Hostname: page-component-76fb5796d-5g6vh Total loading time: 0 Render date: 2024-04-28T00:25:39.068Z Has data issue: false hasContentIssue false

Review of: Human progesterone receptor displays cell cycle-dependent changes in transcriptional activity

Published online by Cambridge University Press:  03 January 2006

C. A. Lange
Affiliation:
Departments of Medicine and Pharmacology, University of Minnesota Cancer Center, Minneapolis, MN, USA.

Abstract

Citation of original article:

R. Narayanan, D. P. Edwards, N. L. Weigel. Human progesterone receptor displays cell cycle-dependent changes in transcriptional activity. Molecular and Cellular Biology 2005; 25(8): 2885–2898.

Abstract of the original article

The human progesterone receptor (PR) contains multiple Ser-Pro phosphorylation sites that are potential substrates for cyclin-dependent kinases, suggesting that PR activity might be regulated during the cell cycle. Using T47D breast cancer cells stably transfected with an mouse mammary tumor virus (MMTV) chloramphenicol acetyltransferase reporter (Cat0) synchronized in different phases of the cell cycle, we found that PR function and phosphorylation is remarkably cell cycle dependent, with the highest activity in S phase. Although PR expression was reduced in the G2/M phase, the activity per molecule of receptor was markedly reduced in both G1 and G2/M phases compared to the results seen with the S phase of the cell cycle. Although PR is recruited to the MMTV promoter equivalently in the G1 and S phases, recruitment of SRC-1, SRC-3, and, consequently, CBP is reduced in G1 phase despite comparable expression levels of SRC-1 and SRC-3. In G2/M phase, site-specific phosphorylation of PR at Ser162 and at Ser294, a site previously reported to be critical for transcriptional activity and receptor turnover, was abolished. Treatment with the histone deacetylase inhibitor trichostatin A elevated G1 and G2/M activity to that of the S phase, indicating that the failure to recruit sufficient levels of active histone acetyltransferase is the primary defect in PR-mediated transactivation.

Type
Journals Club
Copyright
2006 Cambridge University Press

Review

Progesterone receptors (PR) are ligand-activated transcription factors that are also capable of rapidly activating multiple intracellular signaling pathways initiated at or near the plasma membrane. PR exist as either 94 kDa A or 120 kDa B isoforms created by the same gene and mRNAs by the use of alternate promoters and unique translational start sites. The A isoform is an amino-terminally truncated version of the longer B isoform, each containing a C-terminal hormone-binding domain (HBD) and DNA-binding domain (DBD), a hinge region (H), and transcriptional activation function (AF) domains located within both the HBD (AF-1) and N-terminni (AF-2 in PR-A and PR-B; AF-3 in PR-B only). Like other steroid hormone receptor family members (androgen, glucocorticoid, estrogen, and mineralocorticoid receptors), PR are heavily phosphorylated by multiple protein kinases, primarily at N-terminal serine residues. Although the role of PR phosphorylation (i.e. in humans) is not fully understood, it has been shown to influence many aspects of PR transcriptional regulation, including promoter specificity [1], coactivator interaction [2], ligand-dependent [2,3] and independent [4,5] transcriptional activities, receptor turnover [6], and nuclear association [7].

In addition to progestin binding to PR, growth factors independently induce PR phosphorylation at specific sites and by the same or separate kinase pathways [7]. Cross-talk between PR and mitogenic growth factors occurs at multiple levels, including progestin upregulation of epidermal growth factor receptors (EGFRs) and their ligands [8,9]. In the normal breast, EGF potentiates the proliferative actions of progesterone and estrogen, and causes ductal side branching and lobuloalveolar development of the mature mammary gland [10,11]. In breast cancer cells, EGF and progestins synergistically upregulate cyclin D1 and cyclin E protein levels [12]. Cyclins, in turn, regulate progression of cells through the cell cycle by interaction with cyclin-dependent protein kinases (CDKs). For example, D-type cyclins are expressed throughout the cell cycle in response to mitogenic stimulation, while the expression of cyclins E, A, and B (the mitotic cyclin) is periodic. Cyclin D isoforms form complexes with CDK4 and CDK6, and cyclin E associates with CDK2 in S phase, as does cyclin A. Cyclin A also forms complexes with CDK1 in late S and G2, while cyclin B/CDK1 complexes are restricted to M phase [13]. Notably, 8 of 14 phosphorylated Ser residues in PR are CDK2 sites [1417]. Thus, PR phosphorylation is predicted to be induced primarily by cyclinE/CDK2 (G1 to S transition) and/or cyclinA/CDK2 (early S phase) complexes. In addition, progestins are excellent natural synchronizers of the cell cycle, and induce increased CDK2 levels and activity, cyclin D1 expression, and precisely timed S-phase entry that is followed by cell growth inhibition at the G1/S boundary [18]. CDK2 activity is known to increase PR transcriptional activity, both in the presence and absence of progestins [2,4].

Phosphorylation of PR by activated CDK2 suggests a mechanism for the coordinate regulation of PR action during cell cycle progression. This hypothesis was recently directly tested in a paper appearing in the April 2005 issue of Molecular and Cellular Biology by R. Narayanan, D. P. Edwards, and N. L. Weigel entitled, ‘Human progesterone receptor displays cell cycle-dependent changes in transcriptional activity.’ The authors cleverly used three different treatment protocols (designed for capturing cells specifically in either G1, S, or G2/M phases) to create synchronized populations of T47D cells stably expressing an MMTV promoter-driven chloramphenicol acetyltransferase (CAT) reporter gene [19]. Cells enriched for each phase of the cell cycle were then stimulated with progestin (6 h) and CAT activity was measured in cell lysates to determine when PR are most active. In their first set of experiments, Narayanan et al. [19] showed that PR transcriptional activity is highest in S phase relative to G1 and G2/M phases, in which PR are expressed but exist in a state of repressed activity.

The phospho-status of PR in each phase was also examined using available antibodies for selected sites. Notably, Ser162 and Ser294 were hormonally regulated and robustly phosphorylated in both G1 and S phase, but not in G2/M phase. The authors suggest that in G2/M phase, PR is either in an altered conformation or associated with other protein(s) that occlude these sites. An alternative interpretation of these data is that Ser294 phosphorylated PR species are active, but also rapidly degraded [3,6], perhaps preferentially in G2/M phase. This would explain low levels of phospho-Ser294 PR relative to total. It is also possible that cytosolic phosphatases (active in G2/M) mediate the ‘net’ dephosphorylation of ‘shuttling’ PRs, which the authors found to be primarily nuclear during S phase. Regardless of mechanism, a clear correlation exists between PR activity and PR phosphorylation (at Ser162 and Ser294). However, in a previous publication appearing in the same journal [2], these investigators showed that the CDK2-induced increase in PR transcriptional activity (i.e. in the presence of progestin) did not map to phosphorylation sites within PR, but instead stimulated the association of phospho-SRC-1 to PR complexes present at the MMTV promoter. They also found that cyclin A and CDK2 associated with PR in active transcription complexes. In the more recent MCB paper reviewed herein [19], the authors used CHIP assays to show that in addition to steroid receptor coactivator 1 (SRC-1) [2], both SRC-3 and CREB-binding protein (CBP) were preferentially recruited to the MMTV promoter during S phase relative to G1 or G2/M phases.

Finally, the authors showed that PR transcriptional activity in both G1 and G2/M could be restored to the S phase levels by addition of the histone deacetyltransferase (HDAC) inhibitor, trichostatin A (TSA). However, TSA-treatment did not fully restore the ability of progestin to induce PR Ser294 phosphorylation. The authors interpret these data as suggestive that phosphorylation of PR Ser294 is not involved in the mechanism of PR transcriptional activation. An alternative explanation is that histone acetylation and chromatin remodeling (i.e. unwinding) are steps that occur well after PR phosphorylation, which may act to induce persistent nuclear accumulation of PRs during S phase and early co-factor recruitment (i.e. other than SRC isoforms). That is, PR Ser294 phosphorylation may not be required after complexes with sufficient histone acetyltransferase (HAT) activity are appropriately formed.

Perhaps an under-appreciated, but exciting aspect of the Narayanan paper involves the question of hormone sensitivity and cell cycle progression. Interestingly, total levels of PR transcriptional activity in both the G1 and G2/M phases of the cell cycle were highly progestin sensitive, but remained low relative to S phase totals. In contrast, PR transcriptional activity in S phase was heightened, but much less responsive to added hormone (i.e. the basal activity was also very high). As this activity was clearly blocked by the addition of RU486, the authors attribute this PR-dependent activity to residual ligand present after the washout of required serum during the cell cycle synchronization step for S phase enrichment. Yet, both the G1 and G2/M phase protocols required the same washout of added serum, while PR activities remained quite low in the absence of freshly added ligand. Although not addressed by Narayanan and co-workers, these studies have revealed a window (i.e. S phase) of PR ‘hypersensitivity’ in which phosphorylated nuclear receptors may be well-activated by sub-physiologic levels of hormone. Phosphorylation events that induce the recruitment of steroid receptor coactivators (SRC-1, SRC-3, and/or CBP) are predicted to shift the dose–response curve for receptor activation far to the left, lowering the apparent EC50 for transcriptional activation [20]. This suggests that when cells are in S phase, much lower concentrations of PR ligand (i.e. agonists) are sufficient to achieve regulation of gene expression at selected promoters.

What are the implications of these studies for steroid hormone action relevant to breast cancer biology? Breast cancers notoriously display alterations in cell cycle regulation, including increased expression of cyclins D, E and A, loss of p27, and heightened CDK2 activity [2126]. Clearly, increased proliferation as measured by several markers, including cyclin A, predicts a poor prognosis [27]. High cyclin A levels are correlated with proliferation and strongly predict a shorter time to first relapse and shortened survival from diagnosis [28]. Thus, the key question becomes, is there a role for S phase PR action in breast cancer progression? Clinical data suggest that the addition of a progestin to hormone replacement therapy increases breast cancer risk [29]. Tumors that developed in women taking estrogen plus progesterone were larger and of higher-grade relative to estrogen alone or placebo [29], suggesting that progestins actually stimulated breast cancer progression. The paper reviewed herein [19] demonstrated that in PR-positive breast cancer cells, PR activity is highest in S phase, and may be particularly ultrasensitive to low hormone levels. CDK2 can also drive ligand-independent PR activity when p27 levels are low (as in S phase) or knocked-down [4].

What are the relevant PR target genes? Little information exists on the role of PR target genes in breast cancer cell growth control or metastasis. However, nearly half of all PR regulated genes, identified in breast cancer cells, using gene-array approaches encode cell adhesion and membrane-bound proteins or proteins involved in membrane-initiated signaling [3032]. PR is known to upregulate c-myc, STAT5A, cyclin D1, TGF-beta, and EGFR mRNAs, and progestins synergize with EGF to induce increased expression of c-myc, c-fos, p21 and cyclins D1 and E [12,3033]. Clearly much work remains to be done. However, in light of these findings and the new studies reviewed herein [19], inclusion of antiprogestins to existing antiestrogen and combination therapies (that block kinases) should be seriously considered, as blocking PR action in S phase may retard tumor progression and thereby prevent or delay breast cancer recurrence.

References

Qiu M, Lange CA. MAP kinases couple multiple functions of human progesterone receptors: degradation, transcriptional synergy, and nuclear association. J Steroid Biochem Mol Biol 2003; 85: 147157.Google Scholar
Narayanan R, Adigun AA, Edwards DP, Weigel NL. Cyclin-dependent kinase activity is required for progesterone receptor function: novel role for cyclin A/Cdk2 as a progesterone receptor coactivator. Mol Cell Biol 2005; 25: 264277.Google Scholar
Shen T, Horwitz KB, Lange CA. Transcriptional hyperactivity of human progesterone receptors is coupled to their ligand-dependent down-regulation by mitogen-activated protein kinase-dependent phosphorylation of serine 294. Mol Cell Biol 2001; 21: 61226131.Google Scholar
Pierson-Mullany LK, Lange CA. Phosphorylation of progesterone receptor serine 400 mediates ligand-independent transcriptional activity in response to activation of cyclin-dependent protein kinase 2. Mol Cell Biol 2004; 24: 10 54210 557.Google Scholar
Labriola L, Salatino M, Proietti CJ, et al. Heregulin induces transcriptional activation of the progesterone receptor by a mechanism that requires functional ErbB-2 and mitogen-activated protein kinase activation in breast cancer cells. Mol Cell Biol 2003; 23: 10951111.Google Scholar
Lange CA, Shen T, Horwitz KB. Phosphorylation of human progesterone receptors at serine-294 by mitogen-activated protein kinase signals their degradation by the 26S proteasome. Proc Natl Acad Sci USA 2000; 97: 10321037.Google Scholar
Qiu M, Olsen A, Faivre E, Horwitz KB, Lange CA. Mitogen activated protein kinase regulates nuclear association of human progesterone receptors. Mol Endocrinol 2003; 17: 628642.Google Scholar
Lange CA, Richer JK, Horwitz KB. Hypothesis: progesterone primes breast cancer cells for cross-talk with proliferative or antiproliferative signals. Mol Endocrinol 1999; 13: 829836.Google Scholar
Lange CA. Making sense of cross-talk between steroid hormone receptors and intracellular signaling pathways: Who will have the last word? Mol Endocrinol 2004; 18: 269278.Google Scholar
Haslam SZ, Shyamala G. Relative distribution of estrogen and progesterone receptors among the epithelial, adipose, and connective tissue components of the normal mammary gland. Endocrinology 1981; 108: 825830.Google Scholar
Haslam SZ, Counterman LJ, Nummy KA. Effects of epidermal growth factor, estrogen, and progestin on DNA synthesis in mammary cells in vivo are determined by the developmental state of the gland. J Cell Physiol 1993; 155: 7278.Google Scholar
Lange CA, Richer JK, Shen T, Horwitz KB. Convergence of progesterone and epidermal growth factor signaling in breast cancer. Potentiation of mitogen-activated protein kinase pathways. J Biol Chem 1998; 273: 31 30831 316.Google Scholar
Sherr CJ. Cancer cell cycles. Science 1996; 274: 16721677.Google Scholar
Zhang Y, Beck CA, Poletti A, et al. Phosphorylation of human progesterone receptor by cyclin-dependent kinase 2 on three sites that are authentic basal phosphorylation sites in vivo. Mol Endocrinol 1997; 11: 823832.Google Scholar
Zhang Y, Beck CA, Poletti A, Edwards DP, Weigel NL. Identification of phosphorylation sites unique to the B form of human progesterone receptor. In vitro phosphorylation by casein kinase II. J Biol Chem 1994; 269: 31 03431 040.Google Scholar
Zhang Y, Beck CA, Poletti A, Edwards DP, Weigel NL. Identification of a group of Ser-Pro motif hormone-inducible phosphorylation sites in the human progesterone receptor. Mol Endocrinol 1995; 9: 10291040.Google Scholar
Knotts TA, Orkiszewski RS, Cook RG, et al. Identification of a phosphorylation site in the hinge region of the human progesterone receptor and additional amino-terminal phosphorylation sites. J Biol Chem 2001; 276: 84758483.Google Scholar
Groshong SD, Owen GI, Grimison B, et al. Biphasic regulation of breast cancer cell growth by progesterone: role of the cyclin-dependent kinase inhibitors, p21 and p27(Kip1). Mol Endocrinol 1997; 11: 15931607.Google Scholar
Narayanan R, Edwards DP, Weigel NL. Human progesterone receptor displays cell cycle-dependent changes in transcriptional activity. Mol Cell Biol 2005; 25: 28852998.Google Scholar
Simons Jr SS, Oshima H, Szapary D. Higher levels of control: modulation of steroid hormone-regulated gene transcription. Mol Endocrinol 1992; 6: 9951002.Google Scholar
Alkarain A, Jordan R, Slingerland J. p27 deregulation in breast cancer: prognostic significance and implications for therapy. J Mammary Gland Biol Neoplasia 2004; 9: 6780.Google Scholar
Akli S, Keyomarsi K. Cyclin E and its low molecular weight forms in human cancer and as targets for cancer therapy. Cancer Biol Ther 2003; 2: S38S47.Google Scholar
Sutherland RL, Prall OWJ, Watts CKW, Musgrove EA. Estrogen and progestin regulation of cell cycle progression. J Mammary Gland Biol Neoplasia 1998; 3: 6372.Google Scholar
Porter PL, Malone KE, Heagerty PJ, et al. Expression of cell-cycle regulators p27Kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients. Nat Med 1997; 3: 222225.Google Scholar
Gillett C, Fantl V, Smith R, et al. Amplification and overexpression of cyclin D1 in breast cancer detected by immunohistochemical staining. Cancer Res 1994; 54: 18121817.Google Scholar
Keyomarsi K, O'Leary N, Molnar G, et al. Cyclin E, a potential prognostic marker for breast cancer. Cancer Res 1994; 54: 380385.Google Scholar
van Diest PJ, van der Wall E, Baak JP. Prognostic value of proliferation in invasive breast cancer: a review. J Clin Pathol 2004; 57: 675681.Google Scholar
Poikonen P, Sjostrom J, Amini RMV, et al. Cyclin A as a marker for prognosis and chemotherapy response in advanced breast cancer. Br J Cancer 2005; August 9 epub ahead of print 2005; 93: 515519.Google Scholar
Chlebowski RT, Hendrix SL, Langer RD, et al. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the women's health initiative randomized trial. J Am Med Assoc 2003; 289: 32433253.Google Scholar
Richer JK, Jacobsen BM, Manning NG, et al. Differential gene regulation by the two progesterone receptor isoforms in human breast cancer cells. J Biol Chem 2002; 277: 52095218.Google Scholar
Jacobsen BM, Richer JK, Sartorius CA, Horwitz KB. Expression profiling of human breast cancers and gene regulation by progesterone receptors. J Mammary Gland Biol Neoplasia 2003; 8: 257268.Google Scholar
Jacobsen BM, Richer JK, Schittone SA, Horwitz KB. New human breast cancer cells to study progesterone receptor isoform ratio effects and ligand-independent gene regulation. J Biol Chem 2002; 277: 27 79327 800.Google Scholar
Richer JK, Lange CA, Manning NG, Owen G, Powell R, Horwitz KB. Convergence of progesterone with growth factor and cytokine signaling in breast cancer. Progesterone receptors regulate signal transducers and activators of transcription expression and activity. J Biol Chem 1998; 273: 31 31731 326.Google Scholar