Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(43): 39843-51, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11546784

ABSTRACT

Progesterone receptors (PR) contain three activation functions (AFs) that together define the extent to which they regulate transcription. AF1 and AF2 are common to the two isoforms of PR, PR-A and PR-B, whereas AF3 lies within the N-terminal 164 amino acids unique to PR-B, termed the "B-upstream segment" (BUS). To define the BUS regions that contribute to AF3 function, we generated a series of deletion and amino acid substitution mutants and tested them in three backgrounds as follows: BUS alone fused to the PR DNA binding domain (BUS-DBD), the entire PR-B N terminus linked to its DBD (NT-B), and full-length PR-B. Analyses of these mutants identified two regions in BUS whose loss reduces AF3 activity by more than 90%. These are associated with amino acids 54-90 (R1) and 120-154 (R2). R1 contains a consensus (55)LXXLL(59) motif (L1) identical to ones found in nuclear receptor co-activators. R2 is adjacent to a second nuclear receptor box (L2) at (115)LXXLL(119) and contains a conserved tryptophan (Trp-140). Their mutation completely disrupts AF3 activity in a promoter and cell type-independent manner. Critical mutations elicited similar effects on all three B-receptor backgrounds. This underscores the probability that these mutations alter a process linking BUS structure to the function of full-length PR-B in a fundamental way.


Subject(s)
Receptors, Progesterone/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Amino Acid Motifs , Binding Sites , Humans , Membrane Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments , Sequence Deletion , Transcription, Genetic
2.
J Biol Chem ; 276(26): 23825-31, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11328821

ABSTRACT

To understand the basis for functional differences between the two human progesterone receptors (PR), we have carried out a detailed biochemical and biophysical analysis of the N-terminal region of each isoform. Extending our previous work on the A-isoform (Bain, D. L, Franden, M. A., McManaman, J. L., Takimoto, G. S., and Horwitz, K. B. (2000) J. Biol. Chem. 275, 7313-7320), here we present studies on the N-terminal region of the B-isoform (NT-B) and compare its properties to its A-receptor counterpart (NT-A). As seen previously with NT-A, NT-B is quantitatively monomeric in solution, yet undergoes N-terminal-mediated assembly upon DNA binding. Limited proteolysis, microsequencing, and sedimentation analyses indicate that the B-isoform exists in a non-globular, extended conformation very similar to that of NT-A. Additionally, the 164 amino acids unique to the B-isoform (BUS) appear to be in a more extended conformation relative to sequences common to both receptors and do not exist as an independent structural domain. However, sedimentation studies of NT-A and NT-B show differences in the ensemble distribution of their conformational states. We hypothesize that isoform-specific functional differences are not due to structural differences, per se. Rather, the transcriptional element BUS, or possibly other transcription factors, causes a redistribution of the conformational ensemble by stabilizing a more functionally active set of conformations in NT-B.


Subject(s)
Receptors, Progesterone/chemistry , Humans , Kinetics , Protein Conformation , Receptors, Progesterone/metabolism , Response Elements , Transcription, Genetic , Ultracentrifugation
3.
J Mol Endocrinol ; 24(2): 165-82, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10750018

ABSTRACT

Ligand-activated progesterone receptors (PR) bind to DNA at specific progesterone response elements by means of a DNA binding domain (DBD(PR)) containing two highly conserved zinc fingers. DNA-bound PRs regulate transcription via interaction with other nuclear proteins and transcription factors. We have now identified four HeLa cell nuclear proteins that copurify with a glutathionine-S-transferase-human DBD(PR )fusion protein. Microsequence and immunoblot analyses identified one of these proteins as the 113 kDa poly(ADP-ribose) polymerase. The three other proteins were identified as subunits of the DNA-dependent protein kinase (DNA-PK) holoenzyme: its DNA binding regulatory heterodimers consisting of Ku70 and Ku86, and the 460 kDa catalytic subunit, DNA-PK(CS). DNA-PK that was 'pulled-down' by DBD(PR) on the affinity resin was able to (1) autophosphorylate Ku70, Ku86, and DNA-PK(CS), (2) transphosphorylate DBD(PR), and (3) phosphorylate a DNA-PK-specific p53 peptide substrate. DNA-PK was also able to associate with the DBD of the yeast activator GAL4. However, neither a PR DBD mutant lacking a structured first zinc finger (DBD(CYS)) nor the core DBD of the estrogen receptor (DBD(ER)) copurified DNA-PK, suggesting the interaction is not non-specific for DBDs. Lastly, we found that DNA-PK copurified with full-length human PR transiently expressed in HeLa cells, suggesting that the human PR/DNA-PK complex can assemble in vivo. These data show that DNA-PK and DBD(PR) interact, that DBD(PR) is a phosphorylation substrate of DNA-PK, and suggest a potential role for DNA-PK in PR-mediated transcription.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Progesterone/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Autoantigens/isolation & purification , Autoantigens/metabolism , Binding Sites , DNA-Activated Protein Kinase , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Glutathione Transferase/genetics , HeLa Cells , Humans , Ku Autoantigen , Ligands , Methionine/metabolism , Molecular Sequence Data , Phosphorylation , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/isolation & purification , Protein Biosynthesis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/isolation & purification , Receptors, Progesterone/chemistry , Receptors, Progesterone/isolation & purification , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
4.
J Biol Chem ; 275(10): 7313-20, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10702302

ABSTRACT

The role of the N-terminal region in nuclear receptor function was addressed by a biochemical and biophysical analysis of the progesterone receptor A-isoform lacking only the hormone binding domain (NT-A). Sedimentation studies demonstrate that NT-A is quantitatively monomeric, with a highly asymmetric shape. Contrary to dogma, the N-terminal region is structured as demonstrated by limited proteolysis. However, N-terminal structure is strongly stabilized by the DNA binding domain, possibly explaining the lack of structure seen in isolated activation domains. Upon DNA binding, NT-A undergoes N-terminal mediated assembly, suggestive of DNA-induced allostery, and consistent with changes in protease accessibility of sites outside the DNA binding domain. Microsequencing reveals that protease-accessible regions are limited to previously identified phosphorylation motifs and to functional domain boundaries.


Subject(s)
DNA/metabolism , Receptors, Progesterone/chemistry , Binding Sites , Endopeptidases/pharmacology , Humans , Molecular Weight , Protein Folding , Response Elements , Transcription, Genetic
5.
J Steroid Biochem Mol Biol ; 69(1-6): 45-50, 1999.
Article in English | MEDLINE | ID: mdl-10418980

ABSTRACT

Pharmacological antagonists of steroid receptor action had been thought to exert their effects by a passive mechanism driven principally by the ability of the antagonist to compete with agonist for the ligand binding site. However, recent analyses of antagonist-occupied receptor function suggest a more complex picture. Antagonists can be subdivided into two groups, type I, or pure antagonists, and type II, or mixed antagonists that can have variable transcriptional activity based upon differential dimerization and DNA binding properties. This led us to propose that receptor antagonism may not simply be a passive competition for the ligand binding site, but may, in some cases, involve active recruitment of corepressor or coactivator proteins to produce a mixed transcriptional phenotype. We used a yeast two-hybrid screen to identify proteins that interact specifically with antagonist-occupied receptors. Two proteins have been characterized: L7/SPA, a ribosome-associated protein that is localized in both the cytoplasm and nucleus, but with no known extranucleolar nuclear function; and hN-CoR, the human homolog of the mouse thyroid receptor corepressor mN-CoR. In in vivo transcription assays we show that L7/SPA enhances the partial agonist activity of type II mixed antagonists, and that N-CoR and the related corepressor, SMRT, suppresses it. The coregulators do not affect agonists or pure antagonists. Moreover, the net agonist activity seen with mixed antagonists is a function of the ratio of coactivator to corepressor. Based upon these results, we proposed that in breast tumors the inappropriate agonist activity seen with therapeutic antagonists such as tamoxifen is responsible for the hormone-resistant state. To confirm this, we are quantitating coactivator/corepressor ratios in breast tumor cells lines and clinical breast cancers. Results should provide new insights into the mechanisms underlying the progression of breast cancer to hormone resistance, and may suggest strategies for delaying or reversing this process.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/physiopathology , Receptors, Steroid/antagonists & inhibitors , Tamoxifen/pharmacology , Transcription, Genetic , Animals , Drug Resistance, Neoplasm , Humans , Receptors, Steroid/genetics
6.
Breast Cancer Res Treat ; 48(3): 231-41, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9598870

ABSTRACT

Progesterone, through its nuclear receptors (PR), regulates the development and growth of breast cancers. PR also serve as markers of hormone dependence and prognosis in patients with this disease, and functional PR are required to mediate the antiproliferative effects of progestin therapies. We find that normal and malignant breast cells and tissues can express anomalous forms of PR transcripts. We have isolated four variant PR mRNAs that contain precise deletions of exons encoding sections of the DNA- and hormone-binding domains. The transcripts lack exon 2 (PRdelta2), exon 4 (PRdelta4), exon 6 (PRdelta6), or exons 5 and 6 (PRdelta5,6). On immunoblots, PRdelta4, delta6. and delta5, 6 cloned into the background of the PR A-isoform comigrate with similar proteins present in breast tumor extracts; delta6 and delta5, 6 are dominant-negative transcriptional inhibitors of wild-type A- and B-receptors. We propose that expression of variant PR can compromise the accuracy of receptor measurements as markers of hormone-dependent cancers, and can modify the responses of tumors to progestin therapies.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Progesterone/metabolism , Breast/drug effects , Breast/metabolism , Breast Neoplasms/drug therapy , Drug Resistance , Exons , Female , Humans , Progestins/therapeutic use , RNA, Messenger/analysis , Receptors, Progesterone/drug effects , Receptors, Progesterone/genetics , Tumor Cells, Cultured
7.
J Biol Chem ; 273(10): 5455-60, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9488667

ABSTRACT

The B-isoform of human progesterone receptors (PR) contains three activation functions (AF3, AF1, and AF2), two of which (AF1 and AF2) are shared with the A-isoform. AF3 is in the B-upstream segment (BUS), the far N-terminal 164 amino acids of B-receptors; AF1 is in the 392-amino acid N-terminal region common to both receptors; and AF2 is in the C-terminal hormone binding domain. B-receptors are usually stronger transactivators than A-receptors due to transcriptional synergism between AF3 and one of the two downstream AFs. We now show that the N terminus of PR common to both isoforms contains an inhibitory function (IF) located in a 292-amino acid segment lying upstream of AF1. IF represses the activity of A-receptors but is not inhibitory in the context of B-receptors due to constraints imparted by BUS. As a result, IF inhibits AF1 or AF2 but not AF3, regardless of the position of IF relative to BUS. IF is functionally independent and strongly represses transcription when it is fused upstream of estrogen receptors. These data demonstrate the existence of a novel, transferable inhibitory function, mapping to the PR N terminus, which begins to assign specific roles to this large undefined region.


Subject(s)
Gene Expression Regulation/genetics , Receptors, Progesterone/chemistry , HeLa Cells , Humans , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Receptors, Estrogen/chemistry , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Suppression, Genetic/physiology , Trans-Activators/chemistry , Trans-Activators/pharmacology , Transcription, Genetic/genetics , Transfection/genetics
8.
Genes Chromosomes Cancer ; 20(1): 64-72, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9290956

ABSTRACT

The t(14;19)(q32.3;q13.1) is a recurring translocation found in the neoplastic cells of some patients with chronic lymphocytic leukemia (CLL) or other B-lymphocytic neoplasms. We previously cloned the translocation breakpoint junctions present in the leukemic cells from three such patients and identified a gene, BCL3, whose transcription is increased as a result of the translocation. In the present paper, we describe three additional patients with the t(14;19), one with lymphoma and two with CLL, and report the cloning and sequencing of the breakpoint junction in one of these patients as well as in a previously reported patient. We and others have found that the breakpoints on chromosome 14, with one exception, fall within the switch region upstream of the immunoglobulin heavy chain C alpha 1 or C alpha 2 sequences. Several of the breaks within chromosome 19 fall immediately upstream of the BCL3 gene, but several others are more than 16 kb 5' of the gene. Most patients with CLL and the t(14;19) also show trisomy 12.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 19/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Proto-Oncogene Proteins/genetics , Translocation, Genetic , Adult , Aged , B-Cell Lymphoma 3 Protein , Base Sequence , Blotting, Southern , Chromosome Mapping , Cloning, Molecular , Electrophoresis, Gel, Pulsed-Field , Female , Gene Rearrangement , Humans , Karyotyping , Leukemia, B-Cell/genetics , Lymphoma, B-Cell/genetics , Male , Molecular Sequence Data , Sequence Analysis, DNA , Transcription Factors
9.
Mol Endocrinol ; 11(6): 693-705, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9171233

