland/cytology , Rats , Triiodothyronine/administration & dosage
6.
J Clin Invest ; 54(4): 853-65, 1974 Oct.
Article in English | MEDLINE | ID: mdl-4372251

ABSTRACT

We previously reported that putative nuclear receptors for thyroid hormone can be demonstrated by incubation of hormone either with intact GH(1) cells, a rat pituitary tumor cell line, or with isolated GH(1) cell nuclei and rat liver nuclei in vitro. We characterized further the kinetics of triiodothyronine (T3) and thyroxine (T4) binding and the biochemical properties of the nuclear receptor after extraction to a soluble form with 0.4 M KCl. In vitro binding of [(125)I]T3 and [(125)I]T4 with GH(1) cell and rat liver nuclear extract was examined at 0 degrees C and 37 degrees C. Equilibrium was attained within 5 min at 37 degrees C and 2 h at 0 degrees C. The binding activity from GH(1) cells was stable for at least 1 h at 37 degrees C and 10 days at - 20 degrees C. Chromatography on a weak carboxylic acid column and inactivation by trypsin and Pronase, but not by DNase or RNase, suggested that the putative receptor was a nonhistone protein. The estimated equilibrium dissociation constants (K(d)) for hormone binding to the solubilized nuclear binding activity was 1.80 x 10(-10) M (T3) and 1.20 x 10(-9) M (T4) for GH(1) cells and 1.57 x 10(-10) M (T3) and 2.0 x 10(-9) M (T4) for rat liver. These K(d) values for T3 are virtually identical to those which we previously reported with isolated rat liver nuclei and GH(1) cell nuclei in vitro. The 10-fold greater affinity for T3 compared to T4 in the nuclear extract is also identical to that observed with intact GH(1) cells. In addition, the [(125)I]T3 and [(125)I]T4 high-affinity binding in the nuclear extract were inhibited by either nonradioactive T3 or T4, which suggests that the binding activity in nuclear extract was identical for T3 and T4. In contrast, the binding activity for T4 and T3 in GH(1) cell cytosol was markedly different from that observed with nuclear extract (K(d) values were 2.87 x 10(-10) M for T4 and 1.13 x 10(-9) M for T3). Our results indicate that nuclear receptors for T3 and T4 can be isolated in a soluble and stable form with no apparent change in hormonal affinity. This should allow elucidation of the mechanisms of thyroid hormone action at the molecular level.


Subject(s)
Cell Nucleus/metabolism , Receptors, Cell Surface , Thyroxine/metabolism , Triiodothyronine/metabolism , Animals , Cell Fractionation , Cell Line , Chromatography , Chromatography, Gel , Cytosol/metabolism , Histones/metabolism , Iodine Radioisotopes , Kinetics , Liver/metabolism , Liver/ultrastructure , Nucleoproteins/metabolism , Pituitary Neoplasms , Protein Binding , Rats , Serum Albumin, Bovine/metabolism
7.
J Clin Invest ; 91(2): 538-46, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8381821

ABSTRACT

We have examined the c-erbA beta thyroid hormone receptor gene in a kindred, G.H., with a member, patient G.H., who had a severe form of selective pituitary resistance to thyroid hormones (PRTH). This patient manifested inappropriately normal thyrotropin-stimulating hormone, markedly elevated serum free thyroxine (T4) and total triiodothyronine (T3), and clinical hyperthyroidism. The complete c-erbA beta 1 coding sequence was examined by a combination of genomic and cDNA cloning for patient G.H. and her unaffected father. A single mutation, a guanine to adenine transition at nucleotide 1,232, was found in one allele of both these members, altering codon 311 from arginine to histidine. In addition, a half-sister of patient G.H. also harbored this mutant allele and, like the father, was clinically normal. The G.H. receptor, synthesized with reticulocyte lysate, had significantly defective T3-binding activity with a Ka of approximately 5 x 10(8) M-1. RNA phenotyping using leukocytes and fibroblasts demonstrated an equal level of expression of wild-type and mutant alleles in patient G.H. and her unaffected father. Finally, the G.H. receptor had no detectable dominant negative activity in a transfection assay. Thus, in contrast to the many other beta-receptor mutants responsible for the generalized form of thyroid hormone resistance, the G.H. receptor appeared unable to antagonize normal receptor function. These results suggest that the arginine at codon 311 in c-erbA beta is crucial for the structural integrity required for dominant negative function. The ARG-311-HIS mutation may contribute to PRTH in patient G.H. by inactivating a beta-receptor allele, but it cannot be the sole cause of the disease.


Subject(s)
Codon , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptors, Thyroid Hormone/genetics , Adolescent , Adult , Alleles , Arginine , Base Sequence , Child, Preschool , Female , Genes, Dominant , Histidine , Humans , Male , Molecular Sequence Data , Phenotype
8.
Mol Cell Biol ; 13(8): 5057-69, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8393143

ABSTRACT

We report that thyroid hormone (T3) receptor (T3R) can activate the human immunodeficiency virus type 1 (HIV-1) long terminal repeat (LTR). Purified chick T3R-alpha 1 (cT3R-alpha 1) binds as monomers and homodimers to a region in the LTR (nucleotides -104 to -75 [-104/-75]) which contains two tandem NF-kappa B binding sites and to a region (-80/-45) which contains three Sp1 binding sites. In contrast, human retinoic acid receptor alpha (RAR-alpha) and mouse retinoid X receptor beta (RXR-beta) do not bind to these elements. However, RXR-beta binds to these elements as heterodimers with cT3R-alpha 1 and to a lesser extent with RAR-alpha. Gel mobility shift assays also revealed that purified NF-kappa B p50/65 or p50/50 can bind to one but not both NF-kappa B sites simultaneously. Although the binding sites for p50/65, p50/50, and T3R, or Sp1 and T3R, overlap, their binding is mutually exclusive, and with the inclusion of RXR-beta, the major complex is the RXR-beta-cT3R-alpha 1 heterodimer. The NF-kappa B region of the LTR and the NF-kappa B elements from the kappa light chain enhancer both function as T3 response elements (TREs) when linked to a heterologous promoter. The TREs in the HIV-1 NF-kappa B sites appear to be organized as a direct repeat with an 8- or 10-bp gap between the half-sites. Mutations within the NF-kappa B motifs which eliminate binding of cT3R-alpha 1 also abolish stimulation by T3, indicating that cT3R-alpha 1 binding to the Sp1 region does not independently mediate activation by T3. The Sp1 region, however, is converted to a functionally strong TRE by the viral tat factor. These studies indicate that the HIV-1 LTR contains both tat-dependent and tat-independent TREs and reveal the potential for T3R to modulate other genes containing NF-kappa B- and Sp1-like elements. Furthermore, they indicate the importance of other transcription factors in determining whether certain T3R DNA binding sequences can function as an active TRE.


Subject(s)
Gene Expression Regulation, Viral , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Receptors, Thyroid Hormone/physiology , Base Sequence , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Oligonucleotides/chemistry , Receptors, Retinoic Acid , Regulatory Sequences, Nucleic Acid , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transcriptional Activation
9.
Mol Cell Biol ; 20(14): 5048-63, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10866662

ABSTRACT

We describe the cloning and characterization of a new family of nuclear receptor coregulators (NRCs) which modulate the function of nuclear hormone receptors in a ligand-dependent manner. NRCs are expressed as alternatively spliced isoforms which may exhibit different intrinsic activities and receptor specificities. The NRCs are organized into several modular structures and contain a single functional LXXLL motif which associates with members of the steroid hormone and thyroid hormone/retinoid receptor subfamilies with high affinity. Human NRC (hNRC) harbors a potent N-terminal activation domain (AD1), which is as active as the herpesvirus VP16 activation domain, and a second activation domain (AD2) which overlaps with the receptor-interacting LXXLL region. The C-terminal region of hNRC appears to function as an inhibitory domain which influences the overall transcriptional activity of the protein. Our results suggest that NRC binds to liganded receptors as a dimer and this association leads to a structural change in NRC resulting in activation. hNRC binds CREB-binding protein (CBP) with high affinity in vivo, suggesting that hNRC may be an important functional component of a CBP complex involved in mediating the transcriptional effects of nuclear hormone receptors.


Subject(s)
Intracellular Signaling Peptides and Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , CREB-Binding Protein , Cloning, Molecular , Dimerization , Herpes Simplex Virus Protein Vmw65/chemistry , Herpes Simplex Virus Protein Vmw65/metabolism , Hormones/metabolism , Humans , Ligands , Molecular Sequence Data , Mutation , Nuclear Receptor Coactivators , Protein Conformation , Rats , Receptors, Cytoplasmic and Nuclear/chemistry , Response Elements , Sequence Homology, Amino Acid , Trans-Activators/genetics , Transcription, Genetic
10.
Mol Cell Biol ; 21(24): 8371-84, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713274

ABSTRACT

The identification of a novel coregulator for nuclear hormone receptors, designated NRIF3, was recently reported (D. Li et al., Mol. Cell. Biol. 19:7191-7202, 1999). Unlike most known coactivators, NRIF3 exhibits a distinct receptor specificity in interacting with and potentiating the activity of only TRs and RXRs but not other examined nuclear receptors. However, the molecular basis underlying such specificity is unclear. In this report, we extended our study of NRIF3-receptor interactions. Our results suggest a bivalent interaction model, where a single NRIF3 molecule utilizes both the C-terminal LXXIL (receptor-interacting domain 1 [RID1]) and the N-terminal LXXLL (RID2) modules to cooperatively interact with TR or RXR (presumably a receptor dimer), with the spacing between RID1 and RID2 playing an important role in influencing the affinity of the interactions. During the course of these studies, we also uncovered an NRIF3-NRIF3 interaction domain. Deletion and mutagenesis analyses mapped the dimerization domain to a region in the middle of NRIF3 (residues 84 to 112), which is predicted to form a coiled-coil structure and contains a putative leucine zipper-like motif. By using Gal4 fusion constructs, we identified an autonomous transactivation domain (AD1) at the C terminus of NRIF3. Somewhat surprisingly, full-length NRIF3 fused to the DNA-binding domain of Gal4 was found to repress transcription of a Gal4 reporter. Further analyses mapped a novel repression domain (RepD1) to a small region at the N-terminal portion of NRIF3 (residues 20 to 50). The NRIF3 gene encodes at least two additional isoforms due to alternative splicing. These two isoforms contain the same RepD1 region as NRIF3. Consistent with this, Gal4 fusions of these two isoforms were also found to repress transcription. Cotransfection of NRIF3 or its two isoforms did not relieve the transrepression function mediated by their corresponding Gal4 fusion proteins, suggesting that the repression involves a mechanism(s) other than the recruitment of a titratable corepressor. Interestingly, a single amino acid residue change of a potential phosphorylation site in RepD1 (Ser(28) to Ala) abolishes its transrepression function, suggesting that the coregulatory property of NRIF3 (or its isoforms) might be subjected to regulation by cellular signaling. Taken together, our results identify NRIF3 as an interesting coregulator that possesses both transactivation and transrepression domains and/or functions. Collectively, the NRIF3 family of coregulators (which includes NRIF3 and its other isoforms) may play dual roles in mediating both positive and negative regulatory effects on gene expression.


