<<

65 1

Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T35–T48 Endocrinology sensors THEMATIC REVIEW 90 YEARS OF PROGESTERONE Steroid receptors as MAPK signaling sensors in : let the fates decide

Amy R Dwyer1,*, Thu H Truong1,*, Julie H Ostrander1,2 and Carol A Lange1,2,3

1Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA 2Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA 3Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA

Correspondence should be addressed to C A Lange: [email protected]

*(A R Dwyer and T H Truong contributed equally to this work)

This review forms part of a special section on 90 years of progesterone. The guest editors for this section are Dr Simak Ali, Imperial College London, UK and Dr Bert W O’Malley, Baylor College of Medicine, USA

Abstract

Steroid hormone receptors (SRs) are classically defined as ligand-activated transcription Key Words factors that function as master regulators of gene programs important for a wide range ff glucocorticoid receptors of processes governing adult physiology, development, and cell or tissue . ff progesterone receptors A second function of SRs includes the ability to activate cytoplasmic signaling pathways. ff (ER), androgen (AR), and progesterone (PR) receptors bind directly to ff map membrane-associated signaling molecules including mitogenic protein kinases (i.e. c-SRC ff cancer stem cells and AKT), G-proteins, and ion channels to mediate context-dependent actions via rapid activation of downstream signaling pathways. In addition to making direct contact with diverse signaling molecules, SRs are further fully integrated with signaling pathways by virtue of their N-terminal sites that act as regulatory hot-spots capable of sensing the signaling milieu. In particular, ER, AR, PR, and closely related glucocorticoid receptors (GR) share the property of accepting (i.e. sensing) ligand-independent phosphorylation events by proline-directed kinases in the MAPK and CDK families. These signaling inputs act as a ‘second ligand’ that dramatically impacts cell fate. In the face of drugs that reliably target SR ligand-binding domains to block uncontrolled cancer growth, ligand-independent post-translational modifications guide changes in cell fate that confer increased survival, EMT, migration/invasion, stemness properties, and therapy resistance of non-proliferating SR+ cancer cell subpopulations. The focus of this review is on MAPK pathways in the regulation of SR+ cancer cell fate. MAPK-dependent phosphorylation of PR (Ser294) and GR (Ser134) will primarily be discussed in light of the need to target Journal of Molecular changes in breast cancer cell fate as part of modernized combination therapies. Endocrinology (2020) 65, T35–T48

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access

-19-0274 Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T36 Endocrinology sensors

Introduction Closely related to PR within the SR superfamily, the glucocorticoid (GR) is a ligand-activated Of the 48 ligand-activated and ‘orphan’ (i.e. ligands transcription factor responsible for integrating the remain unidentified) nuclear receptors (NRs), six are physiologic, pathological, and therapeutic functions of related members of the SR superfamily whose steroid hormonal (cortisol, endogenous ) and hormone ligands are primarily derived from cholesterol synthetic glucocorticoids like dexamethasone. GR is metabolism: estrogen receptor (ERα, ERβ), progesterone ubiquitously expressed and drives the expression of receptor (PR), glucocorticoid receptor (GR), androgen glucocorticoid-responsive genes in a cell/tissue- and receptor (AR), and mineralocorticoid receptor (MR). PGR is promoter-specific manner after glucocorticoid binding most well-studied as an estrogen-responsive gene, and PR to the GR C-terminal ligand-binding pocket (Miranda expression is used as a clinical biomarker of intact estrogen et al. 2013, Kumar & Thompson 2019). Consequently, GR signaling or functional ER. There are two predominant activation regulates diverse gene programs controlling PR isoforms: full-length PR-B and N-terminally truncated metabolism, immune system, and CNS function to PR-A that is missing the first 164 amino acids found maintain cellular homeostasis. Like other steroid hormone in full-length PR-B, termed the B-upstream segment receptors, GR consists of three functional domains: (BUS). Although PR-A and PR-B share structural and intrinsically disordered N-terminal (NTD), DNA binding sequence identity downstream of the BUS, this segment (DBD), and ligand binding (LBD). NTD and LBD contain contains regulatory components that account for major transcription Activation Function regions, AF1 and AF2, differences in PR-binding partners, promoter selectivity, respectively (Kumar & Thompson 2005). AF2 is strictly and transcriptional activity between these two isoforms. ligand-dependent, while AF1 can act constitutively in A third uterus-specific isoform, termed PR-C, is a further the absence of the LBD (Oakley & Cidlowski 2013), a truncated PR receptor composed of only the hinge region property shared by both PR (Goswami et al. 2014) and AR and hormone binding domain (HBD) that is incapable (Dehm et al. 2008). Like other SRs, functionally distinct of binding DNA but inhibits PR transactivation. PR-C AF domains of GR mediate transcriptional activation signaling occurs specifically in the fundal myometrium by recruiting coregulatory complexes that remodel during the onset of labor and is likely mediated by direct chromatin, target initiation sites, and stabilize RNA competition for C-terminal binding partners (Condon polymerase II for gene transcription (Miranda et al. 2013). et al. 2006). PR-A and PR-B are generally co-expressed Similar to PR isoform structure, a further complexity of in PR+ tissues such as the breast or uterus, but are not GR action is that a large cohort of GR isoforms can be always found together in the same cells (Aupperlee et al. created from a single gene (known as NR3C1) by alternate 2005). Along with ER and Her2, total PR levels, rather splicing and at least eight alternative translational start than individual PR isoforms, are measured clinically sites within the GR N-terminal domain sequence (Turner to classify the luminal A (ER+/PR+) and luminal B & Muller 2005, Duma et al. 2006, Presul et al. 2007). The (ER+/PR-low/null) breast cancer subtypes. However, multiple GR isoforms are expressed in a highly tissue- increasing studies have implicated differential PR isoform specific manner and, when expressed as individual expression in the etiology of breast cancer (Mote et al. 2002, isoforms experimentally, GR isoforms regulate remarkably 2015, Rojas et al. 2017, Lamb et al. 2018). Furthermore, distinct transcriptomes (Lu & Cidlowski 2005). PRs dramatically influence ER transcriptional responses Within the NTD of all SRs, serine, threonine, and lysine (Daniel et al. 2015, Mohammed et al. 2015, Singhal et al. residues can be post-translationally modified (Fig. 1). Our 2018); PR-A can transrepress both ER and PR-B (Abdel- prior studies have primarily defined MAPK-dependent Hafiz et al. 2002), while PR-B is an important ER-binding phospho-PR transcriptomes (Knutson et al. 2012, 2017). partner (Daniel et al. 2015, Mohammed et al. 2015). While the extent to which phosphorylation events modify Measuring PR isoform ratios by immunohistochemistry the GR cistrome is still being studied, such modifications (IHC) has been reported (Sletten et al. 2019) but remains significantly alter GR function Galliher-Beckley( et al. challenging due to the lack of reliable that are 2011). For example, recent collaborative studies from our capable of accurately distinguishing PR-A vs PR-B since group (Regan Anderson et al. 2016, 2018) have established there are no unique amino acid sequences in PR-A. Thus, GR as an important sensor of both life stress (i.e. a defining the context-dependent actions of PR isoforms collection of classical GR ligands acting via the LBD) as is crucial to understanding how imbalanced PR-A/PR-B well as inducible cellular (, reactive oxygen species ratios can be exploited therapeutically in the clinic. (ROS), nutrient starvation), and microenvironmental

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T37 Endocrinology sensors

to the gradual loss of hormone responsiveness in which SRs shift to hormone-independent or resistant states; this process is well-studied with regard to ER, while a similar paradigm is emerging for both PR and GR (Shen et al. 2001, Irusen et al. 2002, Matthews et al. 2004, Arpino et al. 2008, Giulianelli et al. 2008). One mechanism by which SRs become less dependent upon natural ligands occurs through hormone-induced rapid activation of cytoplasmic signaling pathways that, in turn, phosphorylate and sustain SR activation in diminishing ligand (Fig. 2). For example, constitutive ER/PR complexes exist in breast cancer cell lines and primary breast tumors (Daniel et al. 2015); trace levels of either estrogen (E2) or progestin (P4) induce rapid (3–15 min) activation of p42/p44 MAPKs (ERK1/2) via cytoplasmic or membrane-associated ER/PR/c-SRC complexes that have been shown to regulate Figure 1 breast cancer cell proliferation in vitro (Migliaccio et al. Post-translational modifications to steroid receptors. Multiple posttranslational modification sites are present in the N-terminal domain 1998, Boonyaratanakornkit et al. 2001, Ballare et al. 2003). (NTD) of GR, PR, ER, and AR. PR and AR also have a serine phosphorylation We and others have shown that the SR coactivator, site in the hinge (H) region. The annotated pro-directed sites are known PELP1, functions as a cytoplasmic scaffold for docking to be modified by MAPKs and/or CDKs and alter SR transcriptional activity and co-factor binding. DNA-binding and ligand-binding of multiple SRs including GR (Kayahara et al. 2008, domains are also indicated. Regan Anderson et al. 2016), AR (Yang et al. 2012) and both ER and PR (Vadlamudi et al. 2001, Truong et al. () stress, largely acting via p38 MAPK-dependent 2018) as well as the SR coactivator, SRC-3 (Truong et al. phosphorylation of Ser134 (Fig. 2). Herein, we review the 2018). Indeed, a common link between GR signaling in regulation of PR and GR by members of the MAPK family TNBC (discussed subsequently) and PR signaling in ER+ and highlight the functional redundancy of these closely luminal breast cancer is the functional role of PELP1 related SRs as key mediators of advanced luminal (PR) and as a scaffold to bring cytoplasmic kinases and SRs into triple negative (GR) breast cancer phenotypes and therapy- close proximity. In TNBC models, PELP1 expression is resistant tumor progression. A major consequence of the upregulated in response to and cellular integration of SR actions with MAPK signaling pathways stress including hypoxia and ROS production. PELP1 and is the establishment of potent feed-forward signaling GR interactions increase in response to the glucocorticoid loops capable of driving persistent, ligand-independent dexamethasone and creation of ROS by hydrogen SR activation needed for sustained biological responses peroxide (Regan Anderson et al. 2016). Furthermore, relevant to breast cancer cell autonomy and metastatic a transcriptional complex containing PELP1, GR, and dissemination. hypoxia inducible factors (HIFs) is recruited to the breast tumor (Brk/PTK6) promoter; stress-induced Studies of signal transduction Brk/PTK6 signaling promotes breast cancer cell survival, migration, and (Castro & Lange 2010, Lofgren MAPK signaling pathways promote et al. 2011, Regan Anderson et al. 2013). Similarly, in ligand-independent actions of phospho-SRs ER+ breast cancer, PELP1 scaffolds SRC-3, ER, and PR to Reversible post-translational modifications (PTMs) of SRs promote ER phosphorylation (i.e. at Ser118 and Ser167) include phosphorylation, SUMOylation, acetylation, and and expression of E2-induced genes involved in cell ubiquitination. These PTMs may account for the functional survival and stem/progenitor cell formation (Truong et al. diversity of receptor isoforms. This section will primarily 2018). Notably, expression of PR-B (but not progesterone) focus on ligand-independent actions of PR and GR; the was required for E2-induced expression of a distinct hormonal regulation of these SRs in breast cancer models ER/PR target gene signature that predicts greatly shortened have been reviewed (Diep et al. 2015, Scheschowitsch survival in luminal breast cancer patients (Daniel et al. et al. 2017, Truong & Lange 2018). Ligand-independent 2015). Treatment of breast cancer cells with SI-2, an SRC-3 SR activation has been proposed as a contributing factor inhibitor, disrupts ER/PR/PELP1 complexes and blocks

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T38 Endocrinology sensors

Figure 2 Phosphorylation of PR and GR establishes a feed-forward signaling paradigm. Phosphorylation of PR Ser294 by p42/p44 MAPKs (A) and GR Ser134 by stress-activated p38 MAPKs (B) modulates SR target gene selection to drive expression of genes that are able to further promote SR phosphorylation. This establishes a feed-forward signaling loop in the absence of steroid ligand resulting in cell survival, migration, EMT, and non-proliferative/stem cell states.

PELP1-induced tumorsphere formation. Importantly, (MAPKs and CDKs) pathways. As discussed subsequently, the PELP1/GR interaction is enhanced by GR Ser134 these signaling inputs to SR N-termini phospho-sites can phosphorylation, while PR Ser294 phosphorylation is dramatically impact cell fate decisions that are highly required for the PELP1/PR/ER interaction. Thus, SRs relevant to breast cancer progression. participate in cytoplasmic signaling complexes that enable them to rapidly sense changes in the signaling Mitogenic signaling inputs to PR phosphorylation microenvironment through direct PTMs. All SRs can be and transcriptional activity phosphorylated at multiple proline-directed sites within the NTD, and PR and AR also contain these sites in the PR transcriptional activity and promoter selection are hinge region (Fig. 1). Specifically, upregulation of MAPK- dramatically influenced by activation of mitogenic and associated signaling pathways (e.g. Akt, ERK1/2, and p38 -dependent protein kinases (i.e. MAPK, Akt, MAPK) in cancer states can lead to ligand-independent CK2, and cylin-dependent kinases (CDK)) that alter PR activation of SRs via phosphorylation at their N-terminal phosphorylation (Zhang et al. 1997, Knotts et al. 2001, phosphorylation sites (Fig. 1). Ligand-independent Pierson-Mullany & Lange 2004, Daniel et al. 2007, phosphorylation at Ser118 (via MAPK) or Ser167 (via Akt) Faivre et al. 2008, Hagan et al. 2011, Dressing et al. are two of the most well-studied phosphorylation sites 2014), SUMOylation (Daniel et al. 2007, Daniel & Lange in ER (Stellato et al. 2016). Additionally, PR (Ser294, Ser 2009), or acetylation (Daniel et al. 2010). Modification 345, and Ser400) and GR (Ser134) also undergo ligand- of SRs may not appreciably alter transcriptional activity, independent phosphorylation in response to growth as measured using reporter gene assays in vitro, but is a factor or cytokine induced activation of cytoplasmic kinase major mechanism for regulating endogenous target gene

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T39 Endocrinology sensors selection. PTM sites identified on PR that are known to alter Recently, PRs have been implicated in mechanisms of its transcriptional activity at selected endogenous target early luminal breast cancer dissemination; progesterone genes include phosphorylation (S81 unique to PR-B, S294, induced migration of mammary cells from early lesion, S345, and S400), SUMOylation (K388), and acetylation but not primary tumor samples from BALB-NeuT mice (K183, K638, K640, and K641). These PTMs enable PR to (Hosseini et al. 2016). Breast tumor cells co-associate with induce diverse biological outcomes such as anchorage- local and circulating carcinoma-associated fibroblasts independent growth/survival, tumorsphere formation as (c-CAFs) that can be detected as cancer stem cell/c-CAF a readout of stemness properties, and cell cycle-dependent cell co-clusters in both mouse models of breast cancer gene regulation. These biologies are context-dependent as and the blood of patients with metastatic disease (Ao determined by the presence/absence of activated signaling et al. 2015). Related to these studies, fibroblast growth pathways and the availability of coregulators. factor 2 (FGF-2), produced by CAFs, has been shown to For example, PR Ser294 phosphorylation is ligand- activate PR in breast cancer models (Giulianelli et al. 2008). induced (progestins), but also occurs through ligand- This ultimately leads to transcription of known PR target independent pathways in response to tyrosine kinase genes MYC and CCND1, resulting in increased hormone- receptor-induced MAPK and/or CDK independent cell proliferation in mouse mammary tumor activation. Phospho-Ser294 PR-B species are deSUMOylated models. Follow-up studies further defined that FGF-2 at Lys388 and are subject to more rapid ligand-dependent promotes interaction between ER and PR at the MYC receptor downregulation relative to dephosphorylated enhancer and proximal promoter regions (Giulianelli et al. and heavily SUMOylated PR species (Daniel et al. 2007). 2019). Notably, over 700 canonical proteins were recruited Lys388 SUMOylation is a transcriptionally repressive to MYC regulatory sequences following FGF-2 induction, modification at PR target genes associated with cancer which included both PR isoforms (PR-A and PR-B) as well as biology that also stabilizes PRs in the presence of progestin the expression of a novel PR splice variant protein product by slowing their 26S proteasome/ubiquitinoylation- PR-BΔ4. These studies suggest that the PR-BΔ4 variant, dependent degradation. However, a number of genes which has an impaired ligand-binding domain and lacks (i.e. including numerous tumor suppressor genes) are the nuclear localization signal, may play a significant role induced by SUMOylated PRs. Co-mutation of PR-B Lys388 in ligand-independent PR actions, given that it is induced (K388R) rescues transcriptional deficiencies of phospho- and activated by FGF-2 but not progestins. The inclusion mutant PR-B S294A receptors in luciferase assays (Daniel of non-canonical PR isoforms such as PR-BΔ4 in studies of et al. 2007). Interestingly, studies with SUMO-deficient ER+ breast cancer may reveal alternate avenues to prevent mutant PR-B (K388R; a biochemical phospho-mimic) or treat endocrine resistance and effectively block luminal demonstrated that SUMO-deficient PR-B exhibited breast cancer progression and metastasis. Namely, CAF- transcriptional hyperactivity at a unique subset of PR target derived FGF-2 represents a potent second ligand that is genes crucial for cell cycle regulation, proliferation, and sensed by PRs in order to sustain ligand-independent PR survival. This occurred independently of progestins and activation in disseminated cancer cell/CAF clusters and was increased by activated CDK2 (Daniel & Lange 2009). reveals a novel strategy to target phospho-PR actions in Additionally, breast cancer cell models expressing K388R otherwise hormone-ablated breast cancer patients. PR exhibited an alternative gene program significantly Stress signaling inputs to GR phosphorylation and associated with Her2 signaling in luminal B (HER2+) transcriptional activity breast and the emergence of endocrine resistance (Knutson et al. 2012). These studies suggest that, in breast Glucocorticoids (i.e. GR agonists) are one cancer cells with increased MAPK or CDK2 signaling, of the most commonly prescribed and effective treatment phospho-Ser294 PR-B (i.e. SUMO-deficient) species regimens for acute inflammatory conditions as well as promote the expression of transcriptional programs that many chronic inflammatory diseases. However, some can contribute to tumor heterogeneity (e.g. emergence of patients become unresponsive to these treatments due to HER2+ cells) during luminal tumor progression (Knutson the development of glucocorticoid resistance. One of the et al. 2012). Crosstalk between Ser294 and Lys388 in mechanisms that accounts for this resistance is decreased PR-A was less apparent relative to that in PR-B (Daniel glucocorticoid signaling, which is correlated with changes et al. 2007). Additional studies are ongoing to define in GR phosphorylation (Bloom 2004, Tsitoura & Rothman the isoform-specific contributions of SUMO-deficient 2004). GR phosphorylation by ubiquitous protein kinases (i.e. K388R mutant) and Ser294-phosphorylated PR-A. has been shown to lead to altered GR transcriptional

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T40 Endocrinology sensors activity and interaction with other coregulatory proteins and aromatase inhibitors (AIs)) inhibit proliferation (Krstic et al. 1997, Galliher-Beckley et al. 2011). GR is and are very effective treatments for ER-positive phosphorylated on serine residues Ser113, Ser134, Ser141, breast cancer. Yet, the benefits of endocrine therapy Ser203, Ser211, and Ser226 by cyclin-dependent kinases, are not sustained long-term in 10% to 41% of patients MAPK, and casein kinase II (Ismaili & Garabedian 2004) who suffer from late recurrence (Pan et al. 2017). It is (Fig. 1). Three of these GR residues (Ser204, Ser211, and hypothesized that non-proliferative cell states, such as Ser226) are located within the AF1 domain, while the quiescence/tumor cell dormancy, cell survival, plasticity, remainder are found in the NTD (Mason & Housley 1993, and epithelial to mesenchymal transition (EMT) as Ismaili & Garabedian 2004). well as cancer stem cells (CSCs), are not well-targeted Phosphorylation of Ser134 by p38 MAPK is the by standard cancer therapies. Indeed, a growing body most well-studied GR PTM event. Ligand-independent of literature suggests that breast CSCs are resistant to phosphorylation of Ser134 in response to stress-activating current therapeutic approaches (Easwaran et al. 2014, stimuli requires p38 MAPK in U2OS osteosarcoma cells Piva et al. 2014). In fact, these non-proliferative cell (Galliher-Beckley et al. 2011) and TNBC models (Regan states are induced or maintained in response to cellular Anderson et al. 2016). In TNBC cells, p38 MAPK inhibition stress signaling and the selective pressure created by blocked Ser134 GR phosphorylation and abrogated the radiation, chemo-, and endocrine therapy that is largely ability of GR to associate with a GRE-containing region cytostatic rather than cytotoxic, ultimately contributing of the BRK (breast tumor kinase) promoter (Regan to therapy-resistant cancer recurrence and metastasis. Anderson et al. 2016). Ligand-independent regulation For example, chemotherapy has been shown to increase of GR transcriptional activity is further supported by CD44+/CD24- CSC populations and tumorsphere studies showing p38 MAPK-induced phosphorylation formation in breast cancer patients (Li et al. 2008). In of Ser134 increased the association of GR with the 14-3- particular, CSCs tend to be non-proliferative, allowing 3zeta signaling proteins on gene regulatory regions in them to readily evade standard therapies and expand chromatin, which simultaneously resulted in reduced over time (Fillmore & Kuperwasser 2008). Recent studies hormone-dependent transcriptional responses (Galliher- utilizing mouse models of prolactin-induced ER+ breast Beckley et al. 2011). Interestingly, p38 MAPK has been cancer showed that treatment with antiestrogens such as shown to be active in alveolar macrophages of asthmatic fulvestrant or reproducibly stimulated breast patients who exhibited poor response to glucocorticoids CSC self-renewal (Shea et al. 2018). These studies suggest when compared to patients with a normal response that antiestrogen therapies may initially slow tumor (Bhavsar et al. 2008). In contrast, p38 MAPK has also been growth, but also concurrently promote unwanted side shown to phosphorylate Ser211 in human leukemic cells, effects by evoking plasticity and regenerative CSC activity leading to increased dexamethasone-driven apoptosis in largely non-proliferative cancer cell compartments. (Khan et al. 2017). This suggests that p38 MAPK kinase The proliferative effects of progestins in normal phosphorylation of GR impacts ligand-independent breast development are well-established. Progestins GR signaling in a cell-specific manner. In breast cancer are proliferative in the breast (McGowan et al. 2007), models, p38 MAPK signaling is emerging as an important and PR-B is the predominant isoform involved in mediator of stem cell biology (Lu et al. 2018, Xu et al. 2018, normal mammary gland tissue expansion during breast Grun et al. 2019) and other non-proliferative states that development after puberty (Mulac-Jericevic et al. 2003). are relevant to cancer biology (discussed subsequently). Mouse models lacking PR-B, but not PR-A, exhibited defects in mammary gland branching and alveologenesis Studies of cancer cell biology (Mulac-Jericevic et al. 2003). Conversely, using knockout mouse models, PR-A has been shown to be required for PR regulation of breast cancer cell proliferation vs normal uterine development and fertility (Conneely et al. stem cell biology 2001). More recently, PRs have been found to promote Most cancer therapies were historically designed to non-proliferative phenotypes that include cell survival target rapidly dividing tumor cells. In the context of and regulation of cancer stem/stem-like cell self-renewal. ER-positive breast cancer, tumor cells are dependent on For example, progestin stimulation of PR+ mammary ER transcriptional activity to promote proliferation. Thus, epithelial cells induced transcription and secretion of endocrine therapies that target ER function (i.e. selective mitogenic factors such as Wnts, Areg, HB-EGF, and ER modulators (SERMs), selective ER degraders (SERDs), RANKL leading to proliferation of neighboring PR- cells

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T41 Endocrinology sensors

(Fata et al. 2001, Tanos et al. 2013). Separate studies driven secondary tumorsphere formation, an in vitro implicated these same PR signaling outputs (i.e. RANKL and readout of CSC self-renewal (Truong et al. 2019); mutation Wnt4) in the maintenance and expansion of the normal of PR Ser294 to Ala blocked these effects. These findings mammary gland stem compartment (Asselin-Labat et al. indicate that PR Ser294 phosphorylation is a crucial 2010, Joshi et al. 2010). These studies have been extended event that defines both PR-A and PR-B actions in the to show that PR regulates the breast CSC compartment regulation of CSC populations; the increased potency of (Axlund et al. 2013, Cittelly et al. 2013, Hilton et al. 2014, PR-A over PR-B as a driver of breast cancer stemness may Finlay-Schultz & Sartorius 2015, Goodman et al. 2016), be explained by its greater stability (Faivre et al. 2008) and recent work has further shown that PR isoforms have and resistance to deSUMOylation relative to PR-B (Daniel divergent roles in supporting breast CSC properties using et al. 2007). Understanding how SRs contribute to cancer ER+ breast cancer cell line models that either lack both cell fate transitions, plasticity, heterogeneity, and other PRs or express only PR-A, only PR-B, or both isoforms non-proliferative cancer cell biologies is important for (Truong et al. 2019). Measurement of breast CSC markers preventing and targeting therapy resistance. in these models indicated that although PR-A+ breast cancer cells were only weakly proliferative in soft-agar, PR signaling impacts the cell cycle they formed abundant tumorspheres that were enriched for both ALDH+ and CD44+/CD24- cell populations One functional characteristic of CSCs is the ability to relative to PR-B+ cells (which formed fewer but larger maintain a cellular quiescence, reversibly existing in spheres), suggesting a reversible phenotypic flip from the G0 phase of the cell cycle (Bighetti-Trevisan et al. pro-proliferative (PR-B-driven) to non-proliferative CSC 2019). This quiescent state allows for long-term survival (PR-A-driven) programs. Accordingly, S294A PR-A mutant in response to cellular or microenvironmental stress (i.e. models were impaired with regard to ALDH+ tumorsphere chemotherapy, hypoxia, etc.). Signaling molecules that formation, yet exhibited increased proliferation in soft regulate quiescence include cyclin-dependent kinase agar (Truong et al. 2019). These studies suggest tight (CDK) inhibitors (i.e. p21, p27, and p57), as well as the linkage between highly plastic (i.e. proliferative vs CSC) members of the FOXO family of transcription factors. Our regulated cell fates, and implicates PR isoform-specific prior work showed that PRs regulate the cyclin dependent modulation. kinase inhibitor p21 and FOXO1 to promote cellular Divergent roles of PR isoforms may be explained by senescence, the irreversible exit from the cell cycle (i.e. isoform-specific transcriptional control of overlapping permanent accumulation in G0), in ovarian cancer models but distinct gene programs which, in part, is directed by (Diep et al. 2013, 2016). Senescence, FOXO1 and p21 gene PTMs. Notably, phospho-Ser294 species are required for expression, and PR Ser294 phosphorylation were induced both PR-A- and PR-B-induced tumorsphere formation by progestin (ligand-dependent). Chemical inhibition (Knutson et al. 2017, Truong et al. 2019). However, of FOXO1 inhibited PR Ser294 phosphorylation and the phospho-PR-A is preferentially recruited to promoters of PR-dependent senescence phenotype. In these studies, CSC-associated genes (e.g. Wnt4, KLF4, and NOTCH2) PR-B was the dominant isoform required for induction relative to phospho-PR-B; PR isoform recruitment of both FOXO1 and p21 expression as well as progestin- was significantly reduced in phospho-mutant models induced cellular senescence, while PR-A repressed (i.e. S294A). Of note, the effect of PR isoforms on CSC these genes and failed to induce a senescent state upon markers and tumorsphere formation was independent treatment of ovarian cancer cells with progestins. of exogenously added progestin (i.e. ligand-independent Many of the same signaling molecules and transcription in defined media), and the addition of progestin did factors regulate both senescence and quiescence. While a not have a significant effect on tumorsphere number or direct link between PR and CSC quiescence has not yet size. In the context of endocrine therapy resistance, SRs been established in ER+/PR+ breast cancer models, it is may promote tumor progression via ligand-independent possible that context-dependent signals, such as breast vs mechanisms, resulting from the upregulation of growth ovarian cells of origin, cancer type-specific or MAPK/CDK factor and/or stress signaling pathways that occurs signaling pathway activation, and/or ligand-independent following hormone withdrawal (Nicholson et al. 2004, PR phosphorylation, will promote a PR-dependent Need et al. 2015). Relevant to this concept, EGF induces cellular quiescence that contributes to CSC survival. In MAPK-dependent and ligand-independent PR Ser294 support of this, FOXO1 expression increased PR Ser294 phosphorylation, which greatly enhanced PR-A and PR-B phosphorylation and promoted ALDH+ tumorsphere

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T42 Endocrinology sensors formation in both PR-A and PR-B expressing breast cancer proteins (Wu et al. 2004, Regan Anderson et al. 2018). models (Truong et al. 2019). Accordingly, inhibition For example, GR promotes breast tumor kinase (Brk/PTK6) of PR and FOXO1 with onapristone and AS1842856, expression in response to dexamethasone (ligand- respectively, had a synergistic inhibitory effect on CSC dependent), and also in response to hypoxia, cell self-renewal in multiple breast cancer models. Future stress induced by chemotherapy, and anoikis (ligand- studies will examine the cooperative effect of PR isoforms, independent). In TNBC models, hypoxia, , FOXO1, and p21 on CSC quiescence/tumor cell dormancy 5-FU, and non-adherent culture all led to p38-MAPK- in ER+ breast cancer models. dependent GR Ser134 phosphorylation and ligand- independent activation of gene programs implicated in cellular stress-response (Regan Anderson et al. 2016, GR promotes triple negative breast cancer cell 2018). GR-dependent Brk/PTK6 induction protected survival, migration, and invasion cells from chemotherapy-induced cell death and As with PR isoforms, the effects of GR on breast cancer promoted (Regan Anderson et al. 2018). biology are also highly context dependent and most These studies further our mechanistic understanding relevant to non-proliferative cancer cell fates. Like PR of how GR and glucocorticoids promote plasticity expression, GR expression in ER+ luminal breast cancer in order to protect TNBC cells from cell stress and is associated with a better prognosis. In sharp contrast, chemotherapy-induced cell death and may have clinical however, high GR expression is associated with more implications for the routine use of high-dose steroids aggressive triple negative breast cancer (TNBC) biology prior to chemotherapy. and poor prognosis when compared to patients with Cells sense both local external (TME) and internal low GR expression (Pan et al. 2011). In TNBC models, stress (hypoxia) via activation of JNKs and p38 MAPKs, expression of GR and/or GR-associated target genes also historically known as stress-activated protein kinases (i.e. Brk/PTK6) have been associated with pro-survival or SAPKs (Laderoute et al. 1999, Sumbayev & Yasinska signaling, EMT, and cellular migration/invasion in vitro 2005). Notably, upregulation of the p38 MAPK signaling as well as metastasis in vivo (Lofgren et al. 2011, Regan pathway has emerged as a mechanism by which tumor Anderson et al. 2013, 2016, 2018). Recently, activation of cells increase their metastatic capacity. Mechanistic GR with glucocorticoids was shown to drive breast cancer studies have shown that p38 MAPK signaling promotes metastasis in vivo (Obradovic et al. 2019). the invasive and metastatic capacities of breast tumor Cancer cell survival or resisting cell death is an cells (Limoge et al. 2017). As discussed previously, original hallmark of cancer (Hanahan & Weinberg phosphorylation of Ser134 GR is p38 MAPK-dependent 2000). Cellular mechanisms associated with cell survival in osteosarcoma (Galliher-Beckley et al. 2011) and breast include the regulation of pro- and anti-apoptotic BH3 cancer models (Regan Anderson et al. 2016). The role of family proteins, the extrinsic death receptor signaling hypoxia/HIFs in promoting EMT and increased migration molecules, and the intrinsic caspase and apoptosome of TNBC cell models is well-established (Brooks et al. 2016, proteins that ultimately lead to cell death. Intrinsic Tirpe et al. 2019); our most recent studies indicate that activation of cell death pathways is often initiated by GR is a principle driver of TME- (i.e. cytokine) and stress- cell stress caused by DNA damage, ROS production, induced TNBC migration, specifically via p38 MAPK- or metabolic/nutrient stress. GR is a long-established dependent GR Ser134 phosphorylation (C Perez Kerkvliet sensor of physiologic (i.e. hormonal) stress, and more and CA Lange, unpublished results). While these effects recent data indicate that GR is also an exquisite sensor in TNBC cell models were ligand-independent, others of cellular stress and stress signaling within the TME. have reported that glucocorticoid treatment promoted In response to systemic glucocorticoid exposure, GR metastasis in vivo using patient-derived xenograft (PDX) is known to promote cell death in lymphocytes and models (Obradovic et al. 2019). It is important to note monocytes in order to reduce inflammation Strehl( et al. that GR activation by glucocorticoids has been shown to 2019). However, activation of GR also promotes cell suppress the migration of immune cells, highlighting the survival in a variety of epithelial cell types, including tissue- and cell-specific roles of GR/phospho-GR (Stahn in breast cancer models. In studies of TNBC, ligand- & Buttgereit 2008) and underscoring the need to select dependent and ligand-independent GR activation have patients using appropriate biomarkers of phospho-GR been shown to promote expression of pro-survival signaling in GR+ TNBC patients.

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T43 Endocrinology sensors

Ligand-independent SRs are important (Perez Kerkvliet et al. 2020). Thus, targeting phospho-SR therapeutic targets species may be possible using existing agents or newer, more selective SR antagonists paired with the appropriate Phospho-SR species are predicted to be valuable MEK/MAPK or CDK signaling pathway inhibitors (Table 1). biomarkers of aggressive breast cancer behavior. Detection Progestins (megestrol acetate or Megase) as well as of phosphorylated receptors or their respective target gene antiprogestins (RU486, onapristone) were effective in signatures may provide a useful means by which to further early clinical trials, particularly when paired with Tam classify more aggressive breast tumors and thus estimate or ICI 162384 (Robertson et al. 1989, 1999, Nishino et al. risk of therapy-resistant recurrence. An important caveat 1994, Klijn et al. 2000), but had significant toxicity, to targeting phospho-SR species is that these species likely due, in part, to cross-reactivity with ubiquitously may be insensitive to existing receptor antagonists; expressed GRs. This problem may be overcome by use of phosphorylation events confer partial agonist activity lower dose or timed-release antiprogestin preparations, to SR antagonists (Wardell et al. 2010) and phospho-ER as in the case of Apristor (Context Therapeutics, Inc), a species are strongly implicated in the development of clinical formulation of onapristone. Historical clinical tamoxifen resistance (Likhite et al. 2006, Kastrati et al. studies that clearly implicated progestins (namely MPA, 2019). Notably, RU486 (an antagonist for both PR and a PR agonist) in elevated breast cancer risk (Rossouw GR) does not significantly reduce ligand-independent et al. 2002, MWS 2003, Soini et al. 2016) have renewed phosphorylation of PR or GR. However, at least in vitro, interest in targeting PRs as a part of combination ligand-independent actions of phospho-SRs that are endocrine therapies. As a result, selective antiprogestins not further stimulated by agonists are often sensitive to (onapristone and telapristone acetate) have re-entered selected antagonists. For example, ER-scaffolding actions Phase I-II trials with encouraging results (Cottu et al. 2018, of PR-B appear to be insensitive to type I antagonists (i.e. Lee et al. 2020). Similarly, GR antagonists (i.e. RU486) are RU486) that induce PR Ser294 phosphorylation levels being tested in TNBC clinical trials (Nanda et al. 2016). similar to that of PR agonists (Daniel et al. 2015), but are Of concern is that the science of PR and GR (i.e. in breast blocked by type II antagonists (i.e. onapristone) that also cancer models), while growing, lags far behind that of ER block ligand-induced PR Ser294 phosphorylation (Beck or even AR and fewer ligands are available for pre-clinical et al. 1996). Similarly, high basal TNBC cell migration is modeling of PR or GR-driven actions relative to a wealth only weakly stimulated by the GR agonist dexamethasone, of selective receptor modulators for ER or AR. Before but completely blocked by the GR antagonist, RU486 selective SR modulators can be successfully combined

Table 1 PR and GR post-translational modifications and associated ligand-dependent (LD) and ligand-independent (LI) biological outcomes.

Steroid Kinase receptor PTM pathway Biological outcome PR S81 CK2 Proliferation, survival, migration, and transcriptional regulation (Hagan et al. 2011) PR S294 (Phosphorylation) MAPK, CDK Nuclear localization (LI; Qui & Lange 2003), turnover, transcriptional regulation (LD; Daniel & Lange 2009, Shen et al. 2001), proliferation (Daniel et al. 2007), stemness (Truong et al. 2019), senescence (Diep et al. 2013), and cell cycle progression (Moore et al. 2001) PR S345 (Phosphorylation) MAPK, CDK2 Proliferation, migration (Faivre et al. 2008, Dressing et al. 2014) PR K388 (SUMOylation) – Transcriptional regulation and slows PR turnover (Abdel-Hafizet al. 2002, Daniel et al. 2007) PR S400 (Phosphorylation) CDK2 Cell cycle progression and LI transcription, (Zhang et al. 1997, Pierson- Mullany & Lange 2004, Wardell et al. 2010) PR S676 (Phosphorylation) CDK2 Transcriptional activity (Knotts et al. 2001) PR K638-K641 (KxKK, Acetylation – Disrupts nuclear translocation and delays MAPK-induced Ser345 and consensus sequence) Ser294 phosphorylation (Daniel et al. 2010) GR S134 (Phosphorylation) p38 Response to stress-activating stimuli/p38 MAPK, hypoxia (Regan Anderson et al. 2016), stress-induced survival, migration, and stemness (Perez Kerkvliet et al. 2020) GR S211 (Phosphorylation) p38 Apoptosis (LD; Khan et al. 2017) GR S404 (Phosphorylation) GSK-3 Turnover and resistance to LD apoptosis (Galliher-Beckley et al. 2008)

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T44 Endocrinology sensors with existing therapies, the knowledge gap of the roles Biological Chemistry 277 33950–33956. (https://doi.org/10.1074/jbc. that PR/GR play as modulators of other SR (i.e. ER or AR) M204573200) Ao Z, Shah SH, Machlin LM, Parajuli R, Miller PC, Rawal S, Williams AJ, actions and the impact of phosphorylation events to SRs Cote RJ, Lippman ME, Datar RH, et al. 2015 Identification of cancer- on hormone responsiveness and aggressive breast cancer associated fibroblasts in circulating blood from patients with biology must be elucidated. metastatic breast cancer. Cancer Research 75 4681–4687. (https://doi. org/10.1158/0008-5472.CAN-15-1633) Arpino G, Wiechmann L, Osborne CK & Schiff R 2008 Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor Conclusions family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocrine Reviews 29 217–233. (https://doi. In summary, SRs act as highly context-dependent sensors org/10.1210/er.2006-0045) of the cellular signaling environment underpinned by Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER, Yasuda H, Smyth GK, Martin TJ, Lindeman GJ, et al. 2010 Control of PTMs that are intricately controlled by growth factors mammary stem cell function by steroid hormone signalling. Nature and within the and 465 798–802. (https://doi.org/10.1038/nature09027) their intracellular MAPK effectors. Phospho-PR and -GR Aupperlee MD, Smith KT, Kariagina A & Haslam SZ 2005 Progesterone receptor isoforms A and B: temporal and spatial differences in exhibit extensive functional redundancy with regard to expression during murine mammary gland development. shared ligands and binding sites in chromatin/target gene Endocrinology 146 3577–3588. (https://doi.org/10.1210/en.2005- overlap. Both SRs are also capable of initiating feed-forward 0346) Axlund SD, Yoo BH, Rosen RB, Schaack J, Kabos P, Labarbera DV & signaling loops, a paradigm in which their target gene Sartorius CA 2013 Progesterone-inducible cytokeratin 5-positive cells products further phosphorylate the initiating SR, leading in luminal breast cancer exhibit progenitor properties. Hormones and to a sustained signal that can be maintained indefinitely Cancer 4 36–49. (https://doi.org/10.1007/s12672-012-0127-5) Ballare C, Uhrig M, Bechtold T, Sancho E, Di Domenico M, Migliaccio A, in the absence of steroid ligands. Understanding this Auricchio F & Beato M 2003 Two domains of the progesterone mechanism is critical for the treatment of both luminal receptor interact with the estrogen receptor and are required for ER+ and triple negative breast cancers; sustained progesterone activation of the c-Src/Erk pathway in mammalian cells. Molecular and Cellular Biology 23 1994–2008. (https://doi. phospho-SR signaling may contribute to the emergence org/10.1128/mcb.23.6.1994-2008.2003) of therapy-resistant biologies in a minority population of Beck CA, Zhang Y, Weigel NL & Edwards DP 1996 Two types of anti- cancer cells, including non-proliferative stem or dormant progestins have distinct effects on site-specific phosphorylation of human progesterone receptor. Journal of Biological Chemistry 271 cell states, and cell fates associated with EMT, migration, 1209–1217. (https://doi.org/10.1074/jbc.271.2.1209) survival, and therapy resistance. Therapeutically, targeting Bhavsar P, Hew M, Khorasani N, Torrego A, Barnes PJ, Adcock I & this paradigm will likely involve expanding hormonal Chung KF 2008 Relative corticosteroid insensitivity of alveolar macrophages in severe asthma compared with non-severe asthma. modulation/kinase inhibitor repertoires to include agents Thorax 63 784–790. (https://doi.org/10.1136/thx.2007.090027) specifically targeting phospho-SRs and their target genes. Bighetti-Trevisan RL, Sousa LO, Castilho RM & Almeida LO 2019 Cancer stem cells: powerful targets to improve current anticancer therapeutics. Stem Cells International 2019 9618065. (https://doi. org/10.1155/2019/9618065) Declaration of interest Bloom JW 2004 Mitogen-activated protein kinase pathways: therapeutic Carol A Lange serves on the Board of Scientific Advisors for Context targets in steroid resistance? Journal of Allergy and Clinical Immunology Therapeutics. The other authors have nothing to disclose. 114 1055–1058. (https://doi.org/10.1016/j.jaci.2004.09.001) Boonyaratanakornkit V, Scott MP, Ribon V, Sherman L, Anderson SM, Maller JL, Miller WT & Edwards DP 2001 Progesterone receptor contains a proline-rich motif that directly interacts with SH3 Funding domains and activates c-Src family tyrosine kinases. Molecular Cell 8 This work was supported by NIH grants R01CA236948 (C A L and J H O), R01 269–280. (https://doi.org/10.1016/s1097-2765(01)00304-5) CA159712 (C A L), F32 CA210340 (T H T), T32 HL007741 (T H T), and NIH’s Brooks DL, Schwab LP, Krutilina R, Parke DN, Sethuraman A, National Center for Advancing Translational Sciences, grant UL1TR002494 Hoogewijs D, Schorg A, Gotwald L, Fan M, Wenger RH, et al. 2016 (A R D). The content is solely the responsibility of the authors and does ITGA6 is directly regulated by hypoxia-inducible factors and enriches not necessarily represent the official views of the National Institutes of for cancer stem cell activity and invasion in metastatic breast cancer Health’s National Center for Advancing Translational Sciences. The Tickle models. Molecular Cancer 15 26. (https://doi.org/10.1186/s12943-016- Family Land Grant Endowed Chair in Breast Cancer Research (C A L) also 0510-x) supported this work. Castro NE & Lange CA 2010 Breast tumor kinase and extracellular signal-regulated kinase 5 mediate Met receptor signaling to cell migration in breast cancer cells. Breast Cancer Research 12 R60. (https://doi.org/10.1186/bcr2622) References Cittelly DM, Finlay-Schultz J, Howe EN, Spoelstra NS, Axlund SD, Hendricks P, Jacobsen BM, Sartorius CA & Richer JK 2013 Progestin Abdel-Hafiz H, Takimoto GS, Tung L & Horwitz KB 2002 The inhibitory suppression of miR-29 potentiates dedifferentiation of breast cancer function in human progesterone receptor N termini binds SUMO-1 cells via KLF4. Oncogene 32 2555–2564. (https://doi.org/10.1038/ protein to regulate autoinhibition and transrepression. Journal of onc.2012.275)

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T45 Endocrinology sensors

Condon JC, Hardy DB, Kovaric K & Mendelson CR 2006 Up-regulation to specificity protein 1 transcription factors. Molecular Endocrinology of the progesterone receptor (PR)-C isoform in laboring myometrium 22 823–837. (https://doi.org/10.1210/me.2007-0437) by activation of nuclear factor-kappaB may contribute to the onset Fata JE, Chaudhary V & Khokha R 2001 Cellular turnover in the of labor through inhibition of PR function. Molecular Endocrinology mammary gland is correlated with systemic levels of progesterone 20 764–775. (https://doi.org/10.1210/me.2005-0242) and not 17beta- during the estrous cycle. Biology of Conneely OM, Mulac-Jericevic B, Lydon JP & De Mayo FJ 2001 Reproduction 65 680–688. (https://doi.org/10.1095/ Reproductive functions of the progesterone receptor isoforms: biolreprod65.3.680) lessons from knock-out mice. Molecular and Cellular Endocrinology Fillmore CM & Kuperwasser C 2008 Human breast cancer cell lines 179 97–103. (https://doi.org/10.1016/s0303-7207(01)00465-8) contain stem-like cells that self-renew, give rise to phenotypically Cottu PH, Bonneterre J, Varga A, Campone M, Leary A, Floquet A, diverse progeny and survive chemotherapy. Breast Cancer Research 10 Berton-Rigaud D, Sablin MP, Lesoin A, Rezai K, et al. 2018 Phase I R25. (https://doi.org/10.1186/bcr1982) study of onapristone, a type I antiprogestin, in female patients with Finlay-Schultz J & Sartorius CA 2015 Steroid hormones, steroid receptors, previously treated recurrent or metastatic progesterone receptor- and breast cancer stem cells. Journal of Mammary Gland Biology and expressing cancers. PLoS ONE 13 e0204973. (https://doi.org/10.1371/ Neoplasia 20 39–50. (https://doi.org/10.1007/s10911-015-9340-5) journal.pone.0204973) Galliher-Beckley A J, Williams J G, Collins J B & Cidlowski J A 2008 Daniel AR & Lange CA 2009 Protein kinases mediate ligand-independent Glycogen synthase kinase 3beta-mediated serine phosphorylation of derepression of SUMOylated progesterone receptors in breast cancer the human glucocorticoid receptor redirects gene expression profiles. cells. PNAS 106 14287–14292. (https://doi.org/10.1073/ Molecular and Cellular Biology 28 7309–7322. (https://doi. pnas.0905118106) org/10.1128/MCB.00808-08) Daniel AR, Faivre EJ & Lange CA 2007 Phosphorylation-dependent Galliher-Beckley AJ, Williams JG & Cidlowski JA 2011 Ligand- antagonism of SUMOylation derepresses progesterone receptor independent phosphorylation of the glucocorticoid receptor action in breast cancer cells. Molecular Endocrinology 21 2890–2906. integrates cellular stress pathways with nuclear receptor signaling. (https://doi.org/10.1210/me.2007-0248) Molecular and Cellular Biology 31 4663–4675. (https://doi. Daniel AR, Gaviglio AL, Czaplicki LM, Hillard CJ, Housa D & Lange CA org/10.1128/MCB.05866-11) 2010 The progesterone receptor hinge region regulates the kinetics Giulianelli S, Cerliani JP, Lamb CA, Fabris VT, Bottino MC, of transcriptional responses through acetylation, phosphorylation, Gorostiaga MA, Novaro V, Gongora A, Baldi A, Molinolo A, et al. and nuclear retention. Molecular Endocrinology 24 2126–2138. 2008 Carcinoma-associated fibroblasts activate progesterone (https://doi.org/10.1210/me.2010-0170) receptors and induce hormone independent mammary tumor Daniel AR, Gaviglio AL, Knutson TP, Ostrander JH, D'Assoro AB, growth: a role for the FGF-2/FGFR-2 axis. International Journal of Ravindranathan P, Peng Y, Raj GV, Yee D & Lange CA 2015 Cancer 123 2518–2531. (https://doi.org/10.1002/ijc.23802) Progesterone receptor-B enhances estrogen responsiveness of breast Giulianelli S, Riggio M, Guillardoy T, Perez Pinero C, Gorostiaga MA, cancer cells via scaffolding PELP1- and estrogen receptor-containing Sequeira G, Pataccini G, Abascal MF, Toledo MF, Jacobsen BM, et al. transcription complexes. Oncogene 34 506–515. (https://doi. 2019 FGF2 induces breast cancer growth through ligand- org/10.1038/onc.2013.579) independent activation and recruitment of ERalpha and PRBDelta4 Dehm SM, Schmidt LJ, Heemers HV, Vessella RL & Tindall DJ 2008 isoform to MYC regulatory sequences. International Journal of Cancer Splicing of a novel androgen receptor exon generates a constitutively 145 1874–1888. (https://doi.org/10.1002/ijc.32252) active androgen receptor that mediates prostate cancer therapy Goodman CR, Sato T, Peck AR, Girondo MA, Yang N, Liu C, Yanac AF, resistance. Cancer Research 68 5469–5477. (https://doi. Kovatich AJ, Hooke JA, Shriver CD, et al. 2016 Steroid induction of org/10.1158/0008-5472.CAN-08-0594) therapy-resistant cytokeratin-5-positive cells in estrogen receptor- Diep CH, Charles NJ, Gilks CB, Kalloger SE, Argenta PA & Lange CA positive breast cancer through a BCL6-dependent mechanism. 2013 Progesterone receptors induce FOXO1-dependent senescence in Oncogene 35 1373–1385. (https://doi.org/10.1038/onc.2015.193) ovarian cancer cells. Cell Cycle 12 1433–1449. (https://doi. Goswami D, Callaway C, Pascal BD, Kumar R, Edwards DP & Griffin PR org/10.4161/cc.24550) 2014 Influence of domain interactions on conformational mobility Diep CH, Daniel AR, Mauro LJ, Knutson TP & Lange CA 2015 of the progesterone receptor detected by hydrogen/deuterium Progesterone action in breast, uterine, and ovarian cancers. Journal of exchange mass spectrometry. Structure 22 961–973. (https://doi. Molecular Endocrinology 54 R31–R53. (https://doi.org/10.1530/JME-14- org/10.1016/j.str.2014.04.013) 0252) Grun D, Adhikary G & Eckert RL 2019 NRP-1 interacts with GIPC1 and Diep CH, Knutson TP & Lange CA 2016 Active FOXO1 is a key SYX to activate p38 MAPK signaling and cancer stem cell survival. determinant of isoform-specific progesterone receptor transactivation Molecular Carcinogenesis 58 488–499. (https://doi.org/10.1002/ and senescence programming. Molecular Cancer Research 14 141–162. mc.22943) (https://doi.org/10.1158/1541-7786.MCR-15-0431) Hagan CR, Regan TM, Dressing GE & Lange CA 2011 ck2-dependent Dressing GE, Knutson TP, Schiewer MJ, Daniel AR, Hagan CR, Diep CH, phosphorylation of progesterone receptors (PR) on Ser81 regulates Knudsen KE & Lange CA 2014 Progesterone receptor-cyclin D1 PR-B isoform-specific target gene expression in breast cancer cells. complexes induce cell cycle-dependent transcriptional programs in Molecular and Cellular Biology 31 2439–2452. (https://doi. breast cancer cells. Molecular Endocrinology 28 442–457. (https://doi. org/10.1128/MCB.01246-10) org/10.1210/me.2013-1196) Hanahan D & Weinberg RA 2000 The hallmarks of cancer. Cell 100 Duma D, Jewell CM & Cidlowski JA 2006 Multiple glucocorticoid 57–70. (https://doi.org/10.1016/s0092-8674(00)81683-9) receptor isoforms and mechanisms of post-translational Hilton HN, Santucci N, Silvestri A, Kantimm S, Huschtscha LI, modification. Journal of Steroid Biochemistry and Molecular Biology 102 Graham JD & Clarke CL 2014 Progesterone stimulates progenitor 11–21. (https://doi.org/10.1016/j.jsbmb.2006.09.009) cells in normal human breast and breast cancer cells. Breast Cancer Easwaran H, Tsai HC & Baylin SB 2014 Cancer epigenetics: tumor Research and Treatment 143 423–433. (https://doi.org/10.1007/ heterogeneity, plasticity of stem-like states, and drug resistance. s10549-013-2817-2) Molecular Cell 54 716–727. (https://doi.org/10.1016/j. Hosseini H, Obradovic MMS, Hoffmann M, Harper KL, Sosa MS, Werner- molcel.2014.05.015) Klein M, Nanduri LK, Werno C, Ehrl C, Maneck M, et al. 2016 Early Faivre EJ, Daniel AR, Hillard CJ & Lange CA 2008 Progesterone receptor dissemination seeds metastasis in breast cancer. Nature 540 552–558. rapid signaling mediates serine 345 phosphorylation and tethering (https://doi.org/10.1038/nature20785)

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T46 Endocrinology sensors

Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF & Adcock IM protein kinase activity. Journal of Biological Chemistry 274 2002 p38 mitogen-activated protein kinase-induced glucocorticoid 12890–12897. (https://doi.org/10.1074/jbc.274.18.12890) receptor phosphorylation reduces its activity: role in steroid- Lamb CA, Fabris VT, Jacobsen B, Molinolo AA & Lanari C 2018 insensitive asthma. Journal of Allergy and Clinical Immunology 109 Biological and clinical impact of imbalanced progesterone receptor 649–657. (https://doi.org/10.1067/mai.2002.122465) isoform ratios in breast cancer. Endocrine-Related Cancer 25 Ismaili N & Garabedian MJ 2004 Modulation of glucocorticoid receptor R605–R624. (https://doi.org/10.1530/ERC-18-0179) function via phosphorylation. Annals of the New York Academy of Lee O, Sullivan ME, Xu Y, Rogers C, Muzzio M, Helenowski I, Shidfar A, Sciences 1024 86–101. (https://doi.org/10.1196/annals.1321.007) Zeng Z, Singhal H, Jovanovic B, et al. 2020 Selective progesterone Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL, receptor modulators in early-stage breast cancer: a randomized, Stingl J, Waterhouse PD & Khokha R 2010 Progesterone induces placebo-controlled Phase II window-of-opportunity trial using adult mammary stem cell expansion. Nature 465 803–807. (https:// telapristone acetate. Clinical Cancer Research 26 25–34. (https://doi. doi.org/10.1038/nature09091) org/10.1158/1078-0432.CCR-19-0443) Kastrati I, Semina S, Gordon B & Smart E 2019 Insights into how Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, phosphorylation of estrogen receptor at serine 305 modulates Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, et al. 2008 tamoxifen activity in breast cancer. Molecular and Cellular Intrinsic resistance of tumorigenic breast cancer cells to Endocrinology 483 97–101. (https://doi.org/10.1016/j. chemotherapy. Journal of the National Cancer Institute 100 672–679. mce.2019.01.014) (https://doi.org/10.1093/jnci/djn123) Kayahara M, Ohanian J, Ohanian V, Berry A, Vadlamudi R & Ray DW Likhite VS, Stossi F, Kim K, Katzenellenbogen BS & Katzenellenbogen JA 2008 MNAR functionally interacts with both NH2- and COOH- 2006 Kinase-specific phosphorylation of the estrogen receptor terminal GR domains to modulate transactivation. American Journal changes receptor interactions with ligand, deoxyribonucleic acid, of Physiology: Endocrinology and Metabolism 295 E1047–E1055. and coregulators associated with alterations in estrogen and (https://doi.org/10.1152/ajpendo.90429.2008) tamoxifen activity. Molecular Endocrinology 20 3120–3132. (https:// Khan SH, McLaughlin WA & Kumar R 2017 Site-specific doi.org/10.1210/me.2006-0068) phosphorylation regulates the structure and function of an Limoge M, Safina A, Truskinovsky AM, Aljahdali I, Zonneville J, intrinsically disordered domain of the glucocorticoid receptor. Gruevski A, Arteaga CL & Bakin AV 2017 Tumor P38MAPK signaling Scientific Reports 7 15440. (https://doi.org/10.1038/s41598-017-15549- enhances breast carcinoma vascularization and growth by promoting 5) expression and deposition of pro-tumorigenic factors. Oncotarget 8 Klijn JGM, Beex LVAM, Mauriac L, van Zijl JA, Veyret C, Wildiers J, 61969–61981. (https://doi.org/10.18632/oncotarget.18755) Jassem J, Piccart M, Burghouts J, Becquart D, et al. 2000 Combined Lofgren KA, Ostrander JH, Housa D, Hubbard GK, Locatelli A, Bliss RL, treatment with buserelin and tamoxifen in premenopausal Schwertfeger KL & Lange CA 2011 Mammary gland specific metastatic breast cancer: a randomized study. Journal of the National expression of Brk/PTK6 promotes delayed involution and tumor Cancer Institute 92 903–911. (https://doi.org/10.1093/ formation associated with activation of p38 MAPK. Breast Cancer jnci/92.11.903) Research 13 R89. (https://doi.org/10.1186/bcr2946) Knotts TA, Orkiszewski RS, Cook RG, Edwards DP & Weigel NL 2001 Lu NZ & Cidlowski JA 2005 Translational regulatory mechanisms Identification of a phosphorylation site in the hinge region of the generate N-terminal glucocorticoid receptor isoforms with unique human progesterone receptor and additional amino-terminal transcriptional target genes. Molecular Cell 18 331–342. (https://doi. phosphorylation sites. Journal of Biological Chemistry 276 8475–8483. org/10.1016/j.molcel.2005.03.025) (https://doi.org/10.1074/jbc.M009805200) Lu H, Tran L, Park Y, Chen I, Lan J, Xie Y & Semenza GL 2018 Reciprocal Knutson TP, Daniel AR, Fan D, Silverstein KA, Covington KR, Fuqua SA regulation of DUSP9 and DUSP16 expression by HIF1 controls ERK & Lange CA 2012 Phosphorylated and SUMOylation-deficient and p38 MAP kinase activity and mediates chemotherapy-induced progesterone receptors drive proliferative gene signatures during breast cancer stem cell enrichment. Cancer Research 78 4191–4202. breast cancer progression. Breast Cancer Research 14 R95. (https://doi. (https://doi.org/10.1158/0008-5472.CAN-18-0270) org/10.1186/bcr3211) Mason SA & Housley PR 1993 Site-directed mutagenesis of the Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, phosphorylation sites in the mouse glucocorticoid receptor. Journal Schwertfeger KL & Lange CA 2017 Posttranslationally modified of Biological Chemistry 268 21501–21504. progesterone receptors direct ligand-specific expression of breast Matthews JG, Ito K, Barnes PJ & Adcock IM 2004 Defective cancer stem cell-associated gene programs. Journal of Hematology and glucocorticoid receptor nuclear translocation and altered histone Oncology 10 89. (https://doi.org/10.1186/s13045-017-0462-7) acetylation patterns in glucocorticoid-resistant patients. Journal of Krstic MD, Rogatsky I, Yamamoto KR & Garabedian MJ 1997 Mitogen- Allergy and Clinical Immunology 113 1100–1108. (https://doi. activated and cyclin-dependent protein kinases selectively and org/10.1016/j.jaci.2004.03.018) differentially modulate transcriptional enhancement by the McGowan EM, Russell AJ, Boonyaratanakornkit V, Saunders DN, glucocorticoid receptor. Molecular and Cellular Biology 17 3947–3954. Lehrbach GM, Sergio CM, Musgrove EA, Edwards DP & (https://doi.org/10.1128/mcb.17.7.3947) Sutherland RL 2007 Progestins reinitiate cell cycle progression in Kumar R & Thompson EB 2005 Gene regulation by the glucocorticoid antiestrogen-arrested breast cancer cells through the B-isoform of receptor: structure:function relationship. Journal of Steroid progesterone receptor. Cancer Research 67 8942–8951. (https://doi. Biochemistry and Molecular Biology 94 383–394. (https://doi. org/10.1158/0008-5472.CAN-07-1255) org/10.1016/j.jsbmb.2004.12.046) Migliaccio A, Piccolo D, Castoria G, Di Domenico M, Bilancio A, Kumar R & Thompson EB 2019 Role of phosphorylation in the Lombardi M, Gong W, Beato M & Auricchio F 1998 Activation of the modulation of the glucocorticoid receptor’s intrinsically disordered Src/p21ras/Erk pathway by progesterone receptor via cross-talk with domain. Biomolecules 9 95. (https://doi.org/10.3390/biom9030095) estrogen receptor. EMBO Journal 17 2008–2018. (https://doi. Laderoute KR, Mendonca HL, Calaoagan JM, Knapp AM, Giaccia AJ & org/10.1093/emboj/17.7.2008) Stork PJ 1999 Mitogen-activated protein kinase phosphatase-1 (MKP- Miranda TB, Voss TC & Hager GL 2013 High-throughput fluorescence- 1) expression is induced by low oxygen conditions found in solid based screen to identify factors involved in nuclear receptor tumor microenvironments. A candidate MKP for the inactivation of recruitment to response elements. Methods in Molecular Biology 1042 hypoxia-inducible stress-activated protein kinase/c-Jun N-terminal 3–12. (https://doi.org/10.1007/978-1-62703-526-2_1)

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T47 Endocrinology sensors

Mohammed H, Russell IA, Stark R, Rueda OM, Hickey TE, Tarulli GA, Phenotypes in Triple-Negative Breast Cancer. Breast Cancer Research Serandour AA, Birrell SN, Bruna A, Saadi A, et al. 2015 Progesterone 22 39. (https://doi.org/10.1186/s13058-020-01277-8) receptor modulates ERalpha action in breast cancer. Nature 523 Pierson-Mullany LK & Lange CA 2004 Phosphorylation of progesterone 313–317. (https://doi.org/10.1038/nature14583) receptor serine 400 mediates ligand-independent transcriptional Moore R K, Otsuka F & Shimasaki S 2001 Role of ERK1/2 in the activity in response to activation of cyclin-dependent protein kinase Differential Synthesis of Progesterone and Estradiol by Granulosa 2. Molecular and Cellular Biology 24 10542–10557. (https://doi. Cells. Biochemical and Biophysical Research Communications 289 org/10.1128/MCB.24.24.10542-10557.2004) 796–800. (https://doi.org/10.1006/bbrc.2001.6052) Piva M, Domenici G, Iriondo O, Rabano M, Simoes BM, Comaills V, Mote PA, Bartow S, Tran N & Clarke CL 2002 Loss of co-ordinate Barredo I, Lopez-Ruiz JA, Zabalza I, Kypta R, et al. 2014 Sox2 expression of progesterone receptors A and B is an early event in promotes tamoxifen resistance in breast cancer cells. EMBO Molecular breast carcinogenesis. Breast Cancer Research and Treatment 72 Medicine 6 66–79. (https://doi.org/10.1002/emmm.201303411) 163–172. (https://doi.org/10.1023/a:1014820500738) Presul E, Schmidt S, Kofler R & Helmberg A 2007 Identification, tissue Mote PA, Gompel A, Howe C, Hilton HN, Sestak I, Cuzick J, Dowsett M, expression, and glucocorticoid responsiveness of alternative first Hugol D, Forgez P, Byth K, et al. 2015 Progesterone receptor A exons of the human glucocorticoid receptor. Journal of Molecular predominance is a discriminator of benefit from endocrine therapy Endocrinology 38 79–90. (https://doi.org/10.1677/jme.1.02183) in the ATAC trial. Breast Cancer Research and Treatment 151 309–318. Qiu M & Lange C A 2003 MAP kinases couple multiple functions of (https://doi.org/10.1007/s10549-015-3397-0) human progesterone receptors: degradation, transcriptional synergy, Mulac-Jericevic B, Lydon JP, DeMayo FJ & Conneely OM 2003 Defective and nuclear association. The Journal of Steroid Biochemistry and mammary gland morphogenesis in mice lacking the progesterone Molecular Biology 85 147–157. (https://doi.org/10.1016/S0960- receptor B isoform. PNAS 100 9744–9749. (https://doi.org/10.1073/ 0760(03)00221-8) pnas.1732707100) Regan Anderson TM, Peacock DL, Daniel AR, Hubbard GK, Lofgren KA, MWS 2003 Breast cancer and hormone-replacement therapy in the Girard BJ, Schorg A, Hoogewijs D, Wenger RH, Seagroves TN, et al. Million Women Study. Lancet 362 419–427. (https://doi.org/10.1016/ 2013 Breast tumor kinase (Brk/PTK6) is a mediator of hypoxia- S0140-6736(03)14065-2) associated breast cancer progression. Cancer Research 73 5810–5820. Nanda R, Stringer-Reasor EM, Saha P, Kocherginsky M, Gibson J, (https://doi.org/10.1158/0008-5472.CAN-13-0523) Libao B, Hoffman PC, Obeid E, Merkel DE, Khramtsova G, et al. Regan Anderson TM, Ma SH, Raj GV, Cidlowski JA, Helle TM, 2016 A randomized phase I trial of albumin-bound Knutson TP, Krutilina RI, Seagroves TN & Lange CA 2016 Breast paclitaxel with or without mifepristone for advanced breast cancer. tumor kinase (Brk/PTK6) is induced by HIF, glucocorticoid receptor, SpringerPlus 5 947. (https://doi.org/10.1186/s40064-016-2457-1) and PELP1-mediated stress signaling in triple-negative breast cancer. Need EF, Selth LA, Trotta AP, Leach DA, Giorgio L, O'Loughlin MA, Cancer Research 76 1653–1663. (https://doi.org/10.1158/0008-5472. Smith E, Gill PG, Ingman WV, Graham JD, et al. 2015 The unique CAN-15-2510) transcriptional response produced by concurrent estrogen and Regan Anderson TM, Ma S, Perez Kerkvliet C, Peng Y, Helle TM, progesterone treatment in breast cancer cells results in upregulation Krutilina RI, Raj GV, Cidlowski JA, Ostrander JH, Schwertfeger KL, of growth factor pathways and switching from a Luminal A to a et al. 2018 Taxol induces Brk-dependent prosurvival phenotypes in Basal-like subtype. BMC Cancer 15 791–791. (https://doi.org/10.1186/ TNBC cells through an AhR/GR/HIF-driven signaling axis. Molecular s12885-015-1819-3) Cancer Research 16 1761–1772. (https://doi.org/10.1158/1541-7786. Nicholson RI, Staka C, Boyns F, Hutcheson IR & Gee JM 2004 Growth MCR-18-0410) factor-driven mechanisms associated with resistance to estrogen Robertson JFR, Williams MR, Todd J, Nicholson RI, Morgan DAL & deprivation in breast cancer: new opportunities for therapy. Blamey RW 1989 Factors predicting the response of patients with Endocrine-Related Cancer 11 623–641. (https://doi.org/10.1677/ advanced breast cancer to endocrine (megace) therapy. European erc.1.00778) Journal of Cancer and Clinical Oncology 25 469–475. (https://doi. Nishino Y, Schneider MR & Michna H 1994 Enhancement of the org/10.1016/0277-5379(89)90259-9) antitumor efficacy of the antiprogestin, onapristone, by Robertson JFR, Willsher PC, Winterbottom L, Blamey RW & Thorpe S combination with the antiestrogen, ICI 164384. Journal of Cancer 1999 Onapristone, a progesterone receptor antagonist, as first-line Research and Clinical Oncology 120 298–302. (https://doi. therapy in primary breast cancer. European Journal of Cancer 35 org/10.1007/bf01236387) 214–218. (https://doi.org/10.1016/s0959-8049(98)00388-8) Oakley RH & Cidlowski JA 2013 The biology of the glucocorticoid Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martinez P, receptor: new signaling mechanisms in health and disease. Journal of Gonzalez P, Hewitt S, He X, Perou CM, et al. 2017 Progesterone Allergy and Clinical Immunology 132 1033–1044. (https://doi. receptor isoform ratio: a breast cancer prognostic and predictive org/10.1016/j.jaci.2013.09.007) factor for antiprogestin responsiveness. Journal of the National Cancer Obradovic MMS, Hamelin B, Manevski N, Couto JP, Sethi A, Institute 109 djw317. (https://doi.org/10.1093/jnci/djw317) Coissieux MM, Munst S, Okamoto R, Kohler H, Schmidt A, et al. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, 2019 Glucocorticoids promote breast cancer metastasis. Nature 567 Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, 540–544. (https://doi.org/10.1038/s41586-019-1019-4) et al. 2002 Risks and benefits of estrogen plus progestin in healthy Pan D, Kocherginsky M & Conzen SD 2011 Activation of the postmenopausal women: principal results from the Women's Health glucocorticoid receptor is associated with poor prognosis in estrogen Initiative randomized controlled trial. JAMA 288 321–333. (https:// receptor-negative breast cancer. Cancer Research 71 6360–6370. doi.org/10.1001/jama.288.3.321) (https://doi.org/10.1158/0008-5472.CAN-11-0362) Scheschowitsch K, Leite JA & Assreuy J 2017 New insights in Pan H, Gray R, Braybrooke J, Davies C, Taylor C, McGale P, Peto R, glucocorticoid receptor signaling-more than just a ligand-binding Pritchard KI, Bergh J, Dowsett M, et al. 2017 20-year risks of breast- receptor. Frontiers in Endocrinology 8 16. (https://doi.org/10.3389/ cancer recurrence after stopping endocrine therapy at 5 years. New fendo.2017.00016) England Journal of Medicine 377 1836–1846. (https://doi.org/10.1056/ Shea MP, O'Leary KA, Fakhraldeen SA, Goffin V, Friedl A, Wisinski KB, NEJMoa1701830) Alexander CM & Schuler LA 2018 Antiestrogen therapy increases Perez Kerkvliet C, Dwyer AR, Diep CH, Oakley RH, Liddle C, plasticity and cancer stemness of prolactin-induced ERalpha(+) Cidlowski JA & Lange 2020 Glucocorticoid Receptors Are Required mammary carcinomas. Cancer Research 78 1672–1684. (https://doi. Effectors of TGFβ1-induced p38 MAPK Signaling to Advanced Cancer org/10.1158/0008-5472.CAN-17-0985)

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access Journal of Molecular A R Dwyer, T H Truong et al. Steroid receptors as MAPK 65:1 T48 Endocrinology sensors

Shen T, Horwitz KB & Lange CA 2001 Transcriptional hyperactivity of Truong TH, Hu H, Temiz NA, Hagen KM, Girard BJ, Brady NJ, human progesterone receptors is coupled to their ligand-dependent Schwertfeger KL, Lange CA & Ostrander JH 2018 Cancer stem cell down-regulation by mitogen-activated protein kinase-dependent phenotypes in ER(+) breast cancer models are promoted by PELP1/ phosphorylation of serine 294. Molecular and Cellular Biology 21 AIB1 complexes. Molecular Cancer Research 16 707–719. (https://doi. 6122–6131. (https://doi.org/10.1128/mcb.21.18.6122-6131.2001) org/10.1158/1541-7786.MCR-17-0598) Singhal H, Greene ME, Zarnke AL, Laine M, Al Abosy R, Chang YF, Truong TH, Dwyer AR, Diep CH, Hu H, Hagen KM & Lange CA 2019 Dembo AG, Schoenfelt K, Vadhi R, Qiu X, et al. 2018 Progesterone Phosphorylated progesterone receptor isoforms mediate opposing receptor isoforms, agonists and antagonists differentially reprogram stem cell and proliferative breast cancer cell fates. Endocrinology 160 estrogen signaling. Oncotarget 9 4282–4300. (https://doi. 430–446. (https://doi.org/10.1210/en.2018-00990) org/10.18632/oncotarget.21378) Tsitoura DC & Rothman PB 2004 Enhancement of MEK/ERK signaling Sletten ET, Arnes M, Lysa LM, Larsen M & Orbo A 2019 Significance of promotes glucocorticoid resistance in CD4+ T cells. Journal of Clinical progesterone receptors (PR-A and PR-B) expression as predictors for Investigation 113 619–627. (https://doi.org/10.1172/JCI18975) relapse after successful therapy of endometrial hyperplasia: a Turner JD & Muller CP 2005 Structure of the glucocorticoid receptor retrospective cohort study. BJOG 126 936–943. (https://doi. (NR3C1) gene 5′ untranslated region: identification, and tissue org/10.1111/1471-0528.15579) distribution of multiple new human exon 1. Journal of Molecular Soini T, Hurskainen R, Grénman S, Mäenpää J, Paavonen J, Joensuu H & Endocrinology 35 283–292. (https://doi.org/10.1677/jme.1.01822) Pukkala E 2016 Levonorgestrel-releasing intrauterine system and the Vadlamudi RK, Wang RA, Mazumdar A, Kim Y, Shin J, Sahin A & risk of breast cancer: a nationwide cohort study. Acta Oncologica 55 Kumar R 2001 Molecular cloning and characterization of PELP1, a 188–192. (https://doi.org/10.3109/0284186X.2015.1062538) novel human coregulator of estrogen receptor alpha. Journal of Stahn C & Buttgereit F 2008 Genomic and nongenomic effects of Biological Chemistry 276 38272–38279. (https://doi.org/10.1074/jbc. glucocorticoids. Nature Clinical Practice: Rheumatology 4 525–533. M103783200) (https://doi.org/10.1038/ncprheum0898) Wardell SE, Narayanan R, Weigel NL & Edwards DP 2010 Partial agonist Stellato C, Porreca I, Cuomo D, Tarallo R, Nassa G & Ambrosino C 2016 activity of the progesterone receptor antagonist RU486 mediated by The ‘busy life’ of unliganded estrogen receptors. Proteomics 16 an amino-terminal domain coactivator and phosphorylation of 288–300. (https://doi.org/10.1002/pmic.201500261) serine400. Molecular Endocrinology 24 335–345. (https://doi. Strehl C, Ehlers L, Gaber T & Buttgereit F 2019 Glucocorticoids-all- org/10.1210/me.2008-0081) rounders tackling the versatile players of the immune system. Wu W, Chaudhuri S, Brickley DR, Pang D, Karrison T & Conzen SD Frontiers in Immunology 10 1744. (https://doi.org/10.3389/ 2004 Microarray analysis reveals glucocorticoid-regulated survival fimmu.2019.01744) genes that are associated with inhibition of apoptosis in breast Sumbayev VV & Yasinska IM 2005 Regulation of MAP kinase-dependent epithelial cells. Cancer Research 64 1757–1764. (https://doi. apoptotic pathway: implication of reactive oxygen and nitrogen org/10.1158/0008-5472.can-03-2546) species. Archives of Biochemistry and Biophysics 436 406–412. (https:// Xu M, Wang S, Wang Y, Wu H, Frank JA, Zhang Z & Luo J 2018 Role of doi.org/10.1016/j.abb.2005.02.021) p38γ MAPK in regulation of EMT and cancer stem cells. Biochimica et Tanos T, Sflomos G, Echeverria PC, Ayyanan A, Gutierrez M, Delaloye JF, Biophysica Acta (BBA): Molecular Basis of Disease 1864 3605–3617. Raffoul W, Fiche M, Dougall W, Schneider P, et al. 2013 (https://doi.org/10.1016/j.bbadis.2018.08.024) Progesterone/RANKL is a major regulatory axis in the human breast. Yang L, Ravindranathan P, Ramanan M, Kapur P, Hammes SR, Hsieh JT Science Translational Medicine 5 182ra55. (https://doi.org/10.1126/ & Raj GV 2012 Central role for PELP1 in nonandrogenic activation scitranslmed.3005654) of the androgen receptor in prostate cancer. Molecular Endocrinology Tirpe AA, Gulei D, Ciortea SM, Crivii C & Berindan-Neagoe I 2019 26 550–561. (https://doi.org/10.1210/me.2011-1101) Hypoxia: overview on hypoxia-mediated mechanisms with a focus Zhang Y, Beck CA, Poletti A, Clement JPt, Prendergast P, Yip TT, on the role of HIF genes. International Journal of Molecular Sciences 20 Hutchens TW, Edwards DP & Weigel NL 1997 Phosphorylation of E6140. (https://doi.org/10.3390/ijms20246140) human progesterone receptor by cyclin-dependent kinase 2 on three Truong TH & Lange CA 2018 Deciphering steroid receptor crosstalk in sites that are authentic basal phosphorylation sites in vivo. hormone-driven cancers. Endocrinology 159 3897–3907. (https://doi. Molecular Endocrinology 11 823–832. (https://doi.org/10.1210/ org/10.1210/en.2018-00831) mend.11.6.0006)

Received in final form 16 March 2020 Accepted 25 March 2020 Accepted Manuscript published online 25 March 2020

https://jme.bioscientifica.com © 2020 Society for Endocrinology https://doi.org/10.1530/JME-19-0274 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:21:57AM via free access