ABSTRACT

Steroid receptor antagonists, such as the antiestrogen tamoxifen or the antiprogestin RU486, can have inappropriate agonist-like effects in tissues and tumors. To explain this paradox we postulated that coactivators are inadvertently brought to the promoters of DNA-bound, antagonist-occupied receptors. The human (h) progesterone receptor (PR) hinge-hormone binding domain (H-HBD) was used as bait in a two-hybrid screen of a HeLa cDNA library, in which the yeast cells were treated with RU486. We have isolated and characterized two interesting steroid receptor-interacting proteins that regulate transcription in opposite directions. The first is L7/SPA, a previously described 27-kDa protein containing a basic region leucine zipper domain, having no known nuclear function. When coexpressed with tamoxifen-occupied estrogen receptors (hER) or RU486-occupied hPR or glucocorticoid receptors (hGR), L7/SPA increases the partial agonist activity of the antagonists by 3- to 10-fold, but it has no effect on agonist-mediated transcription. The interaction of L7/SPA with hPR maps to the hinge region, and indeed, the hPR hinge region squelches L7/SPA-dependent induction of antagonist-mediated transcription. Interestingly, pure antagonists that lack partial agonist effects, such as the antiestrogen ICI164,384 or the antiprogestin ZK98299, cannot be up-regulated by L7/SPA. We also isolated, cloned, and sequenced the human homolog (hN-CoR) of the 270-kDa mouse (m) thyroid/retinoic acid receptor corepressor. Binding of hN-CoR maps to the hPR-HBD. mN-CoR, and a related human corepressor, SMRT, suppress RU486 or tamoxifen-mediated partial agonist activity by more than 90%. This suppression is completely squelched by overexpression of the hPR H-HBD. Additionally, both corepressors reverse the antagonist-dependent transcriptional up-regulation produced by L7/SPA. Our data suggest that the direction of transcription by antagonist-occupied steroid receptors can be controlled by the ratio of coactivators to corepressors recruited to the transcription complex by promoter-bound receptors. In normal tissues and in hormone-resistant breast cancers in which the agonist activity of mixed antagonists predominates, steroid receptors may be preferentially bound by coactivators. This suggests a strategy by which such partial agonist activity can be eliminated and by which candidate receptor ligands can be screened for this activity.


Subject(s)
DNA-Binding Proteins/physiology , Mifepristone/pharmacology , Nuclear Proteins/physiology , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Repressor Proteins/physiology , Ribosomal Proteins/physiology , Animals , Base Sequence , Binding Sites , COS Cells , DNA, Complementary , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Leucine Zippers , Mice , Mifepristone/antagonists & inhibitors , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Repressor Proteins/genetics , Ribosomal Proteins/isolation & purification , Yeasts
10.
Mol Endocrinol ; 10(10): 1167-77, 1996 Oct.
Article in English | MEDLINE | ID: mdl-9121485

ABSTRACT

The nuclear receptors belong to a superfamily of proteins, many of which are ligand-regulated, that bind to specific DNA sequences and control specific gene transcription. Recent data show that, in addition to contacting the basal transcription machinery directly, nuclear receptors inhibit or enhance transcription by recruiting an array of coactivator or corepressor proteins to the transcription complex. In this review we define the properties of these putative coregulatory factors; we describe the basal and coregulatory factors that are currently known to interact with nuclear receptors; we suggest various mechanisms by which coactivators and corepressors act; we discuss issues that are raised by the presence of multiple, perhaps competing, coregulatory factors; and we speculate how these additional regulatory layers may explain the heterogeneity of hormone responses that are observed in normal and malignant tissues.


Subject(s)
Receptors, Cytoplasmic and Nuclear , Signal Transduction , Animals , Humans
11.
J Biol Chem ; 271(23): 13308-16, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8662865

ABSTRACT

To study the function of human progesterone receptor (hPR) phosphorylation, we have tested four sets of serine to alanine substitution mutants: 10 serine clusters, located in regions common to both hPR isoforms (the M-series mutants) were mutated in A-receptors and B-receptors; 6 serine clusters located in the B-upstream segment (BUS; the B-series mutants) were mutated individually and collectively and cloned into B-receptors and into BUS-DBD-NLS, a constitutive transactivator, in which the AF3 function of BUS is fused to the DNA binding domain (DBD) and nuclear localization signal (NLS) of hPR. Transcription by most of the M-series mutants resembles that of wild-type A- or B-receptors. Mutation of 3 sites, Ser190 at the N terminus of A-receptors, a cluster of serines just upstream of the DBD, or Ser676 in the hinge region, inhibits transcription by 20-50% depending on cell or promoter context. These sites lie outside the AF1 activation function. M-series mutants are substrates for a hormone-dependent phosphorylation step, and they all bind well to DNA. Progressive mutation of the B-series clusters leads to the gradual dephosphorylation of BUS, but only the 6-site mutant, involving 10 serine residues, is completely dephosphorylated. These data suggest that in BUS alternate serines are phosphorylated or dephosphorylated at any time. However, even when BUS is completely dephosphorylated, both BUS-DBD-NLS and full-length B-receptors remain strong transactivators. Mutant B-receptors also do not acquire the dominant negative properties of A-receptors, and they retain the ability to activate transcription in synergy with 8-Br-cAMP and antiprogestins. We conclude that phosphorylation has subtle effects on the complex transcriptional repertoire that distinguishes the two hPR isoforms and does not influence transactivation mediated by AF1 or AF3, but subserves other functions.