Subject(s)
Nuclear Proteins/chemistry , Saccharomyces cerevisiae Proteins , Transcription, Genetic , Alternative Splicing , Amino Acid Sequence , Amino Acids/chemistry , DNA/metabolism , DNA-Binding Proteins , Dimerization , Fungal Proteins/metabolism , Gene Deletion , Gene Expression Regulation , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Two-Hybrid System Techniques
11.
Mol Cell Biol ; 21(7): 2298-311, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259580

ABSTRACT

Members of the type II nuclear hormone receptor subfamily (e.g., thyroid hormone receptors [TRs], retinoic acid receptors, retinoid X receptors [RXRs], vitamin D receptor, and the peroxisome proliferator-activated receptors) bind to their response sequences with or without ligand. In the absence of ligand, these DNA-bound receptors mediate different degrees of repression or silencing of gene expression which is thought to result from the association of their ligand binding domains (LBDs) with corepressors. Two related corepressors, N-CoR and SMRT, interact to various degrees with the LBDs of these type II receptors in the absence of their cognate ligands. N-CoR and SMRT have been proposed to act by recruiting class I histone deacetylases (HDAC I) through an association with Sin3, although they have also been shown to recruit class II HDACs through a Sin3-independent mechanism. In this study, we used a biochemical approach to identify novel nuclear factors that interact with unliganded full-length TR and RXR. We found that the DNA binding domains (DBDs) of TR and RXR associate with two proteins which we identified as PSF (polypyrimidine tract-binding protein-associated splicing factor) and NonO/p54(nrb). Our studies indicate that PSF is a novel repressor which interacts with Sin3A and mediates silencing through the recruitment of HDACs to the receptor DBD. In vivo studies with TR showed that although N-CoR fully dissociates in the presence of ligand, the levels of TR-bound PSF and Sin3A appear to remain unchanged, indicating that Sin3A can be recruited to the receptor independent of N-CoR or SMRT. RXR was not detected to bind N-CoR although it bound PSF and Sin3A as effectively as TR, and this association with RXR did not change with ligand. Our studies point to a novel PSF/Sin3-mediated pathway for nuclear hormone receptors, and possibly other transcription factors, which may fine-tune the transcriptional response as well as play an important role in mediating the repressive effects of those type II receptors which only weakly interact with N-CoR and SMRT.


Subject(s)
RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Mice , PTB-Associated Splicing Factor , Plasmids , Protein Binding , RNA Splicing , RNA-Binding Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Sin3 Histone Deacetylase and Corepressor Complex
12.
Mol Cell Biol ; 13(9): 5725-37, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8395010

ABSTRACT

The receptors for thyroid hormone (T3R), all-trans-retinoic acid (RAR), and 9-cis-retinoic acid (RXR) bind DNA response elements as homo- and heterodimers. The ligand-binding domains of these receptors contain nine conserved heptads proposed to play a role in dimerization. Mutant receptors with changes in the first or last hydrophobic amino acids in the highly conserved ninth heptad of chick T3R alpha [cT3R alpha(L365R) and cT3R(L372R)] and human RAR alpha (hRAR alpha) [hRAR(M377R) and hRAR(L384R)] reveal that this heptad is essential for certain heterodimeric interactions and for diverse functional activities. Without ligands, wild-type receptors form both homodimers and heterodimers, while these mutants form only homodimers. Surprisingly, the cognate ligand for each mutant enables heterodimer formation between cT3R(L365R) and RAR or RXR and between hRAR(M377R) and T3R or RXR. Both cT3R(L365R) and hRAR(M377R) mediate ligand-dependent transcriptional regulation. However, unlike the wild-type receptor, non-ligand-associated cT3R(L365R) does not suppress the basal activity of certain promoters containing thyroid hormone response elements, suggesting that this silencing effect of T3R is mediated by unliganded heterodimers of T3R and endogenous RXR or related factors. Heterodimerization is also necessary for the strong ligand-independent inhibition between T3R and RAR on a common response element, since the ninth-heptad mutants function as poor inhibitors. However, with a T3R-specific response element, hRAR(M377R) acts as a retinoic acid-dependent inhibitor of cT3R, indicating the importance of heterodimerization for this inhibition. Our studies also suggest that the ninth heptad is necessary for the dominant inhibition of wild-type T3Rs by mutant T3Rs, as has been found for the thyroid hormone-resistant syndrome in humans. Thus, the ninth heptad repeat is required for heterodimerization, suppression of basal promoter activity, and dominant negative effects of T3R and RAR. Lastly, the finding that cT3R(L365R) and hRAR(M377R) require ligands for heterodimer formation also raises the possibility that heterodimeric interactions are mediated by the ninth heptad without ligands but by a second region of these receptors with ligands.


