Corepressor Binding to Progesterone and Glucocorticoid Receptors Involves the Activation Function-1 Domain and Is Inhibited by Molybdate

Dongqing Wang and S. Stoney Simons, Jr.

Steroid Hormones Section, National Institute of Diabetes and Digestive and Kidney Diseases/Laboratory of Molecular and Cellular Biology, National Institutes of Health, Bethesda, Maryland 20892

Address all correspondence and requests for reprints to: Dr. S. Stoney Simons, Jr., Building 8, Room B2A-07, National Institute of Diabetes and Digestive and Kidney Diseases/Laboratory of Molecular and Cellular Biology, National Institutes of Health, Bethesda, Maryland 20892. E-mail: steroids{at}helix.nih.gov.


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 
Corepressors are known to interact via their receptor interaction domains (RIDs) with the ligand binding domain in the carboxyl terminal half of steroid/nuclear receptors. We now report that a portion of the activation function-1 domain of glucocorticoid receptors (GRs) and progesterone receptors (PRs), which is the major transactivation sequence, is necessary but not sufficient for corepressor [nuclear receptor corepressor (NCoR) and silencing mediator of retinoid and thyroid hormone receptor (SMRT)] RID binding to GRs and PRs in both mammalian two-hybrid and coimmunoprecipitation assays. Importantly, these two receptor sequences are functionally interchangeable in the context of GR for transactivation, corepressor binding, and corepressor modulatory activity assays. This suggests that corepressors may act in part by physically blocking portions of receptor activation function-1 domains. However, differences exist in corepressor binding to GRs and PRs. The C-terminal domain of PRs has a higher affinity for corepressor than that of GRs. The ability of some segments of the coactivator TIF2 to competitively inhibit corepressor binding to receptors is different for GRs and PRs. With each receptor, the cell-free binding of corepressors to ligand-free receptor is prevented by sodium molybdate, which is a well-known inhibitor of receptor activation to the DNA-binding state. This suggests that receptor activation precedes binding to corepressors. Collectively, these results indicate that corepressor binding to GRs and PRs involve both N- and C-terminal sequences of activated receptors but differ in ways that may contribute to the unique biological responses of each receptor in intact cells.


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 
STEROID HORMONE regulation of gene transcription involves a burgeoning number of proteins acting in concert with the appropriate receptor protein. Among the best studied are the p160 coactivators [steroid receptor coactivator-1, TIF2/glucocorticoid receptor (GR)-interacting protein 1, and amplified in breast cancer 1/pCIP/ACTR/RAC3] and the corepressors [nuclear receptor corepressor (NCoR) and silencing mediator of retinoid and thyroid receptor (SMRT)] (1, 2, 3, 4). However, interest in the corepressors has recently increased with the demonstration that the interacting factors are not limited to ligand-free and antagonist-bound nuclear receptors (5, 6) but also include antagonist-bound androgen receptor (AR) (7, 8), estrogen receptor (ER) (9, 10), GR (11, 12), mineralocorticoid receptor (13), and progesterone receptor (PR) (14, 15) receptors (reviewed in Ref. 16). Thus, corepressors may interact with the antagonist complexes of all steroid/nuclear receptors.

Yet additional targets of corepressors were suggested by the recent observations that corepressors decrease the total amounts of gene expression by receptor-agonist complexes (7, 15, 16, 17, 18, 19, 20). Also, the dose-response curve of steroid receptor-agonist complexes is influenced by corepressors (12, 13, 16, 17, 18, 19). This ability to change the position of the dose-response curve of a given gene in different cells means that the same concentration of circulating hormone can afford unequal levels of gene expression, which is invaluable for allowing differential control of gene expression during development, differentiation, homeostasis, and endocrine therapy (21, 22, 23).

These multiple actions of corepressors raise several mechanistic questions. p160 Coactivators are thought to augment the amount of receptor-mediated gene activation by binding to a highly conserved hydrophobic cleft in the receptor ligand binding domain (LBD), which is often called the activation function (AF)-2 domain and formed by four of the 12 common {alpha}-helices (numbered 3, 4, 5, and 12) (24). A strong corepressor binding site exists in the LBD of nuclear receptors, which overlaps with but is not identical with the site used by p160 coactivators (25, 26, 27). It has been assumed that a similar binding mode exists for steroid receptors. Importantly, the actions of coactivators and corepressors are reversible both at the level of biological activities (i.e. dose-response curve, partial agonist activity, and fold increase of gene activation) (Ref. 17 and Szapary, D., and S. Simons, unpublished results) and for protein-protein interactions in whole-cell and cell-free environments (12). However, different mechanisms may dictate the changes in fold transactivation with agonists vs. modulation of the partial agonist activity of antisteroids and the dose-response curve of agonists. This is because the ability of coactivators to modify the total amount of transactivation is separable from their capacity to modulate the dose-response curve and partial agonist activity (12, 13, 17, 28, 29, 30, 31, 32). These data suggest that corepressors (and coactivators) interact with receptors at another site in addition to the one in the LBD. This hypothesis is supported by the report that the inhibitory region of PR, which is the first 140 amino acids of PR-A (=165–304 of PR-B), is important for PR binding to the receptor interaction domain (RID) of corepressors (33), which is essential for corepressor binding to steroid/nuclear receptors (12, 34). Similarly, N-terminal sequences of ARs (7, 20) and GRs (11) are reported to interact with corepressors. More recently, we found that neither the N- nor the C-terminal sequences of GR are sufficient for the whole-cell or cell-free binding of corepressors (12).

Another important mechanistic question is whether the above putative additional site(s) for corepressor binding to steroid receptors is as conserved among different receptors as the currently identified LBD site. The fact that the dose-response curves and partial agonist activities for PR and GR induction of the same gene in the same cells are unequally affected by overexpression of the same corepressor (19) suggests that different features of each receptor are important. This conclusion was reinforced when PR/GR chimeras having heterologous N- and C-terminal sequences were found to afford responses that were intermediate between those of the wild-type GR and PR (19).

Finally, the biological responses of steroid receptors to corepressors usually require ligand binding, whereas the physical interactions of corepressors with steroid receptors appear to be ligand independent (7, 10, 11, 12, 15, 20, 35). Given these unequal requirements for bound steroid, the relevance of the cell-free physical interactions of corepressors with steroid receptors to the biological actions of corepressors remains an open question.

The purpose of this study, therefore, was first to see whether a second binding site for corepressors does exist in GRs and PRs. Second, we wanted to determine why ligand is required for whole-cell but not cell-free binding of corepressors to GRs and PRs. Finally, we reasoned that a comparison of the features required for corepressor binding ± steroid to GRs vs. PRs might be helpful in understanding the different biological responses of GRs and PRs, even with the same gene. We now report that a second binding site for corepressors is present in the nonhomologous amino-terminal half of GRs and PRs. For both receptors, most of the ligand-free binding of receptors to corepressors is blocked by sodium molybdate, a known inhibitor of receptor-steroid complex activation (36, 37). However, several specific differences offer mechanistic clues about how unequal responses are evoked by GRs and PRs under otherwise identical conditions.


    RESULTS
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 
Interaction of PRs with Corepressors
The receptor interaction domains (RIDs) of the corepressors NCoR and SMRT were defined by their ability to interact with the nuclear receptors (5, 6). We previously used chimeras of these RIDs fused to the GAL4 DNA binding domain (DBD) in mammalian cell assays to detect an association of corepressors with either wild-type GRs or chimeras of GR with the VP16 activation domain (AD), as determined by their ability to induce a GAL4-regulated luciferase reporter plasmid in the presence of agonist and antagonist steroids (12). The same two-hybrid assays are used here to assess the interactions of corepressors with PRs vs. GRs. The data of Fig. 1AGo show a robust interaction between NCoR or SMRT and either PR-A or PR-B complexes of the antiprogestin RU486. A much weaker, but statistically very significant, interaction is seen with agonist (R5020)-bound PRs. This is similar to what was observed for agonist- and antagonist-bound GR interactions with GAL/NCoR-RID (12). Western blots indicate that equal amounts of VP16/PR-A and -B, and GAL/NCoR-RID and GAL/SMRT-RID, are expressed in Cos-7 cells (data not shown).



View larger version (34K):
[in this window]
[in a new window]
 
Fig. 1. Interactions of Corepressors with PRs

A, Steroid-dependent mammalian two-hybrid interactions of GAL/corepressor with VP16/PR chimeras. Triplicate wells of CV-1 cells were transiently transfected with 1.0 ng of GAL/NCoR-RID (or 0.5 ng of GAL/SMRT-RID), 2.5 ng of the indicated VP16/PR chimeras, 5 ng of FRLUC, and 5 ng Renilla TS. After incubating the cells with the specified steroids for 22 h, the cells were harvested, analyzed, and normalized for the expression of the internal Renilla control as described in Materials and Methods. To combine the data of multiple experiments with different absolute amounts of luciferase (per unit of Renilla) activity, the average activity of each set of triplicate wells was then normalized to the level of activity for EtOH with VP16/PR-B. The average normalized values (±SEM) of seven to 11 independent experiments for GAL/NCoR-RID (three experiments for GAL/SMRT-RID) were plotted. P values for VP16/PRs vs. VP16: **, ≤0.0052; ***, ≤0.0009. B, Effect of mutation of CoRNR boxes of NCoR-RID. Triplicate wells of CV-1 cells were transiently transfected as in A except that 1 ng of the mutant GAL/NCoR-RIDm12 was also used. The cells were incubated with steroid, harvested, analyzed, and the average normalized values (activity of GAL/NCoR-RID and VP16/PR-B with no steroid = 1; ± SEM) of two to three independent experiments were plotted as described for panel A. P values: *, ≤0.029; **, 0.0055. C, Competition of GAL/NCoR-RID interactions with VP16/PR and VP16/GR chimeras by exogenous corepressors in mammalian two-hybrid assays. Triplicate wells of CV-1 cells were transiently transfected as in A with 1 ng GAL/NCoR-RID and 2.5 ng VP16/PR-B, or 0.2 ng GAL/NCoR-RID and 10 ng of VP16/GR, plus varying amounts of NCoR or SMRT plasmids (50 or 100 ng) and enough human serum albumin/pCMX vector to maintain a constant molar amount of pCMX plasmid. The cells were incubated with steroid, harvested, analyzed, and the average normalized values (activity of GAL/NCoR-RID and VP16/PR-B or VP16/GR with no steroid or competitor = 1; ± SEM) of five independent experiments were plotted as described for panel A. P values: *, ≤0.041; **, ≤0.0095.