Subject(s)
DNA/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Animals , Binding Sites/genetics , Cell Line , Cloning, Molecular , HeLa Cells , Humans , Molecular Structure , Mutagenesis, Site-Directed , Phosphorylation , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic
12.
Acta Oncol ; 35(2): 129-40, 1996.
Article in English | MEDLINE | ID: mdl-8639307

ABSTRACT

Endocrine therapy used either prophylactically or therapeutically for the treatment of locally advanced or metastatic breast cancers offers many advantages to patients whose tumors contain functional estrogen (ER) and progesterone (PR) receptors. The range of treatments defined as endocrine include surgical ablation of endocrine glands, administration of pharmacologic doses of steroid hormones, chemical blockade of steroid hormone biosynthesis, and inhibition of endogenous steroid hormone action at the tumor with synthetic antagonists. The last of these approaches is the most widely used, making the antiestrogen tamoxifen the preferred first-line therapeutic agent for treatment of hormone-dependent metastatic breast cancer. The wide-spread use of tamoxifen reflects its efficacy and low toxicity, and the fact that it makes good physiological sense to block the local proliferative effects of estrogens directly at the breast. But are estrogens the only hormones with a proliferative impact on the breast and on breast cancers? This chapter focuses on evidence that progesterone also has proliferative actions in the breast; on preliminary data showing that progesterone antagonists may be new tools for the management of metastatic breast cancer; and on recent data suggesting that antiprogestin-occupied PR have novel mechanisms of action that bear on tissue specificity and development of hormone resistance.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Progestins/antagonists & inhibitors , Progestins/physiology , Animals , Breast Neoplasms/pathology , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/physiopathology , Progesterone/antagonists & inhibitors
13.
J Steroid Biochem Mol Biol ; 53(1-6): 9-17, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7626523

ABSTRACT

When hormone antagonists have unexpected agonist-like effects, the clinical consequences are grave. We describe novel molecular mechanisms by which antiprogestin-occupied progesterone receptors behave like agonists. These mechanisms include agonist-like transcriptional effects that do not require receptor binding to DNA at progesterone response elements, or that result from cross-talk between progesterone receptor and other signalling pathways. We discuss the complex structural organization of progesterone receptors and demonstrate that the B-receptor isoform has a unique third activation domain that may confer agonist-like properties in the presence of antiprogestins. By contrast, the A-receptor isoform is a dominant-negative inhibitor. We argue that these novel mechanisms play a role in the apparent hormone resistance of breast cancers and the variable tissue-specific responses to progestins.


Subject(s)
Progestins/antagonists & inhibitors , Receptors, Progesterone/drug effects , Cell Line , Cyclic AMP/pharmacology , DNA-Binding Proteins/genetics , Humans , In Vitro Techniques , Promoter Regions, Genetic , Receptors, Progesterone/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation , Tumor Cells, Cultured
14.
Ciba Found Symp ; 191: 235-49; discussion 250-3, 1995.
Article in English | MEDLINE | ID: mdl-8582200

ABSTRACT

When hormone antagonists have inappropriate agonist-like effects, the clinical consequences are grave. We describe novel molecular mechanisms by which antiprogestin-occupied progesterone receptors behave like agonists. These mechanisms include agonist-like transcriptional effects that do not require receptor binding to DNA at progesterone response elements, or that result from cross-talk between progesterone receptors and other signalling pathways. We discuss the complex structural organization of progesterone receptors, and demonstrate that the B receptor isoform has a unique third activation domain that may confer agonist-like properties in the presence of antiprogestins, whereas the A receptor isoform is a dominant-negative inhibitor. We argue that these novel mechanisms play a role in the apparent hormone resistance of breast cancers and the variable tissue-specific responses to antagonists.