Subject(s)
Carrier Proteins/chemistry , Receptors, Thyroid Hormone/chemistry , Transcription Factors , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , DNA Mutational Analysis , Gene Expression Regulation , Humans , Ligands , Macromolecular Substances , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Rats , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Retinoic Acid , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/genetics , Retinoid X Receptors , Sequence Alignment , Structure-Activity Relationship
13.
Mol Cell Biol ; 19(1): 864-72, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9858609

ABSTRACT

The mdm2 gene is positively regulated by p53 through a p53-responsive DNA element in the first intron of the mdm2 gene. mdm2 binds p53, thereby abrogating the ability of p53 to activate the mdm2 gene, and thus forming an autoregulatory loop of mdm2 gene regulation. Although the mdm2 gene is thought to act as an oncogene by blocking the activity of p53, recent studies indicate that mdm2 can act independently of p53 and block the G1 cell cycle arrest mediated by members of the retinoblastoma gene family and can activate E2F1/DP1 and the cyclin A gene promoter. In addition, factors other than p53 have recently been shown to regulate the mdm2 gene. In this article, we report that thyroid hormone (T3) receptors (T3Rs), but not the closely related members of the nuclear thyroid hormone/retinoid receptor gene family (retinoic acid receptor, vitamin D receptor, peroxisome proliferation activation receptor, or retinoid X receptor), regulate mdm2 through the same intron sequences that are modulated by p53. Chicken ovalbumin upstream promoter transcription factor I, an orphan nuclear receptor which normally acts as a transcriptional repressor, also activates mdm2 through the same intron region of the mdm2 gene. Two T3R-responsive DNA elements were identified and further mapped to sequences within each of the p53 binding sites of the mdm2 intron. A 10-amino-acid sequence in the N-terminal region of T3Ralpha that is important for transactivation and interaction with TFIIB was also found to be important for activation of the mdm2 gene response element. T3 was found to stimulate the endogenous mdm2 gene in GH4C1 cells. These cells are known to express T3Rs, and T3 is known to stimulate replication of these cells via an effect in the G1 phase of the cell cycle. Our findings, which indicate that T3Rs can regulate the mdm2 gene independently of p53, provide an explanation for certain known effects of T3 and T3Rs on cell proliferation. In addition, these findings provide further evidence for p53-independent regulation of mdm2 which could lead to the development of tumors from cells that express low levels of p53 or that express p53 mutants defective in binding to and activating the mdm2 gene.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Receptors, Thyroid Hormone/metabolism , Animals , COUP Transcription Factor I , Cell Line , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Introns , Ligands , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-mdm2 , Proto-Oncogenes , Rats , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Response Elements , Retinoid X Receptors , Structure-Activity Relationship , Transcription Factor TFIIB , Transcription Factors/metabolism , Triiodothyronine/pharmacology , Tumor Suppressor Protein p53/metabolism
14.
Mol Cell Biol ; 11(10): 5164-70, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1656224

ABSTRACT

The ability of a retinoic acid (RA) response element (RARE) in the phosphoenolpyruvate carboxykinase (PEPCK) gene promoter to mediate effects of either RA or thyroid hormone (T3) on gene expression was studied. Fusion gene constructs consisting of PEPCK promoter sequences ligated to the chloramphenicol acetyltransferase (CAT) reporter gene were used for this analysis. While T3 induced CAT expression to a small degree (about twofold) when such constructs were transiently transfected into H4IIE rat hepatoma cells, along with an expression vector encoding the alpha subtype of the T3 receptor (TR), this effect was mediated by promoter sequences distinct from the PEPCK RARE. Although TRs were capable of binding the PEPCK RARE in the form of putative monomers, dimers, and heterodimers with RA receptors (RARs), this element failed to mediate any positive effect of T3 on gene expression. In contrast, the PEPCK RARE mediated six- to eightfold induction of CAT expression by RA. When TRs were coexpressed along with RARs in transfected H4IIE cells, this RA induction was substantially blunted in a T3-independent manner. This inhibitory effect may be due to the binding of nonfunctional TRs or TR-RAR heterodimers to the PEPCK RARE. A model is proposed to explain the previously observed in vivo effects of T3 on PEPCK gene expression.