 
The corepressor RIDs are required for association of GR complexes with NCoR (12). NCoR-RIDm12 contains point mutations in each of the CoRNR box motifs of the two RIDs and no longer interacts with either nuclear receptors (34) or GRs (12). The lower responses of GAL/NCoR-RIDm12 vs. GAL/NCoR-RID with either VP16/PR-B or -A (Fig. 1BGo) argue that the CoRNR box motifs are also essential for the association of both antagonist- and agonist-bound complexes of PR-A and PR-B with NCoR.

We previously documented differing behavior of PRs and GRs to corepressors (19). The much lower luciferase activity for NCoR-RID interacting with PR-B (Fig. 1AGo) than GR (12) suggests that NCoR-RID binding to PRs may be less avid than to GRs. This possibility was assessed by examining the relative ability of increasing amounts of NCoR or SMRT to reduce the interaction of GAL/NCoR-RID with PR-B and GR complexes bound by the antisteroid RU486 (Fig. 1CGo). The approximately equal levels of competition for NCoR-RID interactions with PR-B and GR by NCoR and SMRT suggest that no major differences exist in the affinity of PR- and GR-antagonist complexes for these corepressors.

N Terminus of PR Is Required for Corepressor Binding in Two-Hybrid Assays
Most studies have identified the LBD in the C-terminal half of steroid/nuclear receptors as being the corepressor binding site (14, 25, 26, 27). More recent studies indicate that GR binding to corepressors involves both N- and C-terminal domains (11, 12) and suggest that a combination of these two domains is required for the expression of biological activity of corepressors with both GR and PR (19). We therefore conducted a detailed examination of the PR and GR N-terminal sequences. Note that the numbering of the full-length PR-B (1–933) is used in all truncated PRs.

The chimera lacking the first 467 amino acids, VP16/PR468C (Fig. 2AGoGo), affords close to wild-type activity in the mammalian cell two-hybrid assay (Fig. 2BGoGo). However, deletion of the next 41 residues (468–508 to give VP16/PR509C) dramatically reduces the interaction with NCoR (Fig. 2BGoGo). Interestingly, this 41-amino-acid region is unable to mediate a productive response with GAL/NCoR-RID when present in either the larger sequence of PR395–634 (Fig. 2BGoGo) or the entire N-terminal domain of PR-B (1–535) or PR-A (165–535) (data not shown). In all cases, Western blots show that approximately equal amounts of VP16/PR chimera protein are being expressed (data not shown). The possibility that different amounts of the various expressed chimeric proteins are functionally active was eliminated by showing that the assays with 10-fold higher amounts of chimeric receptor plasmid afforded the same pattern as seen in Fig. 2BGoGo (data not shown). As a further check for functional expressed proteins, all chimeras containing an intact LBD were found to cause steroid-inducible transactivation of a progesterone-regulated luciferase reporter (GREtkLUC) (data not shown). Therefore, we propose that PR-B residues 468–508 are necessary, but not sufficient, for PR association with NCoR under our conditions.



View larger version (27K):
[in this window]
[in a new window]
 
Fig. 2. Localization of an N-Terminal Binding Site in PRs for Corepressors

A, Cartoon of PR segments used in VP16/PR chimeras for this study. PR domains (boxes with various fillings) are identified above the structure of full-length PR-B. The numbering above the wild-type (wt) PR-B sequence indicates the amino acid position of the various domain boundaries for the human PR-B sequence (43 ). B, Ability of GAL/NCoR-RID to interact in mammalian two-hybrid assays with VP16/PR-B containing various receptor deletions. Triplicate wells of CV-1 cells were transiently transfected with 1 ng GAL/NCoR-RID plus the indicated wt or truncated PR chimeras and incubated with steroid, harvested, and analyzed. The average normalized values (activity of GAL/NCoR-RID and VP16/wtPR-B with no steroid = 1; ± SEM) of four to 18 independent experiments were plotted as described for Fig. 1AGo. P values: ***, <0.0001 relative to VP16 controls; {dagger}, ≤0.009; {dagger}{dagger}, 0.0007; {dagger}{dagger}{dagger}, ≤0.0003 relative to the similarly treated wtPR-B sample. C, Ability of GAL/SMRT-RID to interact in mammalian-

 


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 2A. Continued

two-hybrid assays with VP16/PR-B containing various receptor deletions. Triplicate wells of CV-1 cells were transiently transfected with 0.5 ng GAL/SMRT-RID plus the indicated wt or truncated PR chimeras and analyzed. The average normalized values (activity of GAL/SMRT-RID and VP16/wtPR-B with no steroid = 1; ± SEM) of two to seven independent experiments were plotted as described for Fig. 1AGo. P values: ***, ≤0.0002 relative to VP16 controls; {dagger}, ≤0.041, {dagger}{dagger}, ≤0.0061 relative to the similarly treated wtPR-B sample. D, Activity of PR-B with internal deletions in mammalian two-hybrid assays. Triplicate wells of CV-1 cells were transiently transfected with 1 ng GAL/NCoR-RID plus the indicated wt or truncated PR chimeras and incubated with steroid, harvested, and analyzed. The average normalized values (activity of GAL/NCoR-RID and VP16/wtPR-B with no steroid = 1; ± SEM) of two to five independent experiments were plotted as described for Fig. 1AGo. P values: *, ≤0.011 relative to VP16 controls; {dagger}, ≤0.029 relative to the similarly treated wtPR-B sample.

 
A similar analysis indicates that amino acids 468–508 also play a large role in the interactions of PR with SMRT (Fig. 2CGoGo). Again, no major differences are obtained when 10-fold more PR plasmid is used, indicating that sufficient functionally active chimeric protein is present (data not shown). Interestingly, sequences upstream of residue 468 appear to play a larger role in PR binding to SMRT than to NCoR (c.f. Fig. 2GoGo, C vs. B).

To confirm the importance of PR residues 468–508, we examined two internal deletion mutants lacking amino acids 468–508 or 479–508. As seen in Fig. 2DGoGo, these two deletions eliminate most of the interaction of PR with both NCoR and SMRT. These results support the conclusions from Fig. 2GoGo, B and C, that the 41 amino acids from 468–508 of PR-B are necessary for PR binding to corepressors.

Identification of the GR N Terminus Region Required for Corepressor Binding in Two-Hybrid Assays
The above approach with PR was next employed to determine the location of the putative corepressor binding domain in the GR N terminus (11, 12). The data of Fig. 3AGo imply that residues 206–236 are critical for GR binding to NCoR. However, GR constructs containing an internal deletion argue for a slightly larger domain (i.e. amino acids 154–236) (Fig. 3BGo). The equal levels of expressed protein from VP16/GR, /GR{Delta}206–236, and /GR{Delta}154–236 (data not shown) indicate that the low activities of GR deletion constructs in Fig. 3BGo are not due to inadequate levels of receptor protein.



View larger version (29K):
[in this window]
[in a new window]
 
Fig. 3. Localization of an N-Terminal Binding Site in GRs for Corepressors

A, Ability of GAL/NCoR-RID to interact in mammalian two-hybrid assays with VP16/GR containing various receptor deletions. Cartoons (left) represent the GR segments used in the VP16/GR chimeras with the domains for selected activities (boxes with various fillings) being identified above the structure of full-length GR. The numbering above the wild-type (wt) GR sequence indicates the amino acid position of the various domain boundaries for the rat GR sequence (42 ). Triplicate wells of CV-1 cells were transiently transfected with 0.2 ng GAL/NCoR-RID plus 100 ng of the indicated wt or truncated GR chimeras and incubated with steroid, harvested, and analyzed. The average normalized values (activity of GAL/NCoR-RID and VP16/wtGR with no steroid = 1; ±SEM) of three to 12 independent experiments were plotted as described for Fig. 1AGo. P values: ***, <0.0001 relative to VP16 controls; {dagger}, ≤0.02, {dagger}{dagger}, 0.004, {dagger}{dagger}{dagger}, ≤0.0004 relative to the similarly treated wt GR sample. (B) Activity of GR with internal deletions or substitutions in mammalian two-hybrid assays. Cartoons (left) represent the GR segments used in the VP16/GR chimeras as in A. The solid black box in GR/PR/GR represents amino acids 458–514 of hPR-B. Triplicate wells of CV-1 cells were transiently transfected with 0.2 ng GAL/NCoR-RID plus 100 ng of the indicated wt or truncated GR chimeras and incubated with steroid, harvested, and analyzed. The average normalized values (activity of GAL/NCoR-RID and VP16/wt GR with no steroid = 1; ±SEM) of two to eight independent experiments were plotted as described for Fig. 1AGo. P values: *, ≤0.030; **, 0.0036 (alternate Welch t test) relative to the similarly treated wt GR sample.

