Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 18.
Published in final edited form as: Steroids. 2008 Dec 11;74(7):573–576. doi: 10.1016/j.steroids.2008.12.001

Integrated actions of PR and cell cycle machinery regulate breast cancer cell proliferation

Gwen E Dressing 1, Carol A Lange 1,*
PMCID: PMC4871707  NIHMSID: NIHMS82990  PMID: 19118566

Abstract

Multiple laboratories have investigated PR involvement in breast cancer cell cycle progression. There is now a growing body of evidence demonstrating complex interactions between PR and cell cycle regulatory proteins. Here we review the current literature linking PR to cell cycle control and discuss gaps in the current knowledge. A more complete understanding of the relationships between PR and cell cycle regulatory molecules may reveal additional avenues for prevention and treatment of steroid receptor positive breast cancers.

Keywords: progesterone receptor, cell cycle, cyclin D, p21, p27, cyclin dependent kinase 2


The importance of progesterone and the nuclear progesterone receptor (PR) in the progression of steroid hormone receptor-positive breast cancer has recently been a subject of intense study. Exposure of cultured breast cancer cells to progestin initiates cell cycle progression (Figure 1). However, progesterone subsequently induces growth arrest and insensitivity to further progesterone treatment followed by upregulation of tyrosine kinase (erb B) growth factor receptors, in effect “priming” cells for alternate mitogenic stimuli such as EGF [15]. While the mechanisms guiding progestin/PR induced cell cycle progression are currently under investigation, several lines of evidence indicate that PR signaling is interwoven with cell cycle regulation during normal breast development and in breast cancer models, suggesting that this linkage may be therapeutically targeted in the clinic.

Figure 1.

Figure 1

Summary of PR-B Expressing T47D Breast Cancer Cell Proliferation and Cell Cycle Regulatory Protein Expression Following Progestin Exposure. Unsynchronized PR-B expressing T47D breast cancer cells, which are sensitive to progesterone but are not strongly dependent on estrogen, undergo a burst of proliferation upon exposure to progestin (initial 24 hours) followed by growth arrest at the G1/S boundary (dashed line) [1, 3, 26]. Breast cell exposure to continuous progestin results in a transient increase in cyclin D RNA and protein followed by inhibition [1, 5, 22, 23]. Cyclin E levels initially decrease (within 24 hours) upon exposure to progestin followed by a transient cyclin E upregulation (36–60 hours post progestin exposure) [1, 5]. Breast cancer cyclin A levels decrease after 18 hours progestin treatment and remain undetectable through 60 hours [1]. Cyclin B levels also decrease upon progestin treatment of breast cancer cells 36 hours post treatment [1]. Progestin treatment causes a transient increase in CDK2 activity between 12–42 hours post treatment [1, 36]. The cell cycle inhibitor p21Cip1 protein levels transiently increase over the course of 72 hours progestin treatment [1, 39] while p27Kip1 protein levels increase following 40 hours progestin treatment and remain high while progestin is present [1].

PR direct and indirect interaction with cyclins

Over-expression of cyclins in breast tumors has been positively linked to estrogen receptor (ER) and PR status [69]. Notably, similarities exist between cyclin D and PR knockout mice; both models exhibit identical phenotypes characterized by delayed lobuloaveolar development during pregnancy [10, 11]. In addition, several members of the steroid hormone receptor superfamily of ligand-activated transcription factors have been shown to interact with cyclins including ER [12, 13], androgen receptors [1420] and thyroid receptors [21]. These studies have led researchers to more closely examine progesterone and PR interaction with cell cycle regulatory proteins including cyclins, cyclin dependent kinases (CDKs) and members of the p21Cip1/p27Kip1 family of cell cycle regulators.