Subject(s)
Gene Expression Regulation/drug effects , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Promoter Regions, Genetic/physiology , Tretinoin/pharmacology , Triiodothyronine/pharmacology , Animals , Base Sequence , Carrier Proteins/genetics , Chloramphenicol O-Acetyltransferase/genetics , Humans , Models, Genetic , Molecular Sequence Data , Rats , Receptors, Retinoic Acid , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/biosynthesis , Sensitivity and Specificity , Tretinoin/antagonists & inhibitors , Tretinoin/metabolism , Triiodothyronine/metabolism , Tumor Cells, Cultured
15.
Mol Cell Biol ; 17(12): 7195-207, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9372952

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.


Subject(s)
Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Chickens , DNA/genetics , DNA/metabolism , Gene Expression Regulation , HeLa Cells , Humans , In Vitro Techniques , Ligands , Models, Biological , Mutation , Promoter Regions, Genetic , Rats , Transfection , Triiodothyronine/metabolism
16.
Mol Cell Biol ; 15(3): 1817-25, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7862171

ABSTRACT

The ligand-binding domains (LBDs) of the thyroid/retinoid receptor gene subfamily contain a series of heptad motifs important for dimeric interactions. This subfamily includes thyroid hormone receptors (T3Rs), all-trans retinoic acid (RA) receptors (RARs), 9-cis RA receptors (RARs and retinoid X receptors [RXRs]), the 1,25-dihydroxyvitamin D3 receptor (VDR), and the receptors that modulate the peroxisomal beta-oxidation pathway (PPARs). These receptors bind to their DNA response elements in vitro as heterodimers with the RXRs. Unliganded receptors in vivo, in particular the T3Rs, can mediate gene silencing and ligand converts these receptors into a transcriptionally active form. The in vivo interactions of these receptors with RXR were studied by using a GAL4-RXR chimera containing the yeast GAL4 DNA-binding domain and the LBD of RXR beta. GAL4-RXR activates transcription from GAL4 response elements in the presence of 9-cis RA. Unliganded T3R, which does not bind or activate GAL4 elements, represses the activation of GAL4-RXR by 9-cis RA in HeLa cells. However, addition of T3 alone leads to transcriptional activation. These findings suggest that T3R can repress or activate transcription while tethered to the LBD of GAL4-RXR and that heterodimerization can occur in vivo without stabilization by hormone response elements. Similar ligand-dependent activation was observed in HeLa cells expressing RAR, VDR, or PPAR and in GH4C1 cells from endogenous receptors. Replacement of the last 17 amino acids of the LBD of RXRbeta with the 90-amino-acid transactivating domain of the herpes simplex virus VP16 protein leads to a GAL4 constitutive activator that is repressed by wild-type T3R but not by a ninth heptad mutant that does not form heterodimers. This finding suggests that the ninth heptad or T3R is important for gene silencing and that the LBD of RXR does not exhibit silencing activity. This conclusion was verified with GAL4-LBD chimeras and with wild-type receptors in assays using appropriate response elements. These studies indicate that the LBD has diverse functional roles in gene regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression , Multigene Family , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Saccharomyces cerevisiae Proteins , Transcriptional Activation , Animals , Binding Sites , Cell Line , Cells, Cultured , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Binding Proteins/genetics , Fungal Proteins/metabolism , HeLa Cells , Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/metabolism , Humans , Ligands , Molecular Sequence Data , Pituitary Gland/metabolism , Protein Multimerization , RNA, Messenger/metabolism , Rats , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/metabolism , Retinoid X Receptors , Saccharomyces cerevisiae/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Triiodothyronine/pharmacology
17.
Mol Cell Biol ; 19(10): 7191-202, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490654