 
We then asked whether the NCoR-RID binding domain of PR-B amino acids 468–508, implicated in Fig. 2DGoGo, could substitute for the comparable region of GR, i.e. amino acids 154–236. To do this, we created a hybrid chimera of VP16 and GR with PR-B residues 458–514 replacing GR residues 154–236. The fact that this hybrid receptor affords almost as much transactivation activity (and expressed protein; data not shown) as the wild-type VP16/GR (Fig. 3BGo) strongly argues that PR-B amino acids 458–514 comprise an NCoR-RID binding domain and that this domain can replace the comparable domain of GR.

GR and PR N-Terminal Regions Display Functional Redundancy for Gene Induction
The properties of GR, GR{Delta}154–236, and GR/PR/GR in conventional gene induction assays were next examined to determine whether the ability of PR sequences to substitute for GR residues is limited to corepressor binding or includes any of the more elaborate processes involved in transactivation. As shown in Table 1AGo, deletion of amino acids 154–236 to give GR{Delta} has multiple consequences: the total amount of induced gene product is diminished, the fold induction by dexamethasone (Dex) is reduced, the partial agonist activity of the antiglucocorticoid DexOx (32) is decreased, and the EC50 of the dose-response curve is increased. Replacing the missing GR sequence in GR{Delta}154–236 by amino acids 458–514 of PR-B to give GR/PR/GR restores each of these properties to nearly their wild-type levels (Table 1AGo).


View this table:
[in this window]
[in a new window]
 
Table 1. Activity of GR Constructs in Whole-Cell Bioassays

 
The ability of NCoR to modulate the EC50 of the dose-response curve for induction by GR-agonist complexes, and the partial agonist activity of antagonist complexes, varies with cell type. The lack of modulatory activity of transfected NCoR in CV-1 (17), but not 1470.2 (19), cells appears to be due to the already high levels of endogenous NCoR in CV-1 cells (12). Therefore, the capacity of exogenous NCoR to alter the induction properties of GR, GR{Delta}, and GR/PR/GR was determined in 1470.2 cells. This time the deletion in GR, and the replacement by the PR sequence, has little effect on either the total amount of induced gene product or the fold induction by Dex, whereas the changes in partial agonist activity and EC50 are similar to those in CV-1 cells (Table 1BGo). However, the ability of NCoR to reduce the partial agonist activity of DexOx and to increase the EC50, due to a right shift in the dose-response curve, is lost upon deletion of amino acids 154–236 of GR and is reversed, but not completely restored, upon adding PR residues 458–514 (Table 1CGo). Therefore, amino acids 458–514 of PR-B and 154–236 of GR are functionally interchangeable for all of the properties of GRs that we have examined to date.

Competition of NCoR Binding to PRs and GRs by Coactivators
Coactivators and corepressors competitively inhibit the binding of each other to both agonist- and antagonist-bound GRs (12). Because coactivators and corepressors interact with PR-agonist and antagonist complexes (Figs. 1Go and 2GoGo and Refs. 18 and 19), we asked whether coactivators can also compete with corepressors for binding to PRs. We find that transcription intermediary factor (TIF) 2.0, an N-terminal fragment of the coactivator TIF2 (Fig. 4AGo), competes for GAL/NCoR-RID interactions both with PR-B and, as previously reported (12), with GR (Fig. 4BGo). Surprisingly, constructs containing the sequence of TIF2.0, such as TIF2 and a TIF2 mutant (TIF2m123) in which the RIDs have been altered to prevent binding to steroid receptors, still compete for corepressor binding to GRs (Fig. 4AGo and Ref. 12) but not PRs. Similarly, the interaction of RU486-bound PR-B with SMRT-RID is reduced by TIF2.0 but not by TIF2 or TIF2m123 (data not shown). Thus, sequences outside of the N terminus of TIF2 affect its ability to compete for corepressor binding to PR but not GR.



View larger version (22K):
[in this window]
[in a new window]
 
Fig. 4. Competition by Coactivators of NCoR-RID Interactions with PRs and GRs in Mammalian Two-Hybrid Assays

A, TIF2 constructs used in competition assays. Cartoons show the various domains present in TIF2 and the truncated species: RIDs (solid bars), mutated RIDs (half bars), AD1/CBP binding domain (stippled), Q-rich (horizontal striping), and AD2 (cross hatched). B, Coactivator TIF2 inhibition of GAL/NCoR-RID association with VP16/wtPR-B (left) and VP16/wtGR (right). Triplicate wells of CV-1 cells were transiently transfected as in Fig. 1CGo ± 213 ng of TIF2 plasmid, or the molar equivalent of the other TIF2 chimeras, and incubated with steroid, harvested, and analyzed. The average normalized values (activity of GAL/NCoR-RID and VP16/wtPR-B or /wtGR with no steroid = 1; ± SEM) of five (PR-B) and three (GR) independent experiments were plotted as described for Fig. 1AGo. P values relative to the corresponding untreated wt sample: for PR, * = 0.012 (paired t test), for GR, * ≤0.042.

 
NCoR Binding to PRs in Cell-Free Pull-Down Assays
We next inquired whether a direct association of corepressor with PR can be seen in cell-free pull-down assays. Figure 5AGo shows that the immobilization of [35S]methionine-labeled PR-B by glutathione-S-transferase (GST)/NCoR-RID is much greater than that by GST and thus depends upon the presence of NCoR-RID. This binding is relatively independent of ligand. Surprisingly, whereas the biological activities of full-length PR-B and PR468C are much greater than that of PR562C in mammalian cell two-hybrid assays (see Fig. 2BGoGo), densitometric scanning of paired experiments (Fig. 5Go, A and B) shows that 3–8% of the input PR, or PR562C, ± steroid was immobilized by NCoR-RID (data not shown). We therefore examined the binding to GST/NCoR-RID of N- and C-terminal fragments of both PR and GR. As expected (12), neither half of GR binds to NCoR-RID (Fig. 5CGo). In contrast, a PR-B C-terminal fragment (PR562C) binds to NCoR-RID, whereas the N-terminal half (PRN561) does not. Thus, there are significant differences in the ability of the C-terminal domains of GR and PR to bind to NCoR-RID in cell-free pull-down assays.



View larger version (42K):
[in this window]
[in a new window]
 
Fig. 5. NCoR-RID Binding to PRs and GRs in Cell-Free Pull-Down Assays

A, Effect of ligand on PR-B binding to GST/NCoR-RID. [35S]methionine labeled, in vitro-translated PR-B ± the indicated steroids was incubated with glutathione-Sepharose 4B beads containing bound GST ± NCoR-RID. The tightly bound material was washed, eluted, separated on SDS-PAGE gels, and visualized by radioautography as described in Materials and Methods. The left-hand most lane (10%) represents the amount of [35S]methionine-labeled protein in 10% of the material loaded onto each column. Similar results were obtained two additional independent experiments. B, Ability of N-terminal truncated PRs to bind to GST/NCoR-RID. The binding of [35S]methionine-labeled wt PR-B, PR486C, and PR562C (see Fig. 2AGoGo) to immobilized GST ± NCoR-RID was determined as in A. Arrow indicates the position of the labeled PR species. Similar results were obtained in a second independent experiment. C, Comparison of the NCoR binding capacity of PR and GR N- and C-terminal fragments. The binding of [35S]methionine-labeled PR and GR fragments to immobilized GST ± NCoR-RID was determined as in A. Similar results were obtained in a second independent experiment.

 
NCoR Binding to PRs and GRs in Whole-Cell Coimmunoprecipitation Assays
To determine whether the binding of PRs to NCoR seen in the cell-free pull-down assays also occurs in intact cells, we asked whether overexpressed PR-B and GAL/NCoR-RID affords an immunoprecipitable complex. Using anti-GAL antibody for the immunoprecipitation, followed by anti-PR antibody to detect coprecipitated PR-B, Fig. 6AGo shows that PR-B is associated with GAL/NCoR-RID but only when NCoR-RID is present (lanes 8 vs. 7). As in the pull-down assays (Fig. 5AGo), the formation of a PR-B/NCoR-RID complex is ligand independent (lanes 8–10).



View larger version (54K):
[in this window]
[in a new window]
 
Fig. 6. Immunoprecipitation of Whole-Cell Complexes of wt PR-B with Corepressor NCoR

A, Effect of PR ligand on coimmunoprecipitation of PR-B complexes with GAL/NCoR-RID. Cos-7 cells were transiently transfected with 5 µg of PR-B plus equimolar amounts of either GAL (1.6 µg) or GAL/NCoR-RID (2 µg) plasmids and incubated with the indicated steroids (E, EtOH; R, R5020; RU, RU486) for 2 h as described in Materials and Methods. Anti-GAL antibody was used to immunoprecipitate GAL or GAL/NCoR-RID and associated proteins. The amount of PR present in 2% of the inputs (lanes 1–5) and in the immunoprecipitates (lanes 6–10) was detected by Western blotting with anti-PR antibody. Similar results were obtained in a second independent experiment. B, Importance of various PR domains for coimmunoprecipitation of PR-B complexes with GAL/NCoR-RID. Samples were prepared in the absence of steroid and NCoR-bound VP16/PR fragments were detected by Western blotting with anti-VP16 antibody. Similar results were obtained in a second independent experiment. Nonspecifically detected species are marked by {dagger} and *, the latter of which migrate just above PR468C. C, GR domain required for coimmunoprecipitation with GAL/NCoR-RID can be replaced by comparable PR domain. Samples were prepared in the absence of steroid and NCoR-bound VP16/GR fragments were detected as shown in panel B with anti-VP16 antibody. *, Location of a nonspecifically detected species.

 
We then asked whether the selective binding capacity of NCoR to various PR domains seen in the pull-down assays (Fig. 5Go) could be recapitulated under whole-cell conditions. Here, VP16 chimeras of the PR fragments were used to facilitate immunochemical identification. VP16/PR-B is coimmunoprecipitated with GAL/NCoR-RID in the presence of the antiprogestin RU486 in a manner that requires NCoR-RID (compare lanes 7 vs. 6 of Fig. 6BGo), just like the wild-type PR-B (Fig. 6AGo). Thus, the VP16 moiety does not alter the ability of PR-B to associate with GAL/NCoR-RID in whole cells. Of the various PR fragments, the N-terminal species PR-BN is not coimmunoprecipitated (lane 10), whereas the C-terminal fragments PR468C and PR509C are, all in the presence of RU486. However, laser densitometry shows that the amount of receptor coimmunoprecipitated with NCoR-RID is about 6-fold less for PR509C than for full-length PR-B and PR468C (data not shown), suggesting that the efficiency of PR509C binding to NCoR is reduced and that more N-terminal sequences of PR are needed for full binding activity.

Finally, we asked whether the PR sequence 458–514 could restore corepressor binding in a coimmunoprecipitation assay to GRs that are deleted of their corepressor binding site. As expected from the data of Fig. 3BGo, the deletion of GR residues 154–236 dramatically reduces the binding of GR to NCoR-RID under whole-cell conditions (lanes 8 vs. 6 in Fig. 6CGo). However, replacement of the deleted GR sequence with PR amino acids 458–514 rescues the NCoR binding of GR (lanes 7 vs. 6). This reinforces our above conclusions about the importance of these regions for corepressor binding to PRs and GRs.

Activation Is Required for Receptor Binding to NCoR
The binding of PRs to NCoR in the pull-down and coimmunoprecipitation assays is steroid independent (Figs. 5AGo and 6AGo). In contrast, PR interactions with NCoR in whole-cell mammalian two-hybrid assays are steroid dependent (Figs. 1–4GoGoGoGoGo). An identical dichotomy was observed for GR interactions with NCoR-RID (12). It is well known that the DNA binding of steroid receptors requires prior activation of the receptor protein. Furthermore, it is thought that the DNA binding of activated receptors precedes the recruitment of cofactors (38). Therefore, we speculated that a ligand-independent activation of cell-free receptors might be permitting their binding to corepressors. To test this hypothesis, we examined the effect of sodium molybdate, which blocks receptor activation (36, 37), on GR and PR binding to NCoR in the presence and absence of added steroid in pull-down assays. Steroid (1 µM) was added to maximize the amount of steroid-bound receptors that are obtained during in vitro translation (39). Sodium molybdate (20 mM) was present during the in vitro translation reactions to prevent any possible activation of GRs or PRs during their synthesis. As a control for nonspecific ionic effects of molybdate (MoO4–2), we used the iso-electronic sulfate group (SO4–2). The presence of molybdate or sulfate does not alter the amount of in vitro-translated GR (Fig. 7AGo, lanes 1–6), nor do they increase the background binding of GRs to GST (Fig. 7AGo, lanes 10 and 11). However, 20 mM molybdate does selectively inhibit the cell-free binding of GRs ± agonist or antagonist ligands to NCoR-RID (Fig. 7AGo, lanes 7–9 vs. 12–14). Similarly, 20 mM molybdate, but not sulfate, dramatically reduces the cell-free binding of PRs ± agonist or antagonist to GST/NCoR-RID (Fig. 7BGo, lanes 7–9 vs. 12–14). Thus, it appears that the activation of receptors is required for binding to corepressors in pull-down assays and that the binding of ligand-free GRs and PRs to corepressors (Fig. 5AGo and Ref. 12) is due to the formation of ligand-free activated receptors.



View larger version (52K):
[in this window]
[in a new window]
 
Fig. 7. Effect of Molybdate on Cell-Free Binding of Corepressor to PRs and GRs

A, Molybdate but not sulfate inhibits GR binding to NCoR-RID in pull-down assays. Samples were prepared as in Fig. 5Go except that either 20 mM Na2MoO4 or Na2SO4 was present, with the indicated steroids, from the start of the in vitro transcription/translation. Lanes 1–6 show 10% of input. The binding of GR ± 1 µM steroids (RU = RU486) to GST/NCoR-RID (lanes 7–9 and 12–14) or GST (lanes 10–11) in the presence of molybdate vs. sulfate is displayed in lanes 7–9 and 11 vs. lanes 10 and 12–14, respectively. B, Molybdate but not sulfate inhibits PR binding to NCoR-RID in pull-down assays. Samples were processed as in panel A except that the cDNA for PR-B was used instead of GR and 1 µM R (R5020) was used instead of Dex. Similar results were obtained in two additional independent experiments. C, Molybdate but not sulfate inhibits coimmunoprecipitation of PR and GR with GAL/NCoR-RID. Samples were prepared as in Fig. 6Go with the modification that the cell lysates were adjusted to contain either 20 mM Na2MoO4 or Na2SO4 immediately after rupturing the cells. Immunoprecipitation was performed with anti-GAL antibody, whereas antireceptor antibodies were used for Western blotting. Lanes 1–4 give 5% of the input samples (mock and receptor plus GAL or GAL/NCoR-RID), whereas lanes 5–8 show the immunoprecipitated material. The intensity of each band in three to four separate experiments was determined by laser densitometry and the average ± SEM was plotted in panel D (Mo, Na2MoO4; SO4, Na2SO4; Input, 5% of input; IP, immunoprecipitate).

 
As a further test of this hypothesis, we asked whether the ligand-free association of PR-B with NCoR in the whole-cell coimmunoprecipitation experiments (Fig. 6AGo) is also due to an activation of ligand-free receptors that can be blocked by molybdate. We did not try to prevent the activation of ligand-bound PRs within the cells because the ionic nature of sodium molybdate makes it difficult to get a sufficiently high intracellular concentration of molybdate. However, when added immediately after cell lysis, molybdate, but not sulfate, reduces the coimmunoprecipitation of ligand-free PRs and GRs (lanes 7 vs. 8 of Fig. 7CGo, top and bottom panels, respectively) by a factor of 4 (Fig. 7DGo). This argues that activation is responsible for most of the coimmunoprecipitation of ligand-free receptors with NCoR.


    DISCUSSION
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 
A Second Corepressor Binding Site in PRs and GRs
The binding of the corepressors NCoR and SMRT is well known to involve a region of the LBD of steroid/nuclear receptors that overlaps with the coactivator binding site. The current study presents several lines of evidence for a second region in the N-terminal half of PRs and GRs that contributes to the binding of corepressors to full-length receptors bound by both agonists and antagonists. First, amino acids 458–514 of PR-B, and 154–236 of GR, are essential for interactions with NCoR-RID and SMRT-RID in mammalian two-hybrid assays (Figs. 2GoGo and 3Go) in a manner that requires the corepressor RID sequence (Fig. 1BGo). However, this amino-terminal binding domain is not sufficient for wild-type whole-cell interactions of corepressor with either receptor (Fig. 2CGoGo) (12). Similar results are seen under cell-free pull-down (Fig. 5Go) and whole-cell coimmunoprecipitation (Fig. 6Go) conditions, although PR constructs lacking the amino-terminal domain show more association than in the whole-cell bioassays (Fig. 2GoGo). This may be due, in part, to the much higher concentrations of receptors and corepressors that are used in the pull-down and coimmunoprecipitation assays, which would permit the formation of more weakly bound complexes. It has been estimated that protein-protein interactions with affinities lower than 50–100 nM are not detected in two-hybrid assays (40).

We could discern no obvious sequence homology between the two corepressor binding regions of PR and GR. Nevertheless, the PR domain of 458–514 restores NCoR binding to the inert GR deletion mutant (GR{Delta}154–236) under whole-cell coimmunoprecipitation and two-hybrid conditions (Figs. 3BGo and 6CGo) and largely replaces the deleted GR domain in a variety of transactivation responses (Table 1Go). This is strong evidence for the importance of these two, nonhomologous sequences in receptor binding and function of corepressors. These results further suggest that the tertiary structure of the protein, and not amino acid sequence, is critical for corepressor binding to, and the biological activities of, these amino-terminal domains (Fig. 8Go). In sum, these data nicely explain why both N- and C-terminal regions of PR and GR were found to be required for the full response to corepressors (19).



View larger version (18K):
[in this window]
[in a new window]
 
Fig. 8. Model for Corepressor Interactions with GRs vs. PRs

The cartoon of each receptor (numbers indicate amino acid residues) shows a single molecule bound to DNA via its DBD (440–505 for GR, 556–642 for PR). Repression is proposed to occur with agonist and antagonists receptors via corepressor (CoR) binding (thick dashed line; thicker line for CoR interaction with PR-LBD indicates stronger binding) to both the LBD and the amino-terminal region (154–236 of GR, 468–508 of PR), which resides within the AF-1 domain (77–262 of GR, 456–546 of PR). In addition, the N-terminal half of PR including AF-3 (61 ) and an N-terminal region of GR (261–360) (62 ) interact with the LBD (thin dashed line). Coactivators can compete with corepressors for binding to receptor LBD (thick dashed line) (12 ) to cause increased activation. It is further proposed that the dissociation of agonist steroid forces the equilibrium in the direction of CoR-bound receptors. See text for further details.

 
Our results extend the report that N- and C-terminal sequences of GRs participate in corepressor binding (11) and are similar to findings that corepressors interact with amino-terminal residues of ARs (7, 20) and PRs (41). Thus, it appears that corepressor binding to N-terminal domains of the classical steroid receptors may be a general phenomenon.

The corepressor binding sequences that we identified at amino acids 458–514 of human PR(hPR), and 154–236 of rat GR, overlap the AF-1 domains of GRs (residues 98–282) (42) and PRs (456–546) (43). Given the importance of the AF-1 domain for the amount of transactivation (30, 44, 45) and proper RNA splicing (46) in conjunction with the requirement of a corepressor binding sequence in the GR N-terminal domain for robust transactivation (Table 1Go), it is tempting to speculate that the competitive binding of corepressors and other factors to the AF-1 domain is sufficient to block many of the AF-1 activities.

Protein sumoylation has been suggested to facilitate corepressor binding (47). Rat GR is sumoylated at residues K297 and K313 (48), and human PR is sumoylated at K388 (49, 50). However, these residues are outside of the regions that contribute to corepressor binding: GR154–236 and PR458–514 (Figs. 2GoGo, B and C, and 3Go). Therefore, we conclude that the sumoylation sites of GR and PR are not required for corepressor binding.

The whole-cell binding of corepressors to both PRs and GRs appears to be reversible (Fig. 1CGo and Ref. 12). We confirm that coactivators inhibit the interactions of NCoR with GRs (Fig. 4BGo). The amino-terminal portion of TIF2 also counteracts the association of NCoR with PR under conditions where the full-length TIF2 is inactive. Whether this is due to the higher level of expression of the fragment TIF2.0 vs. the full-length TIF2 (our unpublished observations), to an unidentified PR-associated protein that impedes TIF2 binding, or to some unique property of TIF2.0 remains to be determined. Nevertheless, the data with PR support our general hypothesis that coactivators and corepressors competitively inhibit the binding of each other to receptor-agonist and -antagonist complexes (12, 16, 17, 28).

Differences between PRs and GRs
Numerous examples exist for PRs and GRs eliciting different responses even within the same cells (19, 51). The molecular reasons for this are likely to be multifactorial. One component appears to be unequal interactions of corepressors with a combined receptor target of N- and C-terminal domains (19). The present study supports this hypothesis. The quantitative response of GRs in mammalian two-hybrid assays is much greater for NCoR than SMRT (12) but about the same with PRs (Fig. 1AGo). These results, in combination with the recent report that the magnitude of gene expression in two-hybrid assays is determined by the affinity of the two interacting proteins for each other (40), suggest that the affinity of corepressors for GRs and PRs are not equal. Thus, the same cellular concentration of corepressors would be predicted to afford different amounts of corepressor-bound receptors (Fig. 8Go). Cell-specific factors are also important, as shown here by the much greater reduction, upon deleting GR residues 154–236, in the total amount of induced luciferase, and the fold induction of luciferase, in CV-1 cells than in 1470.2 cells (Table 1Go). The identification of these cell-specific factors should be very informative.

We have not been able to identify the amino acids that participate in corepressor binding by the N-terminal domain of PRs and GRs because no interactions are observed for the isolated domains in two-hybrid (Fig. 2BGoGo and Ref. 12), pull-down (Fig. 5CGo and Ref. 12), or coimmunoprecipitation (Fig. 6BGo) assays. We propose that the C-terminal sequence of each receptor plus other cellular proteins contribute to the formation of the final complex. Nevertheless, the fact that the C-terminal half of PR has a higher avidity for NCoR-RID than does the GR C terminus in both pull-down (Fig. 5CGo) and coimmunoprecipitation (Fig. 6BGo) assays is yet another difference between the two receptors (Fig. 8Go).

Coactivators compete for corepressor binding to both PRs and GRs (Fig. 4Go and Ref. 12), but the determinants of this competition are not identical. Sequences outside of TIF2.0, which is the N terminus of TIF2 (Fig. 4AGo), affect its ability to compete for corepressor binding to PR but not GR. Thus, one would expect that the same intracellular ratio of coactivators to corepressors would result in a different proportion of PRs and GRs being associated with coactivators (and corepressors), thereby resulting in unequal amounts of gene activation or repression of common target genes (Fig. 8Go). In summary, these differences in coactivator binding in combination with the unequal affinities of corepressors for binding to selected regions of PRs and GRs expand the mechanisms by which nonidentical responses can be generated by PRs and GRs.

Molybdate Inhibition of Receptor Binding to Corepressors
The present data indicate that the apparent contradiction of why the whole-cell biological responses of GRs and PRs with corepressors, but not the biochemical binding/association, require ligand-bound receptors is due, at least in part, to the activation of ligand-free receptors under cell-free conditions. Activation is defined as a still poorly understood process that converts steroid receptors from a non-DNA-binding to a DNA-binding form and is associated with the dissociation of chaperon proteins such as heat shock protein 90 (37). Steroid-free receptors can be activated (52) under several conditions (53) that do not occur in cells but can be blocked by sodium molybdate (36, 37). The observation that molybdate both prevents activation and reduces the cell-free binding/association of GRs and PRs with NCoR in pull-down and coimmunoprecipitation assays (Fig. 7Go) argues that only activated receptors can bind to corepressors. Whether receptor-bound chaperone proteins block corepressor binding is a logical, but untested, hypothesis even though heat shock protein 90 is thought to bind only to the GR LBD (54). Molybdate, but not sulfate, inhibits the formation of activated complexes in coimmunoprecipitation assays (Fig. 7CGo), suggesting that we are not witnessing a general salt effect and that most of the activation occurs after cell lysis. Molybdate does not have any effect on activated receptors (55, 56), so it is unlikely that molybdate disrupts receptor-corepressor complexes that were preformed in intact cells. Rather, we interpret the data of Fig. 7Go, C and D, as showing that the ligand-free PRs and GRs present in intact cells can be activated by various processes subsequent to cell rupture. Whether the small amount of ligand-free PR and GR that binds to corepressors in the coimmunoprecipitation assays in the presence of 20 mM molybdate (lanes 7 vs. 6 in Fig. 7CGo and lanes labeled "Mo/IP" in Fig. 7DGo) represents the existence of a low level of activated ligand-free receptors in intact cells remains to be established.

The possible presence of ligand-free, activated receptors in intact cells, coupled with the requirement of steroid for GR binding to coactivators in pull-down assays (28), permits an interesting mechanism for the deinduction of steroid-regulated gene transcription (Fig. 8Go). The data of Fig. 7Go suggest that the dissociation of steroid from activated receptors bound to hormone response elements will cause a major decrease in coactivator affinity with little change in the affinity of corepressors. Given the ability of corepressors and coactivators to competitively inhibit the binding of each other to GRs in an equilibrium fashion (12, 17), this decrease in coactivator affinity would result in a markedly increased ratio of corepressors to coactivators that are associated with the hormone response element-bound receptors and a concomitant decrease in transactivation activity. Given the similar corepressor-binding properties of ligand-free PRs and GRs, we propose that the ligand-independent binding of the other classical steroid receptors to corepressors (7, 10, 11, 12, 15, 20, 35) also depends upon the activation state of the receptor. In this case, the above-proposed model for gene deinduction may prove to be general for all of the classical steroid receptors. How many other proteins bind to steroid receptors only after activation remains to be seen.

Some studies with ERs support the hypothesis that cofactor binding to receptors can modify receptor affinity for ligands (Ref. 57 but not Ref. 58). Comparable studies have not been conducted with GRs. However, steroids cannot bind to activated GRs (59). Therefore, the fact that activation is needed for corepressor binding to GRs strongly argues that corepressor binding to GRs cannot alter the affinity of steroid binding to ligand-free GRs. Similarly, the binding of coactivator TIF2 to DNA-bound GRs occurred only with GR-steroid complexes (60). At this point, steroid binding to GRs is again not reversible, and the affinity of steroid binding to GRs cannot be affected. Instead, some step downstream of steroid binding, and GR activation (32), is a more probable target for corepressors and coactivators in their modulation of the dose-response curve of agonists and the partial agonist activity of antagonists (12, 16, 60).


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 
Unless otherwise indicated, all operations were performed at 0 C.

Chemicals
Dex was obtained from Sigma (St. Louis, MO), and promegestone (R5020) was from PerkinElmer Life Sciences (Boston, MA). RU486 was a gift from Etienne Baulieu (Paris, France). Restriction enzymes and DNA polymerase were from New England Biolabs (Beverly, MA), Amersham Biosciences (Piscataway, NJ), or Promega (Madison, WI). [35S]Methionine was from Amersham Biosciences, and sodium molybdate and sodium sulfate were from Baker Chemical (Phillipsburg, NJ).

Plasmids
The Renilla null luciferase reporter was purchased from Promega (Madison, WI), pM vector and GAL/VP16 from CLONTECH (Palo Alto, CA), and pFR-LUC reporter from Stratagene (La Jolla, CA). Human serum albumin/pS65, VP16/GR, VP16/GR361C, VP16/GR407C, pRBAL-GR, pRBAL-GRN523, pRBAL-GR486C have been described previously (12). Chimeras of the VP16 activation domain and PR-B, PR-A, PR-BN, PR-AN, and PR-LBD were gifts from Dean P. Edwards (University of Colorado Health Sciences Center, Denver, CO). Other donated plasmids were received from Hinrich Gronemeyer (Institut de Génétique et de Biologie Moléculaire et Cellulaire, Strasbourg, France: hPR-B, TIF2, TIF2.0, and TIF2m123), Michael Rosenfeld (University of California-San Diego, San Diego, CA: NCoR), Ron Evans (Salk Institute, La Jolla, CA: s-SMRT), Keith Yamamoto (University of California-San Francisco, San Francisco, CA: GR), and Mitch Lazar (University of Pennsylvania School of Medicine: GAL/NCoR-RID [amino acids 1944–2453], GAL/NCoR-RIDm12, GAL/SMRT-RID [amino acids 982–1495], and GST/NCoR-RID).

Several chimeras were synthesized as follows. For VP16/PR468C, the AscI-(filled in) and PstI digest of VP16/PR-B was inserted into EcoRI-(filled in) and PstI-cleaved VP16. For VP16/PR545C, the Bsu36I-(filled in) and SapI fragment of VP16/PR-B was inserted into HincII- and SapI-treated VP16. For VP16/PR562C, the PleI-(filled in) and PstI-restricted VP16/PR-B was inserted into the SalI (filled in) and PstI product from VP16. For VP16/PR395C, the StuI- and XbaI-generated fragment of VP16/PR-B was inserted into the MluI-(filled in) and XbaI of VP16. For VP16/PR395–634, StyI-(fill-in) and EcoRI of VP16/PR395C was inserted into HindIII-(filled in) and EcoRI-treated VP16. VP16/PR479C and VP16/PR509C were constructed using PCR amplification of VP16/PR-B with the forward primers 5'-AGAATTCCCCTGCAAGGCGCCGGGC-3' and 5'-AGAATTCCCCGCGCTCTACCCTGCACTC-3', respectively, and the same reverse primer 5'-GCTCTAGAGCTTTTTATGAAAGAGAAG-3'. The resulting PCR product was digested with EcoRI/XbaI, and inserted into VP16 using the same restriction sites. VP16/PR{Delta}468–508 was prepared using PCR amplification of VP16/PR-B with the forward primer 5'-GCCCCCGCGCTCTACCCTGCACTC-3' and the same reverse primer as for VP16/PR479C. The resulting PCR products were digested with XbaI and inserted into VP16/PR-B using AscI (filled in) and XbaI. VP16/PR{Delta}479–508 was synthesized using PCR amplification of VP16/PR-B with two separate sets of primers: one set primers was 5'-GGAATTCATGACTGAGCTGAAGGCAAAG-3' and 5'-TGGAGGTGGCGCGAACGGGCCCTG-3', the other set was the same as for VP16/PR{Delta}468–508. The resulting PCR product was digested with EcoRI and XbaI and inserted into VP16 using the same restriction sites.

To make VP16/GR{Delta}154–236 and VP16/GR{Delta}206–236 were prepared by amplifying VP16/GR with the same forward primer (5'-GGAATTCATGGACTCCAAAGAATCCTTAGC-3') coupled with the reverse primers 5'-GAAGATCTGTGGGATACAATTTCACACTGCC-3' for VP16/GR{Delta}206–236 or 5'-GAAGATCTGTCGACCTATTGAGGTTTG-3' for VP16/GR{Delta}154–236. Each resulting PCR product was digested with EcoRI and BglII and ligated with two other fragments: the 1.7-kb fragment obtained by BglII and XbaI digestion of VP16/GR and the 3.3 kb fragment from EcoRI/XbaI digestion of VP16. pSVLGR{Delta}154–236 was prepared by cutting pSVLGR with AccI and BglII and then self-ligating after filling-in the 3'-recessed termini with Klenow fragment. The VP16/GR/PR/GR chimera was prepared by ligating three fragments: 1) the PCR amplified product from VP16/PR-B with forward primer 5'-CCATCCAGACCCGGGGAAGCG-3' and reverse primer 5'-GAAGATCTGCAGGGTAGAGCGCGGG-3' that was then digested with SalI and BglII, 2) the 1.7-kb fragment obtained by BglII and XbaI digestion of VP16/GR, and 3) the 3.7-kb fragment from SalI/XbaI digestion of VP16/GR. VP16/GR/PR/GR was digested with SalI and BglII and the 170-bp fragment was inserted into pSVLGR using the same restriction sites to yield pSVLGR/PR/GR.

For pSG5/PR-B, the EcoRI and XbaI (filled in) digestion product of VP16/PR-B was inserted into EcoRI- and BamHI-(filled in) treated pSG5. PSG5/PR562C and pSG5/PR561N were both made by PCR, using VP16/PR-B as the template. For pSG5/PR562C, the primers were 5'-GGAATTCTTACCTCAGAAGATTTGTTTAATC-3' and 5'GAAGATCTCACTTTTTATGAAAGAGAAG-3'. For pSG5/PR561N, the primers were 5'-GGAATTCATGACTGAGCTGAAGGCAAAGG-3' and 5'-GAAGATCTGACTCGAAGCTGTATTGTGG-3'. In both cases, the amplified DNA was then cut with EcoRI and BglII and inserted into the same sites of pSG5.

Cell Culture and Transient Transfection
Monolayer cultures of CV-1, Cos-7, and 1470.2 cells were grown as previously described (18, 30). The total transfected DNA was adjusted to 300 ng/well of a 24-well plate (or 3 µg/60-mm dish) with pBluescriptII SK+ (Stratagene). Renilla TS (Promega) (5–10 ng/well of a 24-well plate) was included as an internal control. Cells were incubated with plasmid DNA, Opti-MEM I, and LipofectAMINE or FuGENE 6 reagent (Roche Molecular Biochemicals, Indianapolis, IN) for 24 h, after which this mixture was replaced by the normal media (10% fetal calf serum, DMEM). The cells were induced with steroids for 20–24 h and then harvested. The cells were lysed and assayed for reporter gene activity using the Dual Luciferase Assay reagents according to manufacturer’s instruction (Promega). Luciferase activity was measured by an EG&G Berthhold (Oak Ridge, TN) luminometer (Microlumat LB 96P). The data were normalized either for total protein or Renilla null luciferase activity.

Mammalian Two-Hybrid Assays
The recommended procedure for the Mammalian Matchmaker two-hybrid assay kit (CLONTECH) was modified slightly by changing from a chloramphenicol acetyltransferase reporter to the luciferase reporter pFRLuc (Stratagene), which is under the control of five repeats of the upstream activating sequence for the binding of GAL4.

Bacterial Expression of Proteins
The pGEX series of plasmids were transformed into Escherichia coli (BL21[DE3]; Stratagene) according to the manufacturer’s procedure. A single colony was picked and inoculated into 3 ml Luria Bertani broth with 100 mM ampicillin. After overnight culture, 1 ml of bacterial culture was diluted into 50 ml of Luria Bertani broth containing 100 mM ampicillin, shaken at 37 C for 2 h, adjusted to 0.5 mM isopropyl-ß-D-thiogalacto-pyranoside, and shaken at 25 C for another 3 h. The cells were harvested by centrifugation, washed once with PBS, resuspended in 10 ml of PBS, and sonicated for 30 cycles at 30% of maximum power (Fisher Scientific sonic dismembrator, model 500). The supernatant was collected for use after centrifugation (5000 x g for 20 min).

In Vitro Transcription and Translation Assays
For each reaction, 1 µg of plasmid DNA was mixed with 2 µl of [35S]-methionine and 40 µl of TNT T7 (or SP6) master mix (Promega) and brought up to a total volume of 50 µl with H2O. The reaction was conducted at 30 C for 90 min. Various steroid hormones (Dex, R5020, or RU486), with or without 20 mM sodium molybdate or sodium sulfate were added into the transcription-translation reaction during the 30 C incubation.

Pull-Down Assays
Sonicated bacterial lysates (0.5 ml) containing overexpressed GST or GST/NCoR-RID were incubated with 20 µl of glutathione-Sepharose 4B beads for 1 h at 0 C. The mixture was centrifuged (12,000 x g), the supernatant discarded, and the matrix was washed with PBS (4 x 1 ml). Each 20 µl sample of immobilized GST or GST-chimera was then incubated overnight at 4 C with 10 µl of hormone prebound, activated, [35S]labeled in vitro-translated GR. The matrix was washed with Buffer H (4 x 1 ml). The immobilized proteins were removed from the beads by heating at 90 C for 5 min in 20 µl of 2x sodium dodecyl sulfate (SDS) loading buffer. The proteins were then separated on 8 or 10% SDS-PAGE gels, and the bound receptor was located by autoradiography.

Coimmunoprecipitation Assays
The day before transfection, Cos-7 cells were seeded into 150-mm dishes at 200,000 cells per dish containing 20 ml of media. On the next day, about 30 µg of DNA/dish was transfected with 60 µl of FuGene reagent. After 2 d, the cells were treated with EtOH ± 1 µM RU486. Cells were lysed 2 h later at room temperature with CytoBuster Protein Extraction Buffer (Novagen, Madison, WI) and clarified by centrifugation at 16,000 x g for 5 min at 4 C. For complexes with GAL-DBD tagged proteins, immunoprecipitation was achieved by incubating with anti-GAL4DBD (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) while rocking (100 cycles/min) at 4 C overnight and then immobilizing the antibody complexes on Protein G Plus/Protein A-Agarose (Oncogen Research Products, San Diego, CA) with rocking (100 cycles/min) at 4 C for another 2 h. In both cases, the agarose beads were then washed three times with 1 ml of ice-cold wash buffer [50 mM Tris (pH 7.5), 150 mM NaCl, 0.1% Nonidet P-40, complete protease inhibitor cocktail (Roche)]. The proteins were extracted with 2x SDS-loading buffer (95 C for 3 min) and separated by gel electrophoresis (8–10% SDS-PAGE), and visualized by Western blotting with anti-PR ({alpha}PR-22) or anti-GR (BUGR-2) antibody (Affinity Bioreagents, Golden, CO).