Work dating back twenty years up to the present shows that acute exposure of growth arrested immortalized breast cancer cells to progestin results in an increase in cyclin D1 mRNA and S phase entry [1, 5, 22] (Figure 1). The ability of anti-progestins to block increases in cyclin D1 [5] and the failure of cells expressing PR-B with a mutation in the DNA binding region to increase cyclin D levels or enter S phase [22, 23] suggested PR involvement. While multiple labs have shown that progestin/PRs are able to induce cyclin D1 gene expression, the PR-dependent mechanisms (i.e. signaling vs. transcriptional activities) are not as clear. Progestin-induced cyclin D1 protein, but not mRNA expression, was dependent upon the sustained activity of c-Src, EGFR and MAPKs, which presumably act to stabilize the short half-life protein through phosphorylation events [24] and/or increase the translation of cyclin D1 mRNAs [25]. Cells expressing PR-B mutants which can bind DNA but cannot rapidly and transiently activate c-Src, showed increased cyclin D and cell cycle progression upon progestin exposure [22, 23]. In similar studies, Faivre, et al. (2007) showed that breast cancer cell proliferation in soft agar required PR DNA binding but was attenuated upon loss of PR rapid signaling events, suggesting that PR-induced protein kinase signaling may promote cell survival. Together, these results demonstrate that the ability of progestin to regulate cyclin D mediated cell cycle progression clearly requires the transcriptional activity of PR-B, yet the precise role and kinetics of extra-nuclear signaling pathways like c-Src and MAPK remain to be defined.

Reciprocal regulation of PR and CDK2

While progestins cause acute upregulation of cyclin D and S phase entry via PR transcriptional activity, chronic exposure of cultured breast cancer cells to progestin results in cell cycle arrest [1, 26] (Figure 1). Long term exposure of breast cancer cell lines to progestin resulted in the inhibition of cyclin D1/CDK4, cyclin D3/CDK4 and cyclin E/CDK2 complex kinase activity as well as decreased overall cyclin D1, cyclin D3 and cyclin E expression [27] (Figure 1). Continuous exposure of T47D cells to progestin also resulted in increased CDK inhibitors p21Cip1 and p27Kip1 proteins [1] and increased association of cyclin E/ CDK2 complexes with kinase inhibiting p27Kip1 [27]. The addition of exogenous cyclin D1 to progestin inhibited cells resulted in the resumption of cell cycle progression and the return of CDK2 activity [27] suggesting that one function of cyclin D1 over-expression is to provide a “sink” for p27Kip1, thereby removing it from cyclin E/ CDK2 complexes. Interestingly, cells which continually over-express cyclin D1 were not responsive to long term progestin inhibition of cell proliferation, yet showed increases in the proportion of cyclin E/CDK2 complexes associated with p27Kip1. These data suggest that cyclin D plays an additional role in progestin regulated cell cycle progression other than simply acting as a p27Kip1 sink [28]. Notably, cyclin E/CDK2 complex kinase activity is hindered by increased p27Kip1, in contrast to cyclin D/ CDK4 complexes which are sensitive to the CDK/4/6 inhibitor p18INK4. T47D cell exposure to progestin increased p18INK4 leading to the upstream inhibition of cyclin E/CDK2 activity and inhibition of cell cycle progression [26]. Thus, chronic exposure of epithelial derived breast cancer cells to progestin results in the upregulation of multiple CDK inhibitors which may act to initially nucleate cyclin/CDK complexes [29], but ultimately decrease the activity of cyclin E/CDK2 and block cell cycle progression (Figure 1). Overexpression of cyclin D, E, or A molecules or loss of p21Cip1 or p27Kip1 in PR positive breast cancer is predicted to bypass these controls [27].