ABSTRACT

Many nuclear receptors are capable of recognizing similar DNA elements. The molecular event(s) underlying the functional specificities of these receptors (in regulating the expression of their native target genes) is a very important issue that remains poorly understood. Here we report the cloning and analysis of a novel nuclear receptor coactivator (designated NRIF3) that exhibits a distinct receptor specificity. Fluorescence microscopy shows that NRIF3 localizes to the cell nucleus. The yeast two-hybrid and/or in vitro binding assays indicated that NRIF3 specifically interacts with the thyroid hormone receptor (TR) and retinoid X receptor (RXR) in a ligand-dependent fashion but does not bind to the retinoic acid receptor, vitamin D receptor, progesterone receptor, glucocorticoid receptor, or estrogen receptor. Functional experiments showed that NRIF3 significantly potentiates TR- and RXR-mediated transactivation in vivo but has little effect on other examined nuclear receptors. Domain and mutagenesis analyses indicated that a novel C-terminal domain in NRIF3 plays an essential role in its specific interaction with liganded TR and RXR while the N-terminal LXXLL motif plays a minor role in allowing optimum interaction. Computer modeling and subsequent experimental analysis suggested that the C-terminal domain of NRIF3 directly mediates interaction with liganded receptors through an LXXIL (a variant of the canonical LXXLL) module while the other part of the NRIF3 protein may still play a role in conferring its receptor specificity. Identification of a coactivator with such a unique receptor specificity may provide new insight into the molecular mechanism(s) of receptor-mediated transcriptional activation as well as the functional specificities of nuclear receptors.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Cell Compartmentation , Computer Simulation , Gene Library , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Retinoid X Receptors , Sequence Analysis, DNA , Two-Hybrid System Techniques
18.
Mol Cell Biol ; 14(9): 5756-65, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065310

ABSTRACT

The ligand-binding domains of thyroid hormone (L-triiodothyronine [T3]) receptors (T3Rs), all-trans retinoic acid (RA) receptors (RARs), and 9-cis RA receptors (RARs and RXRs) contain a series of heptad motifs thought to be important for dimeric interactions. Using a chimera containing amino acids 120 to 392 of chicken T3R alpha (cT3R alpha) positioned between the DNA-binding domain of the yeast GAL4 protein and the potent 90-amino-acid transactivating domain of the herpes simplex virus VP16 protein (GAL4-T3R-VP16), we provide functional evidence that binding of ligand releases T3Rs and RARs from an inhibitory cellular factor. GAL4-T3R-VP16 does not bind T3 and does not activate transcription from a GAL4 reporter when expressed alone but is able to activate transcription when coexpressed with unliganded T3R or RAR. This activation is reversed by T3 or RA, suggesting that these receptors compete with GAL4-T3R-VP16 for a cellular inhibitor and that ligand reverses this effect by dissociating T3R or RAR from the inhibitor. A chimera containing the entire ligand-binding domain of cT3R alpha (amino acids 120 to 408) linked to VP16 [GAL4-T3R(408)-VP16] is activated by unliganded receptor as well as by T3. In contrast, GAL4-T3R containing the amino acid 120 to 408 ligand-binding region without the VP16 domain is activated only by T3. The highly conserved ninth heptad, which is involved in heterodimerization, appears to participate in the receptor-inhibitor interaction, suggesting that the inhibitor is a related member of the receptor gene family. In striking contrast to T3R and RAR, RXR activates GAL4-T3R-VP16 only with its ligand, 9-cis RA, but unliganded RXR does not appear to be the inhibitor suggested by these studies. Further evidence that an orphan receptor may be the inhibitor comes from our finding that COUP-TF inhibits activation of GAL4-T3R-VP16 by unliganded T3R and the activation of GAL4-T3R by T3. These and other results suggest that an inhibitory factor suppresses transactivation by the T3Rs and RARs while these receptors are bound to DNA and that ligands act, in part, by inactivating or promoting dissociation of a receptor-inhibitor complex.