Subject(s)
Hormone Antagonists/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cyclic AMP/physiology , Humans , Transcriptional Activation , Tumor Cells, Cultured
15.
J Steroid Biochem Mol Biol ; 51(5-6): 241-50, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7826885

ABSTRACT

Human progesterone receptors (hPR) exist as two isoforms: 120 kDa B-receptors (hPRB) and N-terminally truncated 94 kDa A-receptors (hPRA). When transfected separately, each isoform exhibits different transcriptional properties that are ligand- and promoter-specific. In human target tissues, both receptor isoforms are present, so that a mixture of three dimeric species, A/A, A/B, and B/B, bind to DNA at progesterone response elements (PRE), and regulate transcription. To study the transcriptional phenotype of pure A/B heterodimers uncontaminated by A/A or B/B homodimers, we exploited the property of the leucine zipper (zip) domains of fos and jun, to form pure heterodimers. Chimeric constructs were made linking the zip of either c-fos or c-jun to the C-terminus of hPRB or hPRA (hPR-zip) to produce A-fos, B-fos, A-jun or B-jun. To determine whether the A- or B-isoform is functionally dominant in the A/B heterodimer, cells expressing hPR-zip chimeras were treated with the progestin antagonist RU486, which produces opposite transcriptional effects with the two isoforms. Gel mobility shift and immune co-precipitation assays show that in the presence of RU486 only pure heterodimers form between A-fos/B-jun or A-jun/B-fos, and bind DNA at PREs. Thus, in these pairs, interactions between the extrinsic fos/jun zipper domains override interactions between the intrinsic hPR dimerization domains. We find that under these conditions, antagonist-occupied B-zip homodimers stimulate transcription, while antagonist-occupied A-zip homodimers are inhibitory, and that pure A/B zip heterodimers have the inhibitory transcriptional phenotype of the A-zip homodimers. We conclude that, in pure heterodimers, A-receptors are dominant negative inhibitors of B-receptors. Additionally, the pure PR-zip heterodimers, unlike wild-type receptors, bind a PRE in the absence of hormone but do not activate transcription. Thus, PR dimerization and PRE binding are necessary but, without hormone, not sufficient to activate transcription.


Subject(s)
Leucine Zippers , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-jun/chemistry , Receptors, Progesterone/chemistry , Base Sequence , Binding, Competitive , Biopolymers/chemistry , Cell Line , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , HeLa Cells , Humans , Mifepristone/metabolism , Molecular Sequence Data , Promegestone/metabolism , Receptors, Progesterone/metabolism , Receptors, Progesterone/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Transcription, Genetic/physiology
16.
Mol Endocrinol ; 8(10): 1347-60, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7854352

ABSTRACT

Human progesterone target tissues contain two progesterone receptors: B-receptors (hPRB), which are 933 amino acids in length, and A-receptors (hPRA), which lack the N-terminal 164 amino acids. The two isoforms differ functionally when they are occupied by agonists or antagonists. We postulated that the unique 164-amino acid, B-upstream segment (BUS) is in part responsible for the functional differences between the two isoforms and have constructed a series of hPR expression vectors encoding BUS fused to isolated down-stream functional domains of the receptors. These include the two transactivation domains: activation function-1 (AF1), located in a 90-amino acid segment just up-stream of the DNA-binding domain (DBD) and nuclear localization signal (NLS), and AF2, located in the hormone-binding domain. BUS is a highly phosphorylated domain, and contains the serine residues responsible for the hPRB triplet protein structure. The construct containing BUS-DBD-NLS binds tightly to DNA when aided by accessory nuclear factors. In HeLa cells, BUS-DBD-NLS strongly and autonomously activates transcription of chloramphenicol acetyltransferase (CAT) from a promoter containing two progesterone response elements (PRE2-TATAtk-CAT). Transcription levels with BUS-DBD-NLS are equivalent to those seen with full-length hPRB, and are higher than those seen with hPRA. BUS specifically requires an intact hPR DBD to be transcriptionally active. DBD mutants that cannot bind DNA or whose DNA binding specificity has been switched to an estrogen response element cannot cooperate in BUS transcriptional activity. The function of BUS-DBD-NLS is promoter and cell specific. It does not transactivate a CAT reporter driven by the mouse mammary tumor virus promoter in HeLa cells and poorly transactivates PRE2-TATAtk-CAT in PR-negative T47D breast cancer cells. However, in the breast cancer cells, BUS-DBD-NLS transactivation of PRE2-TATAtk-CAT can be reconstituted by either elevating cellular levels of cAMP or linking BUS and DBD to AF1 or AF2 of hPR, each of which alone is also inactive in these cells. We conclude that hPRB contains a unique third activation function (AF3) located within BUS and requiring the functional DBD of hPR. Depending on the promoter or cell tested, AF3 can activate transcription autonomously, or it can functionally synergize with AF1 or AF2. Autonomous AF3 function may explain the unexpected transactivating actions of antiprogestin-occupied hPRB, an issue of importance in hormone-resistant breast cancers and in tissue-specific agonist-like effects of hormone antagonists.