PR, like other steroid receptors, is phosphorylated at multiple sites under varying conditions [30, 31]. Of the fourteen confirmed PR-B phosphorylation sites, 8 are cyclin/CDK sites [32]. Differential phosphorylation of PR, and closely related glucocorticoid receptor altered transcriptional activity and is a mechanism for promoter selectivity [3335]. Given that PRs are CDK2 substrates, it is not surprising that PR has been shown to interact with cyclin E/CDK2 and cyclin A/CDK2 complexes [36, 37]. Progestins increase CDK2 kinase activity and PR phosphorylation at serine 400, a CDK2 site that is also basally phosphorylated, and sensitive to mitogens [36]. Forced expression of a constitutively active kinase mutant of CDK2 in breast cancer cells increased both liganded and unliganded PR transcriptional activity on luciferase reporter genes [36]; ser400 phosphorylation via CDK2 kinase activity was required for rapid PR nuclear localization and ligand –independent PR activity [36]. Nonetheless, the functional significance of PR/cyclin/CDK interaction is not fully understood. PR co-immunoprecipitated with cyclin A in the presence and absence of ligand [37]. Overexpression of cyclin A increased ligand dependent PR activity on a MMTV-luc reporter construct and this required CDK2 activity. However, CDK2 did not appear to directly target known PR phosphorylation sites. While cyclin A physically associated with PR in the absence of CDK2, CDK2 activity was required for transcriptional co-activation of PR. Further investigation showed that the association of PR with the cyclin A/ CDK2 complex resulted in the phosphorylation of SRC1 and its increased association with PR, thus enhancing PR transcriptional activity during S phase [37].

PR regulation of cell cycle inhibitors

In addition to positive regulators of cell cycle progression like the cyclins and CDKs, progestins also directly regulate p21Cip1 and p27Kip1 inhibitors of cell cycle progression. Prolonged progestin exposure resulted first in increased expression of p21Cip1 followed by increased p27Kip1 [1]. PR induced upregulation of cell cycle inhibitors is presumed to contribute to the progestin induced cell growth inhibition of cultured breast cancer cells (Figure 1). While increases in p21Cip1 mRNA and protein levels have been detected there are no hormone response elements in the proximal promoters of this gene. However, recent studies demonstrated that PR, when phosphorylated at Ser345, strongly associates with specificity protein 1 (SP1) transcription factors [38] to upregulate transcription of SP1-regulated genes, including p21Cip1 [39] and EGFR [38]. The mechanism of progestin regulation of p27Kip1 has not yet been resolved.

PR strongly associates with S phase cyclins and CDK2, and these associations promote PR transcriptional activity. Thus, PR activity levels change with cell cycle progression and appear highest during S phase [40] when whole cell transcriptional activity is also high. Interestingly, both liganded and unliganded PR activity on MMTV-luciferase reporter constructs are increased during S phase of synchronized cells, and the patterns of PR phosphorylation and SRC-1 recruitment to PRE change during cell cycle progression. Taken together, these studies indicate that CDK2 dependent phosphorylation of PR (Ser400) and differential recruitment of coactivators (SRC-1) may preferentially regulate PR nuclear localization [36] as well as receptor-coactivator interactions at selected promoters during S phase (i.e. when CDK2 activity is highest) relative to G2/M or G1 [40].

Conclusions

Coordination of PR transcriptional activity, phosphorylation state and association with specific cyclin/CDK complexes suggests that PR regulates genes in a cell cycle phase-dependent manner. As most of the current studies looking at PR target genes have used unsynchronized cells, it is possible that key PR target genes have been missed. In particular, a subset of S phase genes may be regulated by phosphorylated PR in the absence of progesterone. The discovery of these genes may provide clues to how progestin/PR may contribute to development and/or progression of breast cancer. A more complete understanding of the coupling of PR target gene regulation to cell cycle control may reveal additional avenues for prevention and treatment of steroid receptor positive breast cancers.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Literature Cited

