Review
The biology of progesterone receptor in the normal mammary gland and in breast cancer

https://doi.org/10.1016/j.mce.2011.10.030Get rights and content

Abstract

This paper reviews work on progesterone and the progesterone receptor (PR) in the mouse mammary gland that has been used extensively as an experimental model. Studies have led to the concept that progesterone controls proliferation and morphogenesis of the luminal epithelium in a tightly orchestrated manner at distinct stages of development by paracrine signaling pathways, including receptor activator of nuclear factor κB ligand (RANKL) as a major paracrine factor. Progesterone also drives expansion of stem cells by paracrine signals to generate progenitors required for alveologenesis. During mid-to-late pregnancy, progesterone has another role to suppress secretory activation until parturition mediated in part by crosstalk between PR and prolactin/Stat5 signaling to inhibit induction of milk protein gene expression, and by inhibiting tight junction closure. In models of hormone-dependent mouse mammary tumors, the progesterone/PR signaling axis enhances pre-neoplastic progression by a switch from a paracrine to an autocrine mode of proliferation and dysregulation of the RANKL signaling pathway. Limited experiments with normal human breast show that progesterone/PR signaling also stimulates epithelial cell proliferation by a paracrine mechanism; however, the signaling pathways and whether RANKL is a major mediator remains unknown. Work with human breast cancer cell lines, patient tumor samples and clinical studies indicates that progesterone is a risk factor for breast cancer and that alteration in progesterone/PR signaling pathways contributes to early stage human breast cancer progression. However, loss of PR expression in primary tumors is associated with a less differentiated more invasive phenotype and worse prognosis, suggesting that PR may limit later stages of tumor progression.

Highlights

► This paper reviews the role of progesterone (P4) in mammary gland development. ► In the mouse P4 controls luminal epithelial proliferation and stem cell expansion. ► These P4 effects are mediated by a RANKL paracrine signaling pathway. ► Dysregulation of RANKL signaling promotes hormone-dependent mouse mammary tumors. ► Whether RANKL has a role in hormone-dependent human breast cancer is unknown. ► PR has tumor suppressor activity at later stages of breast cancer progression.

Introduction

The mammary gland is a hormonally responsive target tissue that develops predominantly after birth, and is capable of undergoing sequential cycles of development through pregnancy, lactation and involution. Because of common developmental and hormonal response features, the mouse mammary gland has served as an experimental system for modeling normal human breast and breast cancer (Conneely et al., 2003). Progesterone (P4) is a key cycling ovarian steroid hormone that is highest in the luteal phase and has a major role to promote glandular differentiation of the endometrium. P4 is also sustained at high levels during pregnancy and is required for maintenance of pregnancy (Anderson and Clarke, 2004, Graham and Clarke, 1997, Howard and Gusterson, 2000, Silberstein et al., 1996). Experimental manipulation of mouse ovarian function has demonstrated the importance of P4 for proliferation and ductal side branching of the mammary gland during puberty and for alveologenesis during pregnancy (Graham and Clarke, 1997). The essential nature of the P4 signaling axis for mammary gland development has also been defined through studies of the progesterone receptor (PR) knockout mouse (PRKO) (Lydon et al., 1995). PR is a member of the nuclear hormone receptor family of ligand-dependent transcription factors that functions by binding to specific target genes either through cis-acting progesterone response elements (PREs) or by tethering to other DNA bound transcription factors. DNA bound receptor recruits co-regulatory proteins that affect chromatin structure and rates of gene transcription (Bulynko and O’Malley, 2010, Kastner et al., 1990, Li and O’Malley, 2003, Mangelsdorf et al., 1995). In addition to its action as a transcription factor, a subpopulation of PR functions outside of the nucleus to mediate rapid (minutes) P4 induced activation of protein phosphorylation signaling cascades (Ballare et al., 2003, Boonyaratanakornkit et al., 2001, Edwards, 2005). In both humans and mice, PR is expressed as two isoforms, PR-A and PR-B that have identical ligand–binding (LBD) and DNA-binding domains (DBD) and differ only in truncation of the amino-terminal domain (NTD) in PR-A (Aupperlee et al., 2005, Aupperlee and Haslam, 2007, Kastner et al., 1990, Li and O’Malley, 2003, Mangelsdorf et al., 1995, Mulac-Jericevic et al., 2000, Shyamala et al., 1998). In human breast cancer cell lines PR-B is a stronger transcriptional activator than PR-A, and the two isoforms have been characterized to regulate different but overlapping subsets of target genes (Graham et al., 2005, Jacobsen et al., 2003). Also, the proliferative effect of P4 in breast cancer cells is mediated primarily by PR-B (Boonyaratanakornkit et al., 2007, Daniel et al., 2009, Faivre et al., 2008, Jacobsen et al., 2003, Skildum et al., 2005). Based on phenotypes of PR isoform selective knockout mice, PR-B is more important for the proliferative responses to P4 in the mammary epithelium, while ovarian and uterine development and function rely primarily on PR-A (Mulac-Jericevic et al., 2000, Mulac-Jericevic et al., 2003). Distinct functions in the mouse in vivo are due in part to differential expression of PR-A and PR-B during development of the mammary gland and in cell compartments of the uterus (Aupperlee et al., 2005, Aupperlee and Haslam, 2007, Kariagina et al., 2007, Mote et al., 2006). This paper reviews the role of P4 and PR in normal mammary gland development, focusing primarily on the mouse model, and in breast cancer based on studies of mouse and human models.

Section snippets

Epithelial cell autonomous actions of PR in the mammary gland

During post-natal development and puberty the mouse mammary gland ductal cap cells proliferate leading to elongation of the ductal tree into the fat pad (Anderson and Clarke, 2004, Graham and Clarke, 1997, Scarpin et al., 2009). In the PRKO mouse, ductal elongation is similar to that of the wild type mouse, establishing that P4/PR signaling is not required for ductal elongation (Lydon et al., 1995). The phenotpye of estrogen receptor alpha (ERα) knockout mice has established the requirement of

Progesterone regulation of cell proliferation and morphogenesis by paracrine pathways

Based on immunohistochemistry PR is exclusively expressed in the epithelial cell compartment of mammary gland ducts with no evidence of expression in myoepithelial cells or stroma (Grimm et al., 2002, Ismail et al., 2003, Shyamala et al., 1997, Shyamala et al., 2002, Silberstein et al., 1996). PR is uniformly expressed in epithelial cells in juvenile mammary gland ducts but switches to a heterogeneous pattern during puberty and in the adult. In the adult, PR is expressed in ∼40% of cells and PR

Paracrine and other mediators of P4/PR signaling in the mouse mammary gland

Several downstream targets have been implicated as effectors of P4 induced cellular proliferation and morphogenesis including Wnt4, amphiregulin, calcitonin, inhibitor of DNA binding 4 (Id4) and receptor activator of nuclear factor κB ligand (RANKL). These genes are known to be involved in proliferation and/or morphogenesis and are induced by P4 in the mammary gland in a manner dependent on PR (Beleut et al., 2010, Brisken et al., 2000, Fernandez-Valdivia et al., 2008, Gavin and McMahon, 1992,

P4/PR suppression of differentiation during pregnancy

In mid-to-late pregnancy, progesterone inhibits milk protein production and closure of tight junctions until parturition (Neville et al., 2002). Tight junctions of the mammary epithelium remain open during pregnancy and their closure during lactation is important to prevent reflux of accumulated milk into the ductal lumen and interstitial spaces (Neville et al., 2002, Nguyen et al., 2001). In the mouse, a precipitous decline in circulating P4 at parturition is associated with increased milk

Crosstalk between prolactin/Stat5 and progesterone/PR signaling pathways

Both P4 and prolactin (PRL) are required for epithelial cell expansion and alveologenesis. Endocrine ablation and gene knockout studies have shown that neither hormone alone is sufficient, and PRKO and PRL receptor knockout (PRLRKO) mice exhibit a similar phenotype suggesting crosstalk between P4 and PRL signaling pathways (Barash, 2006, Bole-Feysot et al., 1998, Brisken and Rajaram, 2006, Fendrick et al., 1998, Goffin and Kelly, 1997, Hennighausen and Robinson, 2008). Gene microarray analysis

P4/PR signaling in mouse mammary gland vs. normal human breast

Segregation of ER/PR positive from steroid hormone induced proliferative cells appears to be conserved in the human breast. As determined by immunohistochemistry, the patterning of ER/PR expression in human breast tissue is similar to that of the mouse mammary gland. ER and PR expression is non-uniform and the two steroid receptors colocalize largely in non-proliferating luminal epithelial cells that are nearby ER/PR negative proliferating cells (Anderson et al., 1998, Anderson, 2002, Anderson