Subject(s)
Peptide Fragments/physiology , Receptors, Progesterone/physiology , Transcriptional Activation , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Binding Sites , Chloramphenicol O-Acetyltransferase/genetics , DNA/metabolism , Drug Synergism , Genes, Reporter , HeLa Cells , Humans , Mammary Tumor Virus, Mouse/genetics , Peptide Fragments/pharmacology , Phosphorylation , Promoter Regions, Genetic , TATA Box , Transcription, Genetic
17.
Cancer Res ; 54(14): 3868-77, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8033109

ABSTRACT

Because progesterone antagonists are growth inhibitors, they are in Phase III clinical trials for the treatment of breast cancer. However, when cellular cAMP levels are elevated, some antiprogestins inappropriately activate transcription. We have proposed that hormone "resistance" may result from such unintended stimulation of breast cancer by antagonists. In transient expression systems, the two natural isoforms of human progesterone receptors (PR), B-receptors and truncated A-receptors, have dissimilar effects on agonist-mediated transcription. We show here that in the presence of 8-Br-cAMP, antiprogestin-occupied B-receptors but not A-receptors become transcriptional activators. Therefore, we developed new model systems to study each PR isoform independently in a breast cancer setting: (a) a stable PR-negative monoclonal subline (T47D-Y) of PR-positive T47D breast cancer cells was selected by flow cytometric PR screening. T47D-Y cells are PR-negative by immunoassays, by ligand binding assay, by growth resistance to progestins, by failure to bind a progesterone response element (PRE) in vitro, and by failure to transactivate PRE-regulated promoters; and (b) T47D-Y cells were stably transfected with expression vectors encoding one or the other PR isoform, and two monoclonal cell lines were selected that express either B-receptors (T47D-YB) or A-receptors (T47D-YA) at levels equal to those seen in natural T47D cells. The ectopically expressed receptors are properly phosphorylated, and like endogenously expressed receptors, they undergo ligand-dependent down-regulation. The expected B:B or A:A homodimers are present in cell extracts from each cell line, but A:B heterodimers are missing in both. In the presence of agonists, cAMP-dependent, transcriptional synergism of PRE-regulated promoters is seen in both cell lines. By contrast, in the presence of the antiprogestins RU486 or ZK112993, inappropriate transactivation occurs in YB cells but not in YA cells. The class of antiprogestins represented by ZK98299, which blocks PR binding to DNA, does not activate transcription in either cell line. We propose that these new cell lines are physiological models for the study of PR isoform-specific antiprogestin resistance in breast cancer.


Subject(s)
Breast Neoplasms/chemistry , Cyclic AMP/pharmacology , Progestins/antagonists & inhibitors , Receptors, Progesterone/analysis , Base Sequence , Breast Neoplasms/pathology , Female , Gonanes/pharmacology , Humans , Mifepristone/analogs & derivatives , Mifepristone/pharmacology , Molecular Sequence Data , Receptors, Progesterone/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured
18.
Mol Endocrinol ; 7(10): 1256-65, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8123133

