a reappraisal of progesterone action in the … 00 mammary gland...journal of mammary gland biology...

14
Journal of Mammary Gland Biology and Neoplasia, Vol. 5, No. 3, 2000 A Reappraisal of Progesterone Action in the Mammary Gland John P. Lydon 1 , Lakshmi Sivaraman 1 , and Orla M. Conneely 1,2 The ovarian hormones estrogen and progesterone and their respective receptors are essential for maintenance of postnatal developmental plasticity of the mammary gland and play a key role in mammary tumorigenesis. Mouse models in which expression of the progesterone receptors was genetically ablated have recently become available. Studies of these models have demonstrated that progesterone is specifically required for pregnancy associated ductal proliferation and lobuloalveolar differentiation of the mammary epithelium, but not for immediate postpubertal ductal morphogenesis. Use of these mice in combination with mam- mary gland transplantation indicates that developmental regulation by progesterone appears to occur through a paracrine mechanism in which progesterone receptor (PR) 3 positive cells represent a subset of non-proliferating epithelial cells that are capable of directing proliferation and/or differentiation of neighboring receptor negative cells. The hierarchical organization of these receptors in the epithelium and their segregation from proliferating cells is a conserved feature in rodent and human mammary tissue. The identification of paracrine mediators of the progesterone response is now an imminent goal as is the delinea- tion of the individual contributions of the two PR isoforms using similar approaches. KEY WORDS: Progesterone; mammary gland; PRKO mouse; epithelial-stromal; PR-A and -B. INTRODUCTION The evolution and propagation of the eutherian mammal has depended on the ovarian steroid hor- mone: progesterone. Although the role of progester- one in uterine biology is well recognized, its involve- ment in mammary gland development and function has been less well understood. However, as described in the ensuing sections, recent studies using rodent models amenable to genetic manipulation together with mammary gland transplantation approaches have begun to unravel the specific developmental role of progesterone in the mammary gland, and have 1 Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030. 2 To whom correspondence should be addressed at E-mail: [email protected] 3 Abbreviations: Progesterone receptor (PR); Progesterone and estrogen receptor knockout (PRKO and ERKO respectively); Terminal end bud (TEB). 325 1083-3021/00/0700-0325$18.00/0 2000 Plenum Publishing Corporation exposed the progesterone-signaling pathway to mo- lecular dissection. The physiological effects of progesterone are mediated by interaction of the hormone with specific intracellular progesterone receptors (PRs) that are members of the nuclear receptor family of transcrip- tion factors (1,2). Progesterone receptors consist of two protein isoforms, termed A and B, that are ex- pressed from a single gene in rodents and humans (3,4). The A and B proteins are produced by initiation of translation at two distinct AUG signals and differ by an amino terminal extension of 128–165 amino acids (depending on species) that is specific to the B protein [the mouse PR (5) is shown in Fig. 1, panel A]. Binding of progesterone to its receptors induces receptor dimerization, binding to specific cis-acting DNA elements in the promoter region of specific target genes and recruitment of coactivator proteins and general transcription factors to regulate tran- scription of responsive target genes [reviewed in (1)]. A significant body of evidence has accumulated in

Upload: ngobao

Post on 13-Jun-2018

227 views

Category:

Documents


0 download

TRANSCRIPT

Journal of Mammary Gland Biology and Neoplasia, Vol. 5, No. 3, 2000

A Reappraisal of Progesterone Action in theMammary Gland

John P. Lydon1, Lakshmi Sivaraman1, and Orla M. Conneely1,2

The ovarian hormones estrogen and progesterone and their respective receptors are essentialfor maintenance of postnatal developmental plasticity of the mammary gland and play a keyrole in mammary tumorigenesis. Mouse models in which expression of the progesteronereceptors was genetically ablated have recently become available. Studies of these modelshave demonstrated that progesterone is specifically required for pregnancy associated ductalproliferation and lobuloalveolar differentiation of the mammary epithelium, but not forimmediate postpubertal ductal morphogenesis. Use of these mice in combination with mam-mary gland transplantation indicates that developmental regulation by progesterone appearsto occur through a paracrine mechanism in which progesterone receptor (PR)3 positivecells represent a subset of non-proliferating epithelial cells that are capable of directingproliferation and/or differentiation of neighboring receptor negative cells. The hierarchicalorganization of these receptors in the epithelium and their segregation from proliferatingcells is a conserved feature in rodent and human mammary tissue. The identification ofparacrine mediators of the progesterone response is now an imminent goal as is the delinea-tion of the individual contributions of the two PR isoforms using similar approaches.

KEY WORDS: Progesterone; mammary gland; PRKO mouse; epithelial-stromal; PR-A and -B.

INTRODUCTION

The evolution and propagation of the eutherianmammal has depended on the ovarian steroid hor-mone: progesterone. Although the role of progester-one in uterine biology is well recognized, its involve-ment in mammary gland development and functionhas been less well understood. However, as describedin the ensuing sections, recent studies using rodentmodels amenable to genetic manipulation togetherwith mammary gland transplantation approacheshave begun to unravel the specific developmentalrole of progesterone in the mammary gland, and have

1 Department of Molecular and Cellular Biology, Baylor Collegeof Medicine, One Baylor Plaza, Houston, Texas, 77030.

2 To whom correspondence should be addressed at E-mail:[email protected]

3 Abbreviations: Progesterone receptor (PR); Progesterone andestrogen receptor knockout (PRKO and ERKO respectively);Terminal end bud (TEB).

325

1083-3021/00/0700-0325$18.00/0 2000 Plenum Publishing Corporation

exposed the progesterone-signaling pathway to mo-lecular dissection.

The physiological effects of progesterone aremediated by interaction of the hormone with specificintracellular progesterone receptors (PRs) that aremembers of the nuclear receptor family of transcrip-tion factors (1,2). Progesterone receptors consist oftwo protein isoforms, termed A and B, that are ex-pressed from a single gene in rodents and humans(3,4). The A and B proteins are produced by initiationof translation at two distinct AUG signals and differby an amino terminal extension of 128–165 aminoacids (depending on species) that is specific to the Bprotein [the mouse PR (5) is shown in Fig. 1, panelA]. Binding of progesterone to its receptors inducesreceptor dimerization, binding to specific cis-actingDNA elements in the promoter region of specifictarget genes and recruitment of coactivator proteinsand general transcription factors to regulate tran-scription of responsive target genes [reviewed in (1)].A significant body of evidence has accumulated in

326 Lydon, Sivaraman, and Conneely

Fig. 1. The progesterone receptor gene encodes two receptor isoforms: PR-A and PR-B. Panel A. The progesteronereceptor contains a long N-terminus region (A/B), a short DNA binding domain (C) and hinge region (D), a ligandbinding domain (E) and a short C-terminus (F). The PR comprises two receptor isoforms, PR-A and PR-B. PR-B isstructurally identical to PR-A, except for an N-terminal extension that varies depending on the species. The mouse PRis depicted; numbers in parