Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Mol Endocrinol ; 27(2): 211-27, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564604

ABSTRACT

The vitamin D receptor (VDR) stimulates transcription as a 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3))-activated heterodimer with retinoid X receptor (RXR). RXR also forms homodimers to mediate 9-cis retinoic acid (9-cis RA)-induced gene expression. Both receptors possess a C-terminal hormone-dependent activation function-2 (AF-2), a highly conserved region that binds coactivators to transduce the transcriptional signal. By replacing single amino acids within the AF-2 of human RXR alpha (hRXR alpha) or mouse RXR beta (mRXR beta), the contribution of these residues to transactivation by the RXR-VDR heterodimer and the RXR-RXR homodimer was evaluated. In 9-cis RA-responsive homodimers, the second and fourth positions of the AF-2 (leucine and glutamate respectively) are essential. However, in the context of an RXR-VDR heterodimer activated by 1,25(OH)(2)D(3), alteration of these two RXR residues has little effect. Instead, AF-2 residues located towards the C-terminus, such as the penultimate position (L455 in hRXR alpha or L441 in mRXR beta), are crucial for RXR-VDR heterodimers. Indeed, L455A mutant RXR exerts a dominant negative effect on RXR-VDR transcriptional responsiveness to 1,25(OH)(2)D(3). Further experiments with a mutant hRXR alpha (F313A) which elicits 9-cis RA-independent transactivation as a homodimer demonstrate that, when heterodimerized with VDR, this RXR mutant is incapable of activating the RXR-VDR heterocomplex in the absence of the VDR ligand. Taken together, these results indicate that RXR is a subordinate, yet essential transcriptional partner in RXR-VDR-mediated activation of gene expression. Furthermore, a functional switch in RXR AF-2 signaling occurs between RXR residues in the homodimeric versus the heterodimeric states, likely reflecting different interactions between subregions of the AF-2 and coactivator(s).


Subject(s)
Receptors, Calcitriol/genetics , Receptors, Retinoic Acid/genetics , Transcription Factors/genetics , Alitretinoin , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Dimerization , Humans , Mice , Models, Biological , Mutation , Protein Structure, Tertiary , Rats , Receptors, Calcitriol/chemistry , Receptors, Retinoic Acid/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Retinoid X Receptors , Signal Transduction , Transcription Factors/chemistry , Transcriptional Activation/drug effects , Transfection , Tretinoin/pharmacology
2.
Mol Cell Endocrinol ; 177(1-2): 145-59, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11377830

ABSTRACT

The functional significance of two unlinked human vitamin D receptor (hVDR) gene polymorphisms was evaluated in twenty human fibroblast cell lines. Genotypes at both a Fok I restriction site (F/f) in exon II and a singlet (A) repeat in exon IX (L/S) were determined, and relative transcription activities of endogenous hVDR proteins were measured using a transfected, 1,25-dihydroxyvitamin D(3)-responsive reporter gene. Observed activities ranged from 2--100-fold induction by hormone, with higher activity being displayed by the F and the L biallelic forms. Only when genotypes at both sites were considered simultaneously did statistically significant differences emerge. Moreover, the correlation between hVDR activity and genotype segregated further into clearly defined high and low activity groups with similar genotypic distributions. These results not only demonstrate functional relevance for both the F/f and L/S common polymorphisms in hVDR, but also provide novel evidence for a third genetic variable impacting receptor potency.


Subject(s)
Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Alleles , Cell Line , Fibroblasts/cytology , Gene Frequency , Genes, Reporter , Genotype , Humans , Polymorphism, Genetic/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptional Activation/genetics , Transfection
3.
Mol Endocrinol ; 14(3): 401-20, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707958

ABSTRACT

The human vitamin D receptor (hVDR) is a ligand-regulated transcription factor that mediates the actions of the 1,25-dihydroxyvitamin D3 hormone to effect bone mineral homeostasis. Employing mutational analysis, we characterized Arg-18/Arg-22, hVDR residues immediately N-terminal of the first DNA binding zinc finger, as vital for contact with human basal transcription factor IIB (TFIIB). Alteration of either of these basic amino acids to alanine also compromised hVDR transcriptional activity. In contrast, an artificial hVDR truncation devoid of the first 12 residues displayed both enhanced interaction with TFIIB and transactivation. Similarly, a natural polymorphic variant of hVDR, termed F/M4 (missing a FokI restriction site), which lacks only the first three amino acids (including Glu-2), interacted more efficiently with TFIIB and also possessed elevated transcriptional activity compared with the full-length (f/M1) receptor. It is concluded that the functioning of positively charged Arg-18/Arg-22 as part of an hVDR docking site for TFIIB is influenced by the composition of the adjacent polymorphic N terminus. Increased transactivation by the F/M4 neomorphic hVDR is hypothesized to result from its demonstrated enhanced association with TFIIB. This proposal is supported by the observed conversion of f/M1 hVDR activity to that of F/M4 hVDR, either by overexpression of TFIIB or neutralization of the acidic Glu-2 by replacement with alanine in f/M1 hVDR. Because the f VDR genotype has been associated with lower bone mineral density in diverse populations, one factor contributing to a genetic predisposition to osteoporosis may be the F/f polymorphism that dictates VDR isoforms with differential TFIIB interaction.


Subject(s)
Protein Isoforms/physiology , Receptors, Calcitriol/physiology , Transcription Factors/metabolism , Transcriptional Activation , Amino Acid Sequence , Amino Acid Substitution , Animals , Bone Density/genetics , COS Cells/drug effects , Calcitriol/pharmacology , Chlorocebus aethiops , DNA/metabolism , Fibroblasts/metabolism , Genetic Predisposition to Disease , Genotype , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Osteoporosis/genetics , Polymorphism, Genetic , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/genetics , Transcription Factor TFIIB , Zinc Fingers/physiology
4.
Biochem Biophys Res Commun ; 267(3): 813-9, 2000 Jan 27.
Article in English | MEDLINE | ID: mdl-10673374

ABSTRACT

Human vitamin D receptor (hVDR) fused to glutathione S-transferase was utilized to detect a VDR-interacting protein (VIP) of approximately 170 kDa. VIP(170) is expressed in osteoblast-like ROS 17/2.8 cells and, to a lesser extent, in COS-7 and HeLa cells. VIP(170) may be a coactivator because it interacts only with 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) ligand-bound hVDR and because a mutation (E420A) in the activation function-2 (AF-2) of hVDR abolishes both receptor-mediated transactivation and VIP(170) binding. Unlike L254G hVDR, a heterodimerization mutant with an intact AF-2, the E420A mutant is only partially attenuated in its association with the retinoid X receptor (RXR) DNA-binding partner. Finally, the ability of overexpressed hVDR to squelch glucocorticoid receptor-mediated transactivation is lost in both the L254G and E420A mutants. These results suggest that several protein-protein interactions, including VDR association with RXR and VIP(170), are required for stabilization of a multimeric complex that transduces the signal for 1,25(OH)(2)D(3)-elicited transactivation.


Subject(s)
Nuclear Proteins/metabolism , Osteoblasts/metabolism , Receptors, Retinoic Acid/metabolism , Trans-Activators , Transcription Factors/metabolism , Amino Acid Substitution , Animals , COS Cells , Calcitriol/pharmacology , Dexamethasone/pharmacology , Dimerization , HeLa Cells , Humans , Mediator Complex , Molecular Weight , Mutagenesis, Site-Directed , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Osteosarcoma , Recombinant Fusion Proteins/biosynthesis , Retinoid X Receptors , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tumor Cells, Cultured
5.
Biochemistry ; 38(49): 16347-58, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10587460

ABSTRACT

The vitamin D receptor (VDR) binds 1,25-dihydroxyvitamin D(3) and mediates its actions on gene transcription by heterodimerizing with retinoid X receptors (RXRs) on direct repeat (DR+3) vitamin D responsive elements (VDREs) located in target genes. The VDRE binding function of VDR has been primarily ascribed to the zinc finger region (residues 24-87). To define the minimal VDRE binding domain for human VDR (hVDR), a series of C-terminally truncated hVDR mutants (Delta134, Delta113, Delta102, Delta90, Delta84, Delta80, and Delta60) was generated and expressed in bacteria. Only the Delta134 and Delta113 mutants bound the VDRE (predominantly as monomers), suggesting that, in addition to the conserved zinc finger region of hVDR, as many as 25 amino acids in a C-terminal extension (CTE) participate in DNA binding. Site-directed mutagenesis of conserved charged residues in full-length hVDR was then performed to dissect the functional significance of the CTE (residues 88-112) in the context of the complete hVDR-RXR-VDRE interaction. Functional assays revealed that E98K/E99K, R102A/K103A/R104A, and K109A/R110A/K111A mutant hVDRs possessed dramatically reduced DNA binding and transcriptional activities, whereas distinct point mutants, such as K103A, bound to DNA normally but lacked transcriptional activity. Therefore, the boundary for the minimal DNA-binding domain in hVDR extends C-terminal of the zinc fingers to Lys-111, with clusters of highly conserved charged amino acids playing a crucial role in binding to the DR+3 element. Further, individual residues in this region (e.g., Lys-103) may lie on the opposing face of a DNA-binding alpha-helix, where they could contact transcriptional coactivators or basal transcription factors.


Subject(s)
DNA-Binding Proteins/chemistry , Peptide Fragments/chemistry , Receptors, Calcitriol/chemistry , Trans-Activators/chemistry , Zinc Fingers , Amino Acid Sequence , Amino Acid Substitution/genetics , Conserved Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dimerization , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/physiology , Point Mutation , Protein Structure, Tertiary/genetics , Receptors, Calcitriol/genetics , Receptors, Calcitriol/physiology , Sequence Deletion , Trans-Activators/genetics , Trans-Activators/physiology , Zinc Fingers/genetics
6.
J Cell Biochem ; 75(3): 462-80, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10536369

ABSTRACT

The vitamin D receptor (VDR) is a transcription factor believed to function as a heterodimer with the retinoid X receptor (RXR). However, it was reported [Schräder et al., 1994] that, on putative vitamin D response elements (VDREs) within the rat 9k and mouse 28k calcium binding protein genes (rCaBP 9k and mCaBP 28k), VDR and thyroid hormone receptor (TR) form heterodimers that transactivate in response to both 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) and triiodothyronine (T(3)). We, therefore, examined associations of these receptors on the putative rCaBP 9k and mCaBP 28k VDREs, as well as on established VDREs from the rat osteocalcin (rOC) and mouse osteopontin (mOP) genes, plus the thyroid hormone response element (TRE) from the rat myosin heavy chain (rMHC) gene. In gel mobility shift assays, we found no evidence for VDR-TR heterodimer interaction with any tested element. Further, employing these hormone response elements linked to reporter genes in transfected cells, VDR and TR mediated responses to their cognate ligands only from the rOC/mOP and rMHC elements, respectively, while the CaBP elements were unresponsive to any combination of ligand(s). Utilizing the rOC and mOP VDREs, two distinct repressive actions of TR on VDR-mediated signaling were demonstrated: a T(3)-independent action, presumably via direct TR-RXR competition for DNA binding, and a T(3)-dependent repression, likely by diversion of limiting RXR from VDR-RXR toward the formation of TR-RXR heterodimers. The relative importance of these two mechanisms differed in a response element-specific manner. These results may provide a partial explanation for the observed association between hyperthyroidism and bone demineralization/osteoporosis.


Subject(s)
DNA/metabolism , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , COS Cells , DNA, Complementary/genetics , Dimerization , Humans , In Vitro Techniques , Ligands , Mice , Models, Biological , Protein Structure, Quaternary , Rats , Receptor Cross-Talk , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/genetics , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/chemistry , Receptors, Thyroid Hormone/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoid X Receptors , Transcription Factors/chemistry , Transcription Factors/genetics , Transcriptional Activation
7.
Mol Endocrinol ; 10(12): 1617-31, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8961271

ABSTRACT

Hereditary hypocalcemic vitamin D-resistant rickets is attributable to defects in the nuclear receptor for 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3]. Two novel point mutations (I314S and R391C) identified in the hormone-binding domain of the human vitamin D receptor (VDR) from patients with hereditary hypocalcemic vitamin D-resistant rickets confer the receptor with sharply reduced 1,25-(OH)2D3-dependent transactivation. These natural mutations, especially R391C, also lead to a second specific consequence, namely impaired heterodimeric interaction with retinoid X receptor (RXR). While the transactivation ability of the I314S mutant can be largely restored by providing excess 1,25-(OH)2D3, R391C activity is more effectively restored with exogenous RXR. These observations are reflected also in the clinical course of each patient: the patient bearing the I314S mutation showed a nearly complete cure with pharmacological doses of a vitamin D derivative, whereas the patient bearing R391C responded only partially to such therapy. Further tests with patient fibroblasts and transfected cells show that the activity of the I314S VDR mutant is augmented somewhat by added RXR, while transactivation by the R391C mutant is best corrected by RXR in the presence of excess hormone. Thus, the effects of hormone vs. RXR in bolstering these mutant VDRs, such that they mediate efficient transactivation, are not entirely separable. The unique properties of these genetically altered receptors establish a new subclass of natural human VDR mutants that illustrate, in vivo, the importance of both 1,25-(OH)2D3 binding and heterodimerization with RXR in VDR action.


Subject(s)
Calcitriol/pharmacology , Point Mutation , Receptors, Calcitriol/drug effects , Receptors, Calcitriol/genetics , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , COS Cells/metabolism , Calcitriol/metabolism , Calcitriol/pharmacokinetics , Child, Preschool , Cloning, Molecular , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Drug Resistance/genetics , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Genes, Dominant , Humans , Hypocalcemia/drug therapy , Hypocalcemia/genetics , Infant , Metabolic Diseases/genetics , Molecular Sequence Data , Phenotype , Protein Conformation , Receptors, Calcitriol/chemistry , Receptors, Retinoic Acid/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoid X Receptors , Transcription Factors/chemistry , Transcriptional Activation/drug effects , Transfection
8.
J Biol Chem ; 271(9): 5143-9, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617794

ABSTRACT

The significance of conserved cysteines at positions 288, 337, and 369 in the hormone binding domain of the human vitamin D receptor was evaluated by individual site-directed mutagenesis to glycine. Neither nuclear localization nor heterodimerization with retinoid X receptors in binding to the vitamin D-responsive element was appreciably affected by altering these cysteines, but vitamin D hormone (1,25-(OH)2D3) activated transcription was compromised significantly in the C288G and C337G mutants. Only the C288G mutant displayed depressed (3-fold) 1,25-(OH)2D3 ligand binding affinity at 4 degrees C, in vitro, although at elevated temperatures (23-37 degrees C), ligand binding was attenuated severely in C288G, moderately in C337G and very mildly in C369G. The degree of impairment of ligand binding at physiologic temperatures correlated with the requirement for increased concentrations of 1,25-(OH)2D3 ligand to maximally stimulate transcriptional activity in co-transfected COS-7 cells. Thus cysteine 288 and, to a lesser extent, cysteine 337 are important for high affinity hormone binding to the vitamin D receptor, which ultimately leads to ligand-dependent transcriptional activation.


Subject(s)
Calcitriol/pharmacology , Cysteine , Liver/metabolism , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Calcitriol/metabolism , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Conserved Sequence , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Point Mutation , Protein Multimerization , Rats , Receptors, Calcitriol/isolation & purification , Receptors, Retinoic Acid/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Retinoid X Receptors , Thermodynamics , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Transfection
9.
J Cell Biochem ; 58(3): 315-27, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7593254

ABSTRACT

Organ culture of 19-day-old chick embryo duodena was utilized to evaluate the mechanism of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3)-dependent calbindin-D28k (CaBP) expression. Duodenal CaBP and 1,25(OH)2D3 receptor (VDR) expression were assessed by Western blot analysis, while CaBP and VDR mRNA levels were determined by Northern blot analysis. In untreated duodena, both VDR protein and mRNA were present, while CaBP protein and mRNA were undetectable. Treatment of cultured duodena with 25 nM 1,25(OH)2D3 resulted in detectable CaBP mRNA after 4 h which continued to increase during a 24 h time period. Under these conditions, localization of [3H-1 beta]1 alpha,25(OH)2D3 in duodenal chromatin is rapid (< or = 30 min). Thus, the delayed accumulation of detectable CaBP mRNA cannot be explained by slow nuclear binding of 1,25(OH)2D3. The inclusion of 1.6 microM actinomycin D in the organ culture partially inhibited the 1,25(OH)2D3-regulated increase in CaBP mRNA, which implies that there is a transcriptional component involved in the increased CaBP mRNA levels. Similarly, quantitative polymerase chain reaction studies allowed the detection of CaBP pre-mRNA and mRNA sequences 1 h after hormone treatment, suggesting that CaBP gene transcription is initiated rapidly. Treatment of cultures with 36 microM cycloheximide 1 h prior to 1,25(OH)2D3 addition resulted in superinduction of VDR mRNA levels but sharply reduced CaBP steady-state mRNA levels. This dramatic reduction in CaBP mRNA reveals that 1,25(OH)2D3-mediated CaBP expression is dependent on ongoing protein synthesis. Thus, we propose that a labile auxiliary protein or other cofactor, which may or may not be 1,25(OH)2D3-dependent, is necessary for 1,25(OH)2D3-mediated CaBP gene transcription in chick duodena.


Subject(s)
Calcitriol/metabolism , Duodenum/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , S100 Calcium Binding Protein G/genetics , Animals , Calbindins , Calcitriol/pharmacology , Chick Embryo , Chromatin/metabolism , Chromosome Mapping , Duodenum/drug effects , Gene Expression Regulation/drug effects , Kinetics , Organ Culture Techniques , Polymerase Chain Reaction , Protein Biosynthesis/drug effects , Receptors, Calcitriol/metabolism , Transcription, Genetic/drug effects
10.
J Steroid Biochem Mol Biol ; 53(1-6): 583-94, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7626514

ABSTRACT

The nuclear vitamin D receptor (VDR) binds the 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) hormone with high affinity and elicits its actions to stimulate gene expression in target cells by binding to the vitamin D-responsive element (VDRE). VDREs in such positively controlled genes as osteocalcin, osteopontin, beta 3 integrin and vitamin D-24-OHase are direct hexanucleotide repeats with a spacer of three nucleotides. The present studies of VDR/VDRE interaction utilized full-length human vitamin D receptor (hVDR) that was overexpressed in E. coli, purified to near homogeneity (> 95%), and its authenticity confirmed by demonstrating high affinity hormone binding and reactivity to monoclonal antibody 9A7 gamma. The expressed hVDR displays strict dependence on the family of retinoid X receptors (RXRs) for binding to the vitamin D-responsive element (VDRE) in the rat osteocalcin gene. Similar overexpression in E. coli of the DNA binding domain (delta 134), containing only residues 4-133 of hVDR, generated a receptor species that possesses intrinsic DNA binding activity. Both full-length and delta 134 hVDRs retain similar DNA binding specificities when tested with several natural hormone responsive elements, indicating that the N-terminal zinc finger region determines hVDR-DNA sequence selectivity. The C-terminal region of the molecule is required for hormone binding and confers the receptor with the property of very high affinity DNA binding, via heterodimerization between hVDR and RXR. A natural ligand for the RXR co-receptor, 9-cis retinoic acid, suppresses both VDR-RXR binding to the VDRE and 1,25(OH)2D3 stimulated transcription, indicating that 9-cis retinoic acid recruits RXR away from VDR to instead form RXR homodimers.


Subject(s)
Calcitriol/physiology , Receptors, Calcitriol/genetics , Base Sequence , Cloning, Molecular , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Escherichia coli , Humans , In Vitro Techniques , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Receptors, Calcitriol/chemistry , Recombinant Proteins , Regulatory Sequences, Nucleic Acid , Structure-Activity Relationship
11.
Mol Endocrinol ; 8(2): 159-72, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8170472

ABSTRACT

The human vitamin D receptor (hVDR) requires another nuclear protein(s), designated receptor auxiliary factor (RAF), for optimal binding to the vitamin D-responsive element (VDRE). To determine the region in hVDR required to form a heterodimer with RAF on the VDRE, mutant hVDR cDNAs were constructed by site-directed mutagenesis and transfected into COS-7 cells. A truncated hVDR, lacking 25 C-terminal amino acids (delta 403-427), showed complex production in combination with endogenous RAF in COS-7 cells. Complex development was markedly enhanced by adding a rat liver nuclear fraction, which contains RAF activity, or either the alpha or beta form of the retinoid-X receptor, which has been reported to be closely related or identical to RAF. In contrast, either a C-terminal truncation of 46 amino acids (delta 382-427) or single point mutations at lysine-382, methionine-383, glutamine-385, or leucine-390 dramatically reduced the ability of hVDR to heterodimerize with RAF. Binding of 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] hormone was undetectable in delta 382-427 truncated hVDR, whereas the delta 403-427 mutant hVDR exhibited significant 1,25-(OH)2D3 ligand binding, although the dissociation constant was approximately 10-fold higher than that of the wild-type receptor. Surprisingly, the delta 403-427 mutant hVDR did not mediate measurable transcriptional activation in cotransfection experiments with a VDRE-GH reporter gene construct. These results indicate that hVDR residues between cysteine-403 and serine-427 are required for very high affinity 1,25-(OH)2D3 ligand binding and transcriptional activation, but are not involved in heterodimerization. The region of hVDR between lysine-382 and arginine-402, probably the domain containing heptad 9, plays an essential role in the heterodimerization of hVDR with RAF. However, based upon additional point mutagenesis experiments, it is likely that other regions of the hormone-binding domain, such as that including heptad 4 (leucine-325 to leucine-332), also contribute to the protein-protein interactions required for the high affinity, specific binding of hVDR to the VDRE.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Calcitriol/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Amino Acid Sequence , Animals , Base Sequence , Calcitriol/metabolism , Humans , Liver , Molecular Sequence Data , Mutation/physiology , Point Mutation , Rats , Receptors, Calcitriol/chemistry , Transcription, Genetic/physiology
12.
Biochem Biophys Res Commun ; 197(2): 478-85, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8267583

ABSTRACT

We report here that highly purified human vitamin D receptor (hVDR) derived from E. coli or baculovirus expression systems does not exhibit saturable, high affinity 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) ligand binding when these preparations alone are analyzed. Inclusion of rat liver nuclear extract, which does not itself contain detectable 1,25(OH)2D3 binding activity, is required to endow hVDR isolated from bacterial or insect cells with the property of high affinity hormone binding (Kd = 0.13-0.22 nM). This observation should facilitate the valid assay of 1,25(OH)2D3 binding activity and kinetics in samples of overexpressed hVDR. Moreover, since rat liver nuclear extract contains retinoid X receptors and possibly other auxiliary factors capable of forming heterodimers with hVDR that in turn associate with vitamin D responsive elements, we hypothesize that like DNA binding, 1,25(OH)2D3 binding to hVDR requires the cooperation of a co-receptor or some uncharacterized receptor activating/stabilizing factor.


Subject(s)
Calcitriol/metabolism , Cell Nucleus/metabolism , Liver/metabolism , Receptors, Calcitriol/metabolism , Tissue Extracts/metabolism , Animals , Baculoviridae/genetics , Cell Line , Cloning, Molecular , DNA-Binding Proteins/metabolism , Escherichia coli/metabolism , Humans , Kinetics , Moths , Osteocalcin/genetics , Rats , Receptors, Calcitriol/biosynthesis , Receptors, Calcitriol/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
13.
Mol Cell Biol ; 13(9): 5907-17, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8395017

ABSTRACT

The vitamin D receptor (VDR) binds the vitamin D-responsive element (VDRE) as a heterodimer with an unidentified receptor auxiliary factor (RAF) present in mammalian cell nuclear extracts. VDR also interacts with the retinoid X receptors (RXRs), implying that RAF may be related to the RXRs. Here we demonstrate that highly purified HeLa cell RAF contained RXR beta immunoreactivity and that both activities copurified and precisely coeluted in high-resolution hydroxylapatite chromatography. Furthermore, an RXR beta-specific antibody disrupted VDR-RAF-VDRE complexes in mobility shift assays. These data strongly indicate that HeLa RAF is highly related to or is identical to RXR beta. Consequently, the effect of the 9-cis retinoic acid ligand for RXRs was examined in 1,25-dihydroxyvitamin D3 [1,25(OH)2D3]-activated gene expression systems. Increasing concentrations of 9-cis retinoic acid (1 nM to 1 microM) markedly reduced 1,25(OH)2D3-dependent accumulation of osteocalcin mRNA in osteoblast-like ROS 17/2.8 cells. All-trans retinoic acid also interfered with vitamin D responsiveness, but it was consistently less potent than the 9-cis isomer. Transient transfection studies revealed that attenuation by 9-cis retinoic acid was at the transcriptional level and was mediated through interactions at the osteocalcin VDRE. Furthermore, overexpression of both RXR beta and RXR alpha augmented 1,25(OH)2D3 responsiveness in transient expression studies. Direct analysis of VDRE binding in mobility shift assays demonstrated that heteromeric interactions between VDR and RXR were enhanced by 1,25(OH)2D3 and were not affected appreciably by 9-cis retinoic acid, except that inhibition was observed at high retinoid concentrations. These data suggest a regulatory mechanism for osteocalcin gene expression that involves 1,25(OH)2D3-induced heterodimerization of VDR and unliganded RXR. 9-cis retinoic acid may attenuate 1,25(OH)2D3 responsiveness by diverting RXRs away from VDR-mediated transcription and towards other RXR-dependent transcriptional pathways.


Subject(s)
Calcitriol/pharmacology , Gene Expression Regulation , Osteocalcin/genetics , Receptors, Cell Surface/physiology , Receptors, Retinoic Acid , Receptors, Steroid/physiology , Transcription Factors , Tretinoin/pharmacology , Animals , Calcitriol/antagonists & inhibitors , Gene Expression , HeLa Cells , Humans , Ligands , Promoter Regions, Genetic , RNA, Messenger/genetics , Rats , Receptors, Calcitriol , Recombinant Proteins/metabolism , Retinoid X Receptors
14.
J Biol Chem ; 268(20): 15118-26, 1993 Jul 15.
Article in English | MEDLINE | ID: mdl-8392065

ABSTRACT

We have reported previously that the human vitamin D receptor (hVDR) is selectively phosphorylated by protein kinase C-beta (PKC-beta), in vitro, on a serine residue in the sequence RRS51MKRK, which is located between the two zinc fingers of hVDR and is potentially important to its transacting function (Hsieh, J.-C., Jurutka, P.W., Galligan, M.A., Terpening, C.M., Haussler, C.A., Samuels, D.S., Shimizu, Y., Shimizu, N., and Haussler, M.R. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 9315-9319). In the present experiments we evaluated this phosphorylation event using a series of hVDR mutants in which serine 51 or its flanking residues were modified. Alteration of serine 51 to a non-phosphorylatable residue resulted in an approximately 60% reduction in basal hVDR phosphorylation in intact cells but did not diminish 1,25-dihydroxyvitamin D3-stimulated phosphorylation. Such mutations also abolished subsequent phosphorylation of immunoprecipitated hVDR by purified PKC-beta, in vitro, as did replacement of basic residues on either side of serine 51. Mutation of serine 51 to glycine (S51G) or to aspartic acid (S51D), as well as altering the basic residues flanking serine 51, abolished the interaction of hVDR with the vitamin D-responsive element (VDRE) as monitored by gel mobility shift analysis. Thus, we conclude that unmodified serine 51 and its surrounding basic residues are crucial not only for PKC-beta substrate recognition but also for the optimal VDRE binding of native hVDR. In transactivation assays, S51G and S51D possessed only 35 and 10% of wild-type hVDR activity, respectively. Mutation of serine 51 to threonine (S51T) restored phosphorylation by PKC-beta, in vitro, to about 40% of wild-type and transactivation to 45% of that of wild-type hVDR. Alteration of serine 51 to alanine, which is the residue in the corresponding position of the glucocorticoid, progesterone, mineral-ocorticoid, and androgen receptors, eliminated PKC-beta phosphorylation but completely preserved the specific DNA binding activity and transactivation capacity of hVDR. Thus, phosphorylation of hVDR at serine 51 is not required for either VDRE binding or transactivation. Finally, incubation of Escherichia coli-expressed hVDR with PKC-beta elicits marked phosphorylation of the receptor and significantly inhibits its ability to complex with the VDRE. We therefore speculate that posttranslational modification of hVDR at serine 51 may constitute a negative regulatory loop which could be operative when target cells are subject to PKC activation events.


Subject(s)
Protein Kinase C/metabolism , Receptors, Steroid/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcitriol/metabolism , Catalysis , Cell Line , DNA/metabolism , Down-Regulation , Enhancer Elements, Genetic , Escherichia coli , Haplorhini , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Receptors, Calcitriol , Receptors, Steroid/genetics , Sequence Homology, Amino Acid , Transcriptional Activation
15.
J Biol Chem ; 266(28): 18808-13, 1991 Oct 05.
Article in English | MEDLINE | ID: mdl-1655763

ABSTRACT

A baculovirus expression vector system (BEVS) was used to overproduce the full-length human vitamin D receptor (hVDR) in Spodoptera frugiperda ovarian cells. hVDR was expressed to a level of 0.5% of the total soluble protein in this system. Western analysis demonstrated that the baculovirus-generated protein had electrophoretic and immunologic properties equivalent to those of hVDR expressed in mammalian cells. The BEVS-derived receptor displayed specificity and high affinity (apparent Kd = 0.7 nM) for the 1,25(OH)2D3 ligand. Recombinant hVDR generated a specific protein-DNA complex with a duplex oligomer containing a vitamin D-responsive element (VDRE) in gel mobility shift assays. The intensity of the VDR.VDRE complex was not affected by 1,25(OH)2D3. However, the complex exhibited increased mobility in the presence of hormone, possibly the result of a 1,25(OH)2D3-dependent conformational change. A nuclear extract obtained from CV-1 cells markedly enhanced the intensity of this VDR.VDRE complex and produced an additional distinct VDR-dependent complex, thus implicating a role for nuclear auxiliary factors in multiple high affinity VDR.VDRE interactions. Finally, methylation interference studies defined the guanine residues contacted when the putative VDR-auxiliary factor complex associates with the rat osteocalcin VDRE; specifically, all of the GC base pairs in the sequence GGGTGAATGAGGACA. Therefore, these results show that the BEV system elicits high level expression of hVDR with critical functional characteristics being preserved.


Subject(s)
Baculoviridae/genetics , Receptors, Steroid/genetics , Vitamin D/metabolism , Animals , Base Sequence , Blotting, Western , Cloning, Molecular , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Vectors , Humans , Methylation , Molecular Sequence Data , Osteocalcin/genetics , Osteocalcin/metabolism , Rats , Receptors, Calcitriol , Receptors, Steroid/metabolism
16.
Proc Natl Acad Sci U S A ; 88(20): 9315-9, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-1656468

ABSTRACT

The vitamin D receptor (VDR) is known to be a phosphoprotein and inspection of the deduced amino acid sequence of human VDR (hVDR) reveals the conservation of three potential sites of phosphorylation by protein kinase C (PKC)--namely, Ser-51, Ser-119, and Ser-125. Immunoprecipitated extracts derived from a rat osteoblast-like osteosarcoma cell line that contains the VDR in high copy number were incubated with the alpha, beta, and gamma isozymes of PKC, and VDR proved to be an effective substrate for PKC-beta, in vitro. When hVDR cDNAs containing single, double, and triple mutations of Ser-51, Ser-119, and Ser-125 were expressed in CV-1 monkey kidney cells, immunoprecipitated and phosphorylated by PKC-beta, in vitro, the mutation of Ser-51 selectively abolished phosphorylation. Furthermore, when transfected CV-1 cells were treated with phorbol 12-myristate 13-acetate, a PKC activator, phosphorylation of wild-type hVDR was enhanced, whereas that of the Ser-51 mutant hVDR was unaffected. Therefore, Ser-51 is the site of hVDR phosphorylation by PKC, both in vitro and in vivo. To evaluate the functional role of Ser-51 and its potential phosphorylation, hVDR-mediated transcription was tested using cotransfection with expression plasmids and a reporter gene that contained a vitamin D response element. Mutation of Ser-51 markedly inhibited transcriptional activation by the vitamin D hormone, suggesting that phosphorylation of Ser-51 by PKC could play a significant role in vitamin D-dependent transcriptional activation. Therefore, the present results link the PKC signal transduction pathway of growth regulation and tumor promotion to the phosphorylation and function of VDR.


Subject(s)
Mutagenesis, Site-Directed , Protein Kinase C/metabolism , Receptors, Steroid/genetics , Serine , Transcriptional Activation , Amino Acid Sequence , Animals , Brain/enzymology , Calcitriol/metabolism , Cell Line , Cloning, Molecular , Escherichia coli/genetics , Genetic Vectors , Humans , Mice , Phosphorylation , Protein Kinase C/isolation & purification , Receptors, Calcitriol , Receptors, Steroid/metabolism , Recombinant Proteins/metabolism , Transcription, Genetic , Transfection
17.
Mol Endocrinol ; 5(3): 373-85, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1653893

ABSTRACT

The gene for rat bone gla protein (BGP) was isolated and 1250 basepairs (bp), including 1100 bp of 5' flanking DNA, were placed up-stream of the human GH reporter gene. After transient transfection into the osteoblast-like rat osteosarcoma cell line ROS 17/2.8, the BGP promoter demonstrated a low level of basal activity that was increased approximately 10-fold by the addition of 10(-8) M 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3]. A single 250-bp fragment (-523 to -274) was sufficient to confer hormone inducibility upon both heterologous and homologous promoters. Deletion studies, complemented by evaluation with synthetic oligomers, enabled localization of the 1,25-(OH)2D3 response element to within 19 bp (-456 to -438), containing an element with an imperfect direct repeat [GGTGA(N4)GGACA] and homology to other steroid-responsive elements. Gel retardation assays demonstrated that partially purified chick intestinal 1,25-(OH)2D3 receptor bound specifically and with high affinity to a DNA fragment containing the putative 1,25-(OH)2D3 response element, and this binding was perturbed by monoclonal antibodies to the 1,25-(OH)2D3 receptor. Surprisingly, the 250-bp fragment, when linked in an antisense orientation with respect to the BGP promoter, blocked basal and hormone-dependent gene expression. However, a 246-bp fragment 5' to the 250-bp element (-1100 to -855) restored 20-fold inducibility when linked to the first fragment in the same orientation, suggesting cooperativity between at least two elements to achieve the hormonal regulation observed in this gene.


Subject(s)
Bone and Bones/metabolism , Calcitriol/pharmacology , Calcium-Binding Proteins/genetics , Enhancer Elements, Genetic , Extracellular Matrix Proteins , Gene Expression Regulation , Repetitive Sequences, Nucleic Acid , Animals , Base Sequence , Bone and Bones/drug effects , Chickens , DNA/chemistry , Humans , Intestines/chemistry , Models, Molecular , Molecular Sequence Data , Rats , Receptors, Calcitriol , Receptors, Steroid/genetics , Restriction Mapping , Matrix Gla Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...