ABSTRACT

When antagonist-occupied steroid receptors have agonist-like effects, the clinical consequences are grave. We present evidence that human progesterone B-receptors (hPRB) when occupied by progesterone antagonists, inappropriately activate transcription by an unusual mechanism that does not require the canonical progesterone response element (PRE). In HeLa cells cotransfected with a PRE-tk-chloramphenicol acetyltransferase reporter and a hPRB expression vector, strong transcription is seen not only when receptors are activated by the agonist R5020, but also in the presence of the three antiprogestins, RU486, ZK112993, and ZK98299. Human PRB occupied by ZK98299 do not bind to a PRE, suggesting that the transcriptional stimulation is independent of DNA binding. Indeed, a tk-chloramphenicol acetyltransferase promoter-reporter lacking the PRE loses transcriptional activation by the agonist, but retains transactivation by the three antagonists. The PRE-independent antagonist-induced transcription requires that hPRB have an intact DNA-binding domain, but hPR target gene specificity is not required, because a hPRB mutant that binds an estrogen response element still activates transcription. It appears that antagonist-occupied hPR activate transcription without binding to a PRE, perhaps by interacting with tethering proteins instead. Even a gene that is not a normal progesterone target could be aberrantly activated. Human cells contain equimolar amounts of hPRB and the N-terminally truncated natural isotype, hPRA. Unlike hPRB, hPRA are not transcriptionally activated by progesterone antagonists. We, therefore, tested the effects of antagonists when the two receptor isotypes are coexpressed and found that A-receptors can annul the inappropriate transcription by B-receptors. Thus, when both receptor forms are present, the hPRA phenotype is dominant. Moreover, pure hPRB/hPRA heterodimers, produced by fos/jun leucine zipper domain-hPR chimeras, also have the inactive transcriptional phenotype of hPRA. Our studies suggest not only that the two hPR isotypes are functionally quite different, but also that some of the agonist-like transcriptional effects of antagonist-occupied B-receptors proceed through novel mechanisms.


Subject(s)
DNA/metabolism , Progesterone/antagonists & inhibitors , Receptors, Progesterone/physiology , Transcription, Genetic , Binding Sites , Genes, Reporter , Genes, fos , Genes, jun , Gonanes/pharmacology , HeLa Cells , Humans , Leucine Zippers , Mifepristone/analogs & derivatives , Mifepristone/pharmacology , Phenotype , Progesterone/pharmacology , Promoter Regions, Genetic , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Recombinant Fusion Proteins , Transfection
19.
J Biol Chem ; 268(13): 9262-6, 1993 May 05.
Article in English | MEDLINE | ID: mdl-8387487

ABSTRACT

When steroid hormone antagonists have inappropriate agonist effects, the clinical consequences are grave. Progesterone antagonists bind to two naturally occurring isoforms of human progesterone receptors (hPR), hPRB and the NH2-terminally truncated hPRA, and usually inhibit agonist-stimulated transcription. It is shown here that elevation of cAMP levels in a human breast cancer cell line leads to the functional reversal of progesterone antagonist action. While hPR occupied by the antagonists RU486 and ZK112993 are transcriptionally inactive, the antagonist-occupied receptors become strong activators of transcription in the presence of 8-Br-cAMP. However, this functional switch does not occur with the progesterone antagonist ZK98299, which, unlike RU486 and ZK112993, is unable to induce hPR binding to DNA. This suggests that the 8-Br-cAMP-induced transcriptional reversal requires that the antagonist-occupied receptors be bound to DNA. Even with agonist-occupied hPR, addition of 8-Br-cAMP results in a synergistic increase in transcriptional activity. When hPRA alone are transiently expressed in COS-1 cells, transcription of a reporter gene is stimulated by the agonist R5020 and by 8-Br-cAMP and is synergistic when both are present; but the 8-Br-cAMP-dependent component of transcription proceeds in the absence of hPRA, in the absence of the progesterone response element, and in the presence of a DNA-binding domain mutant of hPRA that cannot bind to the progesterone response element. Additionally, under the intracellular conditions in which 8-Br-cAMP activates antagonist-hPR complexes, there is no protein kinase A-mediated phosphorylation of the receptors. We discuss a model in which a gene that is independently transcribed by cAMP-responsive factors and by hPR can be selected for positive or negative regulation on the transcription complex due to additive or cooperative interactions between the two DNA-bound factors.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Antineoplastic Agents/pharmacology , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Progesterone/antagonists & inhibitors , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Breast Neoplasms , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Female , Gonanes/pharmacology , Humans , Mifepristone/analogs & derivatives , Mifepristone/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Phosphates/metabolism , Promegestone/pharmacology , Receptors, Progesterone/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
20.
Trends Endocrinol Metab ; 4(1): 1-7, 1993.
Article in English | MEDLINE | ID: mdl-18407126

ABSTRACT

All steroid receptors are phosphoproteins and several, including progesterone receptors (PRs), become hyperphosphorylated upon binding of ligand. PR phosphorylation is complex, occurring in different cellular compartments and perhaps requiring multiple serine kinases. A model that is emerging proposes that PR phosphorylation is progressive, occurring in at least a three-stage cascade. However, the functional significance of this phosphorylation cascade remains unclear.

SELECTION OF CITATIONS
SEARCH DETAIL
...