Subject(s)
Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/antagonists & inhibitors , Animals , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Herpes Simplex Virus Protein Vmw65/physiology , Humans , In Vitro Techniques , Ligands , Macromolecular Substances , Rats , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Triiodothyronine/pharmacology
19.
Mol Cell Biol ; 15(8): 4507-17, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7623841

ABSTRACT

The effects of the thyroid hormone (3,5,3'-triiodo-L-thyronine [T3]) on gene transcription are mediated by nuclear T3 receptors (T3Rs). alpha- and beta-isoform T3Rs (T3R alpha and -beta) are expressed from different genes and are members of a superfamily of ligand-dependent transcription factors that also includes the receptors for steroid hormones, vitamin D, and retinoids. Although T3 activates transcription by mediating a conformational change in the C-terminal approximately 220-amino-acid ligand-binding domain (LBD), the fundamental mechanisms of T3R-mediated transcriptional activation remain to be determined. We found that deletion of the 50-amino-acid N-terminal A/B domain of chicken T3R alpha (cT3R alpha) decreases T3-dependent stimulation of genes regulated by native thyroid hormone response elements about 10- to 20-fold. The requirement of the A/B region for transcriptional activation was mapped to amino acids 21 to 30, which contain a cluster of five basic amino acids. The A/B region of cT3R alpha is not required for T3 binding or for DNA binding of the receptor as a heterodimer with retinoid X receptor. In vitro binding studies indicate that the N-terminal region of cT3R alpha interacts efficiently with TFIIB and that this interaction requires amino acids 21 to 30 of the A/B region. In contrast, the LBD interacts poorly with TFIIB. The region of TFIIB primarily involved in the binding of cT3R alpha includes an amphipathic alpha helix contained within residues 178 to 201. Analysis using a fusion protein containing the DNA-binding domain of GAL4 and the entire A/B region of cT3R alpha suggests that this region does not contain an intrinsic activation domain. These and other studies indicate that cT3R alpha mediates at least some of its effects through TFIIB in vivo and that the N-terminal region of DNA-bound cT3R alpha acts to recruit and/or stabilize the binding of TFIIB to the transcription complex. T3 stimulation could then result from ligand-mediated changes in the LBD which may lead to the interaction of other factors with cT3R alpha, TFIIB, and/or other components involved in the initiation of transcription.


Subject(s)
Gene Expression Regulation , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Triiodothyronine/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cells, Cultured , Conserved Sequence , DNA Mutational Analysis , Humans , Ligands , Molecular Sequence Data , Protein Binding , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Transcription Factor TFIIB
20.
Pharmacol Ther ; 33(2-3): 317-32, 1987.
Article in English | MEDLINE | ID: mdl-3310036

ABSTRACT

Photoaffinity label probes of iodothyronines can interact with nuclear receptors in intact cells and in solubilized receptor preparations. These probes have certain advantages over a chemical affinity label in analyzing receptor structure. First, a photoaffinity label probe covalently cross-links only after photoactivation. Therefore, it is possible to demonstrate with appropriate competitive inhibition studies that the photoaffinity label probe associates with the receptor in question. Secondly, since cross-linking only occurs after photolysis, it is possible to adjust the concentration of the photoaffinity label to maximize association with "specific" binding sites relative to "non-specific" associations prior to covalent linkage by photoactivation. The different [125I]iodothyronine-PAL analogues may be useful as probes of the thyroid hormone receptor binding domain since PAL compounds with different affinities for receptor may photocouple to different receptor residues within or proximate to the hormone binding region. These probes may also be useful as an adjunct to receptor purification and in probing the organization of the receptor in chromatin. Lastly, they may provide insights into possible alterations of receptor structure in patients with partial end organ resistance to thyroid hormone (Refetoff et al., 1967; Eil et al., 1982).


Subject(s)
Receptors, Thyroid Hormone/analysis , Affinity Labels , Animals , Humans , Photochemistry
SELECTION OF CITATIONS
SEARCH DETAIL