Western Blotting
SDS-PAGE gels were equilibrated in transfer buffer for 15 min at room temperature before electrophoretic transfer of the proteins to nitrocellulose membranes (Schleicher & Schuell BioScience, Keene, NH) in a Bio-Rad (Hercules, CA) small (150–200 mA overnight) or large (350 mA overnight) Transblot apparatus. The nitrocellulose membranes were stained with Ponceau S (0.02% Ponceau S and 0.04% glacial acetic acid in water) to localize the molecular weight markers. The VP16 fusion proteins were probed with rabbit VP16 polyclonal antibody (CLONTECH) and the GAL4 fusion proteins were probed with mouse GAL4 DBD monoclonal antibody (CLONTECH). Antibody complexes were visualized by ECL detection reagents as described by the manufacturer (Amersham Biosciences). Western blot films were scanned using a Bio-Rad Model GS-800 Calibrated Imaging Densitometer and the densitometry quantitated using Bio-Rad Quantity One software.

Statistical Analysis
Unless otherwise noted, all experiments were performed in triplicate several times. The values of n independent experiments were then analyzed for statistical significance by the two-tailed Student’s t test using the program InStat 2.03 for Macintosh (GraphPad Software, San Diego, CA). When the difference between the SDs of two populations is significantly different, then the Mann-Whitney test or the Alternate Welch t test is used.


    ACKNOWLEDGMENTS
 
We thank Dean P. Edwards, Ron Evans, Hinrich Gronemeyer, Mitch Lazar, Keith Yamamoto, and Michael Rosenfeld for generously donating plasmids, Tomoshige Kino (National Institute of Child Health and Human Development, National Institutes of Health) for critical review of the manuscript, and members of the Steroid Hormones Section for helpful comments.


    FOOTNOTES
 
First Published Online March 17, 2005

Abbreviations: AD, Activation domain; AF, activation function; AR, androgen receptor; GAL, GAL4 DNA binding domain; DBD, DNA binding domain; Dex, dexamethasone; ER, estrogen receptor; GST, glutathione-S-transferase; GR, glucocorticoid receptor; hPR, human PR; LBD, ligand binding domain; NCoR, nuclear receptor corepressor; PR, progesterone receptor; RID, receptor interaction domain; SMRT, silencing mediator of retinoid and thyroid receptor; TIF, transcription intermediary factor; VP16, VP16 activation domain.

Received for publication January 7, 2005. Accepted for publication March 7, 2005.


    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 RESULTS
 DISCUSSION
 MATERIALS AND METHODS
 REFERENCES
 

  1. McKenna NJ, Nawaz Z, Tsai SY, Tsai M-J, O’Malley BW 1998 Distinct steady-state nuclear receptor coregulator complexes exist in vivo. Proc Natl Acad Sci USA 95:11697–11702[Abstract/Free Full Text]
  2. Underhill C, Qutob MS, Yee SP, Torchia J 2000 A novel nuclear receptor corepressor complex, N-CoR, contains components of the mammalian SWI/SNF complex and the corepressor KAP-1. J Biol Chem 275:40463–40470[Abstract/Free Full Text]
  3. Huang EY, Zhang J, Miska EA, Guenther MG, Kouzarides T, Lazar MA 2000 Nuclear receptor corepressors partner with class II histone deacetylases in a Sin3-independent repression pathway. Genes Deve 14:45–54
  4. Li J, Wang J, Nawaz Z, Liu JM, Qin J, Wong J 2000 Both corepressor proteins SMRT and N-CoR exist in large protein complexes containing HDAC3. EMBO J 19:4342–4350[Abstract/Free Full Text]
  5. Horlein AJ, Naar AM, Heinzel T, Torchia J, Gloss B, Kurokawa R, Ryan A, Kamei Y, Soderstrom M, Glass CK, Rosenfeld MG 1995 Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor. Nature 377:397–404[CrossRef][Medline]
  6. Chen JD, Evans RM 1995 A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377:454–457[CrossRef][Medline]
  7. Dotzlaw H, Moehren U, Mink S, Cato AC, Iniguez LJA, Baniahmad A 2002 The amino terminus of the human AR is target for corepressor action and antihormone agonism. Mol Endocrinol 16:661–673[Abstract/Free Full Text]
  8. Fu M, Wang C, Wang J, Zhang X, Sakamaki T, Yeung YG, Chang C, Hopp T, Fuqua SA, Jaffray E, Hay RT, Palvimo JJ, Janne OA, Pestell RG 2002 Androgen receptor acetylation governs trans activation and MEKK1-induced apoptosis without affecting in vitro sumoylation and trans-repression function. Mol Cell Biol 22:3373–3388[Abstract/Free Full Text]
  9. Smith CL, Nawaz Z, O’Malley BW 1997 Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4-hydroxytamoxifen. Mol Endocrinol 11:657–666[Abstract/Free Full Text]
  10. Zhang X, Jeyakumar M, Petukhow S, Bagchi MK 1998 A nuclear receptor corepressor modulates transcriptional activity of antagonist-occupied steroid hormone receptor. Mol Endocrinol 12:513–524[Abstract/Free Full Text]
  11. Schulz M, Eggert M, Baniahmad A, Dostert A, Heinzel T, Renkawitz R 2002 RU486-induced glucocorticoid receptor agonism is controlled by the receptor N terminus and by corepressor binding. J Biol Chem 277:26238–26243[Abstract/Free Full Text]
  12. Wang Q, Blackford Jr JA, Song L-N, Huang Y, Simons Jr SS 2004 Equilibrium interactions of corepressors and coactivators modulate the properties of agonist and antagonist complexes of glucocorticoid receptors. Mol Endocrinol 18:1376–1395[Abstract/Free Full Text]
  13. Wang Q, Richter WF, Anzick SL, Meltzer PS, Simons Jr SS 2004 Modulation of transcriptional sensitivity of mineralocorticoid and estrogen receptors. J Steroid Biochem Mol Biol 91:197–210[CrossRef][Medline]
  14. Jackson TA, Richer JK, Bain DL, Takimoto GS, Tung L, Horwitz KB 1997 The partial agonist activity of antagonist-occupied steroid receptors is controlled by a novel hinge domain-binding coactivator L7/SPA and the corepressors N-CoR or SMRT. Mol Endocrinol 11:693–705[Abstract/Free Full Text]
  15. Wagner BL, Norris JD, Knotts TA, Weigel NL, McDonnell DP 1998 The nuclear corepressors NCoR and SMRT are key regulators of both ligand- and 8-bromo-cyclic AMP-dependent transcriptional activity of the human progesterone receptor. Mol Cell Biol 18:1369–1378[Abstract/Free Full Text]
  16. Simons Jr SS 2003 The importance of being varied in steroid receptor transactivation. Trends Physiol Sci 24:253–259
  17. Szapary D, Huang Y, Simons Jr SS 1999 Opposing effects of corepressor and coactivators in determining the dose-response curve of agonists, and residual agonist activity of antagonists, for glucocorticoid receptor regulated gene expression. Mol Endocrinol 13:2108–2121[Abstract/Free Full Text]
  18. Giannoukos G, Szapary D, Smith CL, Meeker JEW, Simons Jr SS 2001 New antiprogestins with partial agonist activity: potential selective progesterone receptor modulators (SPRMs) and probes for receptor- and coregulator-induced changes in progesterone receptor induction properties. Mol Endocrinol 15:255–270[Abstract/Free Full Text]
  19. Song L-N, Huse B, Rusconi S, Simons Jr SS 2001 Transactivation specificity of glucocorticoid vs. progesterone receptors: role of functionally different interactions of transcription factors with amino- and carboxyl-terminal receptor domains. J Biol Chem 276:24806–24816[Abstract/Free Full Text]
  20. Cheng S, Brzostek S, Lee SR, Hollenberg AN, Balk SP 2002 Inhibition of the dihydrotestosterone-activated androgen receptor by nuclear receptor corepressor. Mol Endocrinol 16:1492–1501[Abstract/Free Full Text]
  21. Karim FD, Thummel CS 1992 Temporal coordination of regulatory gene expression by the steroid hormone ecdysone. EMBO J 11:4083–4093[Abstract]
  22. Gurdon JB, Bourillot PY 2001 Morphogen gradient interpretation. Nature 413:797–803[CrossRef][Medline]
  23. Dubrulle J, Pourquie O 2004 fgf8 mRNA decay establishes a gradient that couples axial elongation to patterning in the vertebrate embryo. Nature 427:419–422[CrossRef][Medline]
  24. Robyr D, Wolffe AP, Wahli W 2000 Nuclear hormone receptor coregulators in action: diversity for shared tasks. Mol Endocrinol 14:329–347[Free Full Text]
  25. Hu X, Lazar MA 1999 The CoRNR motif controls the recruitment of corepressors by nuclear hormone receptors. Nature 402:93–96[CrossRef][Medline]
  26. Nagy L, Kao H-Y, Love JD, Li C, Banayo E, Gooch JT, Krishna V, Chatterjee K, Evans RM, Schwabe JWR 1999 Mechanism of corepressor binding and release from nuclear hormone receptors. Genes Dev 13:3209–3216[Abstract/Free Full Text]
  27. Perissi V, Staszewski LM, McInerney EM, Kurokawa R, Krones A, Rose DW, Lambert MH, Milburn MV, Glass CK, Rosenfeld MG 1999 Molecular determinants of nuclear receptor-corepressor interaction. Genes Dev 13:3198–3208[Abstract/Free Full Text]
  28. He Y, Szapary D, Simons Jr SS 2002 Modulation of induction properties of glucocorticoid receptor-agonist and -antagonist complexes by coactivators involves binding to receptors but is independent of ability of coactivators to augment transactivation. J Biol Chem 277:49256–49266[Abstract/Free Full Text]
  29. Oshima H, Simons Jr SS 1993 Sequence-selective interactions of transcription factor elements with tandem glucocorticoid-responsive elements at physiological steroid concentrations. J Biol Chem 268:26858–26865[Abstract/Free Full Text]
  30. Szapary D, Xu M, Simons Jr SS 1996 Induction properties of a transiently transfected glucocorticoid-responsive gene vary with glucocorticoid receptor concentration. J Biol Chem 271:30576–30582[Abstract/Free Full Text]
  31. Zeng H, Plisov SY, Simons Jr SS 2000 Ability of the glucocorticoid modulatory element (GME) to modify glucocorticoid receptor transactivation indicates parallel pathways for the expression of GME and glucocorticoid response element activities. Mol Cell Endocrinol 162:221–234[CrossRef][Medline]
  32. Chen S, Sarlis NJ, Simons Jr SS 2000 Evidence for a common step in three different processes for modulating the kinetic properties of glucocorticoid receptor-induced gene transcription. J Biol Chem 275:30106–30117[Abstract/Free Full Text]
  33. Giangrande PH, Pollio G, McDonnell DP 1997 Mapping and characterization of the functional domains responsible for the differential activity of the A and B isoforms of the human progesterone receptor. J Biol Chem 272:32889–32900[Abstract/Free Full Text]
  34. Hu X, Li Y, Lazar MA 2001 Determinants of CoRNR-dependent repression complex assembly on nuclear hormone receptors. Mol Cell Biol 21:1747–1758[Abstract/Free Full Text]
  35. Lavinsky RM, Jepsen K, Heinzel T, Torchia J, Mullen T-M, Schiff R, Del-Rio AL, Ricote M, Ngo S, Gemsch J, Hilsenbeck SG, Osborne CK, Glass CK, Rosenfeld MG, Rose DW 1998 Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR and SMRT complexes. Proc Natl Acad Sci USA 95:2920–2925[Abstract/Free Full Text]
  36. Dahmer MK, Housley PR, Pratt WB 1984 Effects of molybdate and endogenous inhibitors on steroid-receptor inactivation, transformation, and translocation. Ann Rev Physiol 46:67–81[CrossRef][Medline]
  37. Pratt WB, Toft DO 1997 Steroid receptor interactions with heat shock protein and immunophilin chaperones. Endocr Rev 18:306–360[Abstract/Free Full Text]
  38. McKenna NJ, O’Malley BW 2002 Combinatorial control of gene expression by nuclear receptors and coregulators. Cell 108:465–474[CrossRef][Medline]
  39. Stancato LF, Hutchison KA, Krishna P, Pratt WB 1996 Animal and plant cell lysates share a conserved chaperone system that assembles the glucocorticoid receptor into a functional heterocomplex with hsp90. Biochemistry 35:554–561[CrossRef][Medline]
  40. de Felipe KS, Carter BT, Althoff EA, Cornish VW 2004 Correlation between ligand-receptor affinity and the transcription readout in a yeast three-hybrid system. Biochemistry 43:10353–10363[CrossRef][Medline]
  41. Giangrande PH, Kimbrel EA, Edwards DP, McDonnell DP 2000 The opposing transcriptional activities of the two isoforms of the human progesterone receptor are due to differential cofactor binding. Mol Cell Biol 20:3102–3115[Abstract/Free Full Text]
  42. Simons Jr SS 1994 Function/activity of specific amino acids in glucocorticoid receptors. Vitam Horm 48:49–130
  43. Ballare C, Uhrig M, Bechtold T, Sancho E, Di DM, Migliaccio A, Auricchio F, Beato M 2003 Two domains of the progesterone receptor interact with the estrogen receptor and are required for progesterone activation of the c-Src/Erk pathway in mammalian cells. Mol Cell Biol 23:1994–2008[Abstract/Free Full Text]
  44. Hollenberg SM, Giguere V, Segui P, Evans RM 1987 Colocalization of DNA-binding and transcriptional activation functions in the human glucocorticoid receptor. Cell 49:39–46[CrossRef][Medline]
  45. Iniguez-Lluhi JA, Lou DI, Yamamoto KR 1997 Three amino acid substitutions selectively disrupt the activation but not the repression function of the glucocorticoid receptor N terminus. J Biol Chem 272:4149–4156[Abstract/Free Full Text]
  46. Auboeuf D, Honig A, Berget SM, O’Malley BW 2002 Coordinate regulation of transcription and splicing by steroid receptor coregulators. Science 298:416–419[Abstract/Free Full Text]
  47. Gill G 2004 SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev 18:2046–2059[Abstract/Free Full Text]
  48. Tian S, Poukka H, Palvimo JJ, Janne OA 2002 Small ubiquitin-related modifier-1 (SUMO-1) modification of the glucocorticoid receptor. Biochem J 367:907–911[CrossRef][Medline]
  49. Abdel-Hafiz H, Takimoto GS, Tung L, Horwitz KB 2002 The inhibitory function in human progesterone receptor N termini binds SUMO-1 protein to regulate autoinhibition and transrepression. J Biol Chem 277:33950–33956[Abstract/Free Full Text]
  50. Chauchereau A, Amazit L, Quesne M, Guiochon-Mantel A, Milgrom E 2003 Sumoylation of the progesterone receptor and of the steroid receptor coactivator SRC-1. J Biol Chem 278:12335–12343[Abstract/Free Full Text]
  51. Wan Y, Nordeen SK 2002 Overlapping but distinct gene regulation profiles by glucocorticoids and progestins in human breast cancer cells. Mol Endocrinol 16:1204–1214[Abstract/Free Full Text]
  52. Willmann T, Beato M 1986 Steroid-free glucocorticoid receptor binds specifically to mouse mammary tumour virus DNA. Nature 324:688–691[CrossRef][Medline]
  53. Simons Jr SS 1979 Factors influencing association of glucocorticoid receptor-steroid complexes with nuclei, chromatin, and DNA: interpretation of binding data. In: Baxter JD, Rousseau GG, eds. Glucocorticoid hormone action. Berlin: Springer-Verlag; 161–187
  54. Kaul S, Murphy PJM, Chen J, Brown L, Pratt WB, Simons Jr SS 2002 Mutations at positions 547–553 of rat glucocorticoid receptors reveal that hsp90 binding requires the presence, but not defined composition, of a seven amino acid sequence at the amino terminus of the ligand binding domain. J Biol Chem 277:36223–36232[Abstract/Free Full Text]
  55. Distelhorst CW, Benutto BM 1985 Activation of the rat liver cytosol glucocorticoid receptor by Sephacryl S-300 filtration in the presence and absence of molybdate. Physical properties of the receptor and evidence for an activation inhibitor. J Biol Chem 260:2153–2159[Abstract]
  56. Cavanaugh AH, Simons Jr SS 1990 Glucocorticoid receptor binding to calf thymus DNA. 2. Role of a DNA-binding activity factor in receptor heterogeneity and a multistep mechanism of receptor activation. Biochemistry 29:996–1002[CrossRef][Medline]
  57. Gee AC, Carlson KE, Martini PGV, Katzenellenbogen BS, Katzenellenbogen JA 1999 Coactivator peptides have a differential stabilizing effect on the binding of estrogens and antiestrogens with the estrogen receptor. Mol Endocrinol 13:1912–1923[Abstract/Free Full Text]
  58. Liu J, Knappenberger KS, Kack H, Andersson G, Nilsson E, Dartsch C, Scott CW 2003 A homogeneous in vitro functional assay for estrogen receptors: coactivator recruitment. Mol Endocrinol 17:346–355[Abstract/Free Full Text]
  59. Danze PM, Richard C, Formstecher P, Dautrevaux M 1990 Steroid receptor exchange assay in the presence of acetonitrile: application to the study of glucocorticoid- and anti-glucocorticoid-receptor complexes. Steroids 55:10–16[CrossRef][Medline]
  60. Cho S, Blackford Jr JA, Simons Jr SS 2005 Role of activation function domain 1, DNA binding, and coactivator in the expression of partial agonist activity of glucocorticoid receptor complexes. Biochemistry, 44:3547–3561[CrossRef][Medline]
  61. Tetel MJ, Giangrande PH, Leonhardt SA, McDonnell DP, Edwards DP 1999 Hormone-dependent interaction between the amino- and carboxyl-terminal domains of progesterone receptor in vitro and in vivo. Mol Endocrinol 13:910–924[Abstract/Free Full Text]
  62. Cho S, Blackford Jr JA, Kagan BL, Simons Jr SS 2005 Glucocorticoid receptor ligand binding domain is sufficient for the modulation of both the dose-response curve of receptor-agonist complexes and the partial agonist activity of receptor-antisteroid complexes by glucocorticoid receptors, coactivator TIF2, and Ubc9. Mol Endocrinol 19:290–311[Abstract/Free Full Text]




This Article
Abstract
Full Text (PDF)
All Versions of this Article:
19/6/1483    most recent
Author Manuscript (PDF)
Purchase Article
View Shopping Cart
Alert me when this article is cited
Alert me if a correction is posted
Citation Map
Services
Email this article to a friend
Similar articles in this journal
Similar articles in PubMed
Alert me to new issues of the journal
Download to citation manager
Request Copyright Permission
Google Scholar
Articles by Wang, D.
Articles by Simons, S. S.
Articles citing this Article
PubMed
PubMed Citation
Articles by Wang, D.
Articles by Simons, S. S., Jr.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
Endocrinology Endocrine Reviews J. Clin. End. & Metab.
Molecular Endocrinology Recent Prog. Horm. Res. All Endocrine Journals