  • [1].Groshong SD, Owen GI, Grimison B, Schauer IE, Todd MC, Langan TA, Sclafani RA, Lange CA, Horwitz KB. Biphasic regulation of breast cancer cell growth by progesterone: role of the cyclin-dependent kinase inhibitors, p21 and p27(Kip1) Molecular Endocrinology. 1997;11:1593–607. doi: 10.1210/mend.11.11.0006. [DOI] [PubMed] [Google Scholar]
  • [2].Lange CA. Making sense of cross-talk between steroid hormone receptors and intracellular signaling pathways: who will have the last word? Molecular Endocrinology. 2004;18:269–78. doi: 10.1210/me.2003-0331. [DOI] [PubMed] [Google Scholar]
  • [3].Lange CA, Richer JK, Horwitz KB. Hypothesis: Progesterone primes breast cancer cells for cross-talk with proliferative or antiproliferative signals. Molecular Endocrinology. 1999;13:829–36. doi: 10.1210/mend.13.6.0290. [DOI] [PubMed] [Google Scholar]
  • [4].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. Journal of Biological Chemistry. 1998;273:31308–16. doi: 10.1074/jbc.273.47.31308. [DOI] [PubMed] [Google Scholar]
  • [5].Musgrove EA, Hamilton JA, Lee CSL, Sweeney KJE, Watts CKW, Sutherland RL. Growth factor, steroid, and steroid antagonist regulation of cyclin gene expression associated with changes in T-47D human breast cancer cell cycle progression. Molecular and Cellular Biology. 1993;13:3577–3587. doi: 10.1128/mcb.13.6.3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Reis-Fihlo JS, Savage K, Lambros M, James M, Steele D, Jones R, Dowsett M. Cyclin D1 protein overexpression and CCND1 amplification in breast carcinomas: an immunohistochemical and chromogenic in situ hybridisation analysis. Modern Pathology. 2006;19:999–1009. doi: 10.1038/modpathol.3800621. [DOI] [PubMed] [Google Scholar]
  • [7].Millar E, Tran K, Marr P, Graham PH. p27KIP-1 cyclin A and cyclin D1 protein expression in ductal carcinoma in situ of the breast: p27KIP-1 correlates with hormone receptor status but not with local recurrence. Pathology International. 2007;57:183–189. doi: 10.1111/j.1440-1827.2007.02079.x. [DOI] [PubMed] [Google Scholar]
  • [8].Hui R, Cornish AL, McClelland RA, Roberson JF, Blamey RW, Musgrove EA, Nicholson RI, Sutherland RL. Cyclin D1 and estrogen receptor messenger RNA levels are positively correlated in primary breast cancer. Clinical Cancer Research. 1996;2:923–8. [PubMed] [Google Scholar]
  • [9].Al-Kuraya K, Schraml P, Torhorst J, Tapia C, Zaharieva B, Novotny H, Spichtin H, Maurer R, Mirlacher M, Kochili O, Zuber M, Dieterich H, Mross F, Wilber K, Simon R, Sauter G. Prognostic relevance of gene amplifications and coamplifications in breast cancer. Cancer Research. 2004;64:8534–40. doi: 10.1158/0008-5472.CAN-04-1945. [DOI] [PubMed] [Google Scholar]
  • [10].Sicinski P, Donaher JP, Parker SB, Li T, Fazeli A, Gardner H, Haslam SZ, Bronson RT, Elledge SJ, Weinberg RA. Cyclin D1 provides a link between development and oncogenesis in the retina and breast. Cell. 1995;82:621–30. doi: 10.1016/0092-8674(95)90034-9. [DOI] [PubMed] [Google Scholar]
  • [11].Mulac-Jericevic B, Lydon JP, DeMayo FJ, Conneely OM. Defective mammary gland morphogeneis in mice lacking the progesterone receptor B isoform. Proceeding of the National Academy of Sciences. 2003;100:9744–9. doi: 10.1073/pnas.1732707100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Zwijzen RM, Buckle RS, Hijmans EM, Loomans CJ, Bernards R. Ligand-independent recruitment of steroid receptor coactivators to estrogen receptor by cyclin D1. Genes and Development. 1998;12:3488–98. doi: 10.1101/gad.12.22.3488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].McMahon C, Suthiphongchai T, DiRenzo J, Ewen ME. P/CAF associates with cyclin D1 and potentiates its activation of the estrogen receptor. Proceeding of the National Academy of Sciences. 1999;96:5382–7. doi: 10.1073/pnas.96.10.5382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Knudsen KE, Cavenee WK, Arden KC. D-type cyclins complex with the androgen receptor and inhibit its transcriptional transactivation ability. Cancer Research. 1999;59:2297–301. [PubMed] [Google Scholar]