Progesterone regulated RANKL signaling in mammary tumorigenesis

A hallmark of pre-neoplastic lesions and mammary tumors is an increase in the proportion of proliferating ER/PR positive cells, suggesting a switch from a paracrine to an autocrine mode of regulation by steroid hormones (Anderson, 2002, Brisken, 2002, Brisken and Rajaram, 2006, Rosen, 2003). In mouse models of hormone-dependent mammary tumors, an increase in the percentage of proliferating ER/PR positive cells is associated with hyperplasias and development of ductal carcinoma in situ (DCIS) (

Progesterone/PR signaling in human breast cancer

The estrogen/ER signaling axis is a major player in stimulating growth and progression of the luminal subtype human breast cancers and indeed targeted anti-estrogen therapies have been successful in treatment of ER positive tumors (Bast et al., 2001). Early on, progesterone was assumed to be of little consequence in the etiology or progression of breast cancer, in part due to its well established role to inhibit proliferation of the uterine endometrium and to promote differentiation of uterine

Conclusion

Much of our understanding of the biology of PR in the normal mammary gland and the role of progesterone/PR signaling in tumor progression comes from studies with mouse experimental models. In part due to lack of suitable experimental models, uncertainties remain concerning the mechanism of action of progesterone and PR in the normal human breast and in the etiology and progression of human breast cancer. Available data indicates that progesterone as a proliferative hormone is a risk factor for

Acknowledgements

Some of the work described in this review was supported in part by NIH Grants DK049030 and HD038129 (DPE) and NIH Training Grant T32 HD007165 (AEO).

References (216)

  • B. Groner et al.

    Hormonal regulation of transcription factor activity in mammary epithelial cells

    Mol. Cell. Endocrinol.

    (1994)
  • E. Anderson et al.

    Estrogen responsiveness and control of normal human breast proliferation

    J. Mammary Gland Biol. Neoplasia

    (1998)
  • E. Anderson

    The role of oestrogen and progesterone receptors in human mammary development and tumorigenesis

    Breast Cancer Res.

    (2002)
  • E. Anderson et al.

    Steroid receptors and cell cycle in normal mammary epithelium

    J. Mammary Gland Biol. Neoplasia

    (2004)
  • T.J. Anderson et al.

    Cell turnover in the “resting” human breast: influence of parity, contraceptive pill, age and laterality

    Br. J. Cancer

    (1982)
  • D. Apter et al.

    Some endocrine characteristics of early menarche, a risk factor for breast cancer, are preserved into adulthood

    Int. J. Cancer

    (1989)
  • M.L. Asselin-Labat et al.

    Steroid hormone receptor status of mouse mammary stem cells

    J. Natl. Cancer Inst.

    (2006)
  • M.L. Asselin-Labat et al.

    Control of mammary stem cell function by steroid hormone signalling

    Nature

    (2010)
  • M.D. Aupperlee et al.

    Progesterone receptor isoforms A and B: temporal and spatial differences in expression during murine mammary gland development

    Endocrinology

    (2005)
  • M.D. Aupperlee et al.

    Differential hormonal regulation and function of progesterone receptor isoforms in normal adult mouse mammary gland

    Endocrinology

    (2007)
  • R. Bagheri-Yarmand et al.

    Metastasis-associated protein 1 deregulation causes inappropriate mammary gland development and tumorigenesis

    Development

    (2004)
  • C. Ballare et al.

    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.

    (2003)
  • A.M. Bamberger et al.

    Modulation of AP-1 activity by the human progesterone receptor in endometrial adenocarcinoma cells

    Proc. Natl. Acad. Sci. USA

    (1996)
  • A.M. Bamberger et al.

    Progesterone receptor isoforms, PR-B and PR-A, in breast cancer: correlations with clinicopathologic tumor parameters and expression of AP-1 factors

    Horm Res.

    (2000)
  • I. Barash

    Stat5 in the mammary gland: controlling normal development and cancer

    J. Cell Physiol.

    (2006)
  • M.H. Barcellos-Hoff et al.

    Transforming growth factor-beta and breast cancer: mammary gland development

    Breast Cancer Res.

    (2000)
  • V.J. Bardou et al.

    Progesterone receptor status significantly improves outcome prediction over estrogen receptor status alone for adjuvant endocrine therapy in two large breast cancer databases

    J. Clin. Oncol.

    (2003)
  • R.C. Bast et al.

    2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: clinical practice guidelines of the American Society of Clinical Oncology

    J. Clin. Oncol.

    (2001)
  • M. Baum et al.

    Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Tamoxifen Alone or in Combination) trial efficacy and safety update analyses

    Cancer

    (2003)
  • C. Beger et al.

    Identification of Id4 as a regulator of BRCA1 expression by using a ribozyme-library-based inverse genomics approach

    Proc. Natl. Acad. Sci. USA

    (2001)
  • M. Beleut et al.

    Two distinct mechanisms underlie progesterone-induced proliferation in the mammary gland

    Proc. Natl. Acad. Sci. USA

    (2010)
  • V. Beral

    Breast cancer and hormone-replacement therapy in the million women study

    Lancet

    (2003)
  • T. Berger et al.

    Disruption of the Lcn2 gene in mice suppresses primary mammary tumor formation but does not decrease lung metastasis

    Proc. Natl. Acad. Sci. USA

    (2010)
  • L. Bernstein

    Epidemiology of endocrine-related risk factors for breast cancer

    J. Mammary Gland Biol. Neoplasia

    (2002)
  • L. Bjornstrom et al.

    Cross-talk between Stat5b and estrogen receptor-alpha and -beta in mammary epithelial cells

    J. Mol. Endocrinol.

    (2001)
  • L. Bjornstrom et al.

    Signal transducers and activators of transcription as downstream targets of nongenomic estrogen receptor actions

    Mol. Endocrinol.

    (2002)
  • T. Blick et al.

    Epithelial mesenchymal transition traits in human breast cancer cell lines

    Clin. Exp. Metastasis

    (2008)
  • J.L. Boerner et al.

    Estrogen negatively regulates epidermal growth factor (EGF)-mediated signal transducer and activator of transcription 5 signaling in human EGF family receptor-overexpressing breast cancer cells

    Mol. Endocrinol.

    (2005)
  • C. Bole-Feysot et al.

    Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice

    Endocr. Rev.

    (1998)
  • V. Boonyaratanakornkit et al.

    The role of extranuclear signaling actions of progesterone receptor in mediating progesterone regulation of gene expression and the cell cycle

    Mol. Endocrinol.

    (2007)
  • C. Brisken et al.

    A paracrine role for the epithelial progesterone receptor in mammary gland development

    Proc. Natl. Acad. Sci. USA

    (1998)
  • C. Brisken et al.

    Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling

    Genes Dev.

    (2000)
  • C. Brisken

    Hormonal control of alveolar development and its implications for breast carcinogenesis

    J. Mammary Gland Biol. Neoplasia

    (2002)
  • C. Brisken et al.

    Alveolar and lactogenic differentiation

    J. Mammary Gland Biol. Neoplasia

    (2006)
  • Bulynko, Y.A., O’Malley, B.W., 2010. Nuclear Receptor Coactivators: Structural and Functional Biochemistry....
  • A.C. Buser et al.

    Progesterone receptor repression of prolactin/signal transducer and activator of transcription 5-mediated transcription of the beta-casein gene in mammary epithelial cells

    Mol. Endocrinol.

    (2007)
  • A.C. Buser et al.

    Progesterone receptor directly inhibits beta-casein gene transcription in mammary epithelial cells through promoting promoter and enhancer repressive chromatin modifications

    Mol. Endocrinol.

    (2011)
  • K.M. Cadigan et al.

    Wnt signaling: a common theme in animal development

    Genes Dev.

    (1997)
  • J. Cao et al.

    Estradiol represses prolactin-induced expression of Na+/taurocholate cotransporting polypeptide in liver cells through estrogen receptor-alpha and signal transducers and activators of transcription 5a

    Endocrinology

    (2004)
  • R.P. Carnevale et al.

    Progestin effects on breast cancer cell proliferation, proteases activation, and in vivo development of metastatic phenotype all depend on progesterone receptor capacity to activate cytoplasmic signaling pathways

    Mol. Endocrinol.

    (2007)
  • Cited by (141)

    • Clinical implementation of biomarkers and signaling pathway as novel targeted therapeutics in breast cancer

      2023, Treatment Landscape of Targeted Therapies in Oncology: Challenges and Opportunities
    View all citing articles on Scopus
    View full text