  • [15].Reutens AT, Fu M, Wang C, Albanese C, McPhaul MJ, Sun Z, Balk SP, Janne O, Palvimo JJ, Pestell RG. Cyclin D1 binds the androgen receptor and regulates hormone-dependent signaling in a p300/CBP-associated factor (P/CAF)-dependent manner. Molecular Endocrinology. 2001;15:797–811. doi: 10.1210/mend.15.5.0641. [DOI] [PubMed] [Google Scholar]
  • [16].Petre CE, Wetherill YB, Danielsen M, Knudsen KE. Cyclin D1: mechanism and consequence of androgen receptor co-repressor activity. Journal of Biological Chemistry. 2002;277:2207–15. doi: 10.1074/jbc.M106399200. [DOI] [PubMed] [Google Scholar]
  • [17].Petre-Draviam CE, Cook SL, Burd CJ, Marshall TW, Wetherill YB, Knudsen KE. Specificity of cyclin D1 for androgen receptor regulation. Cancer Research. 2003;63:4903–13. [PubMed] [Google Scholar]
  • [18].Burd CJ, Petre CE, Moghadam H, Wilson EM, Knudsen KE. Cyclin D1 binding to the androgen receptor (AR) NH2-terminal domain inhibits activation of fuction 2 association and reveals dual roles for AR corepression. Molecular Endocrinology. 2005;19:607–20. doi: 10.1210/me.2004-0266. [DOI] [PubMed] [Google Scholar]
  • [19].Petre-Draviam CE, Williams EB, Burd CJ, Gladden A, Moghadam H, Meller J, Diehl JA, Knudsen KE. A central domain of cyclin D1 mediates nuclear receptor corepressor activity. Oncogene. 2005;24:431–44. doi: 10.1038/sj.onc.1208200. [DOI] [PubMed] [Google Scholar]
  • [20].Burd CJ, Petre CE, Morney LM, Wang YL, Revelo MP, Haiman CA, Lu SS, Fenoglio-Preiser CM, Li J, Knudsen ES, Wong J, Knudsen KE. Cyclin D1b variant influences prostate cancer growth through aberrant androgen receptor regulation. Proceeding of the National Academy of Sciences. 2006;103:2190–5. doi: 10.1073/pnas.0506281103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Lin HM, Zhao L, Cheng SY. Cyclin D1 is a ligand-independent co-repressor for thyroid hormone receptors. Journal of Biological Chemistry. 2002;277:28733–41. doi: 10.1074/jbc.M203380200. [DOI] [PubMed] [Google Scholar]
  • [22].Faivre E, Lange CA. Progesterone receptors upregulate Wnt-1 to induce epidermal growth factor receptor transactivation and c-Src-dependent sustained activation of Erk1/2 mitogen-activated protein kinase in breast cancer cells. Molecular and Cellular Biology. 2007;27:466–480. doi: 10.1128/MCB.01539-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].McGowan EM, Russell AJ, Boonyaratanakornkit V, Saunders DN, Lehrbach GM, Sergio CM, Musgrove EA, Edwards DP, Sutherland RL. Progestins reinitiate cell cycle progression in antiestrogen-arrested breast cancer cells through the B-isoform of progesterone receptor. Cancer Research. 2007;67:8942–51. doi: 10.1158/0008-5472.CAN-07-1255. [DOI] [PubMed] [Google Scholar]
  • [24].Guo Y, Stacey DW, Hitomi M. Post-transcriptional regulation of cyclin D1 expression during G2 phase. Oncogene. 2002;21:7545–56. doi: 10.1038/sj.onc.1205907. [DOI] [PubMed] [Google Scholar]
  • [25].Rosenwald IB, Kaspar R, Rousseau D, Gehrke L, Leboulch P, Chen JJ, Schmidt EV, Sonenberg N, London IM. Eukaryotic translation initiation factor 4E regulates expression of cyclin D1 at transcriptional and post-transcriptional levels. J Biol Chem. 1995;270:21176–80. doi: 10.1074/jbc.270.36.21176. [DOI] [PubMed] [Google Scholar]
  • [26].Swarbrick A, Lee CSL, Sutherland RL, Musgrove EA. Cooperation of p27/kip and p18/INK4c in progestin-mediated cell cycle arrest in T-47D breast cancer cells. Molecular and Cellular Biology. 2000;20:2581–91. doi: 10.1128/mcb.20.7.2581-2591.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Musgrove EA, Swarbrick A, Lee CSL, Cornish AL, Sutherland RL. Mechanisms of cyclin-dependent kinase inactivation by progestins. Molecular and Cellular Biology. 1998;18:1812–25. doi: 10.1128/mcb.18.4.1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Musgrove EA, Hunter LK, Lee CSL, Swarbrick A, Hui R, Sutherland RL. Cyclin D1 overexpression induces progestin resistance in T-47D breast cancer cells despite p27Kip1 association with cyclin E-Cdk2. Journal of Biological Chemistry. 2001;276:47675–83. doi: 10.1074/jbc.M106371200. [DOI] [PubMed] [Google Scholar]
  • [29].LaBaer J, Garrett MD, Stevenson LF, Slingerland JM, Sandhu C, Chou HS, Fattaey A, Harlow E. New functional activities for the p21 family of CDK inhibitors. Genes Dev. 1997;11:847–62. doi: 10.1101/gad.11.7.847. [DOI] [PubMed] [Google Scholar]
  • [30].Weigel NL. Steroid hormone receptors and their regulation by phosphorylation. Biochemical Journal. 1996;319:657–67. doi: 10.1042/bj3190657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Weigel NL, Bai W, Zhang Y, Beck CA, Edwards DP, Poletti A. Phosphorylation and progesterone receptor function. Journal of Steroid Biochemistry and Molecular Biology. 1995;53:509–14. doi: 10.1016/0960-0760(95)00098-k. [DOI] [PubMed] [Google Scholar]
  • [32].Moore NL, Narayanan R, Weigel NL. Cyclin dependent kinase 2 and the regulation of human progesterone receptor activity. Steroids. 2007;72:202–9. doi: 10.1016/j.steroids.2006.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Galliher-Beckley AJ, Williams JG, Collins JB, Cidlowski JA. GSK-3{beta}-mediated Serine Phosphorylation of the Human Glucocorticoid Receptor Re-directs Gene Expression Profiles. Mol Cell Biol. 2008 doi: 10.1128/MCB.00808-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Chen W, Dang T, Blind RD, Wang Z, Cavasotto CN, Hittelman AB, Rogatsky I, Logan SK, Garabedian MJ. Glucocorticoid receptor phosphorylation differentially affects target gene expression. Mol Endocrinol. 2008;22:1754–66. doi: 10.1210/me.2007-0219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Daniel A, Qiu M, Faivre E, Ostrander J, Skildum A, Lange C. Linkage of progestin and epidermal growth factor signaling: phosphorylation of progesterone receptors mediates transcriptional hypersensitivity and increased ligand-independent breast cancer cell growth. Steroids. 2007;72:188–201. doi: 10.1016/j.steroids.2006.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].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. Molecular and Cellular Biology. 2004;24:10542–57. doi: 10.1128/MCB.24.24.10542-10557.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].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. Molecular and Cellular Biology. 2005;25:264–77. doi: 10.1128/MCB.25.1.264-277.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Faivre EJ, Daniel AR, Hillard CJ, Lange CA. Progesterone Receptor Rapid Signaling Mediates Ser345 Phosphorylation and Tethering to Sp1 Transcription Factors. Molecular Endocrinology. 2008 doi: 10.1210/me.2007-0437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Owen GI, Richer JK, Tung L, Takimoto G, Horwitz KB. Progesterone regulates transcription of the p21(WAF1) cyclin- dependent kinase inhibitor gene through Sp1 and CBP/p300. J Biol Chem. 1998;273:10696–701. doi: 10.1074/jbc.273.17.10696. [DOI] [PubMed] [Google Scholar]
  • [40].Narayanan R, Edwards DP, Weigel NL. Human progesterone receptor displays cell cycle-dependent changes in transcriptional activity. Molecular and Cellular Biochemistry. 2005;25:2885–98. doi: 10.1128/MCB.25.8.2885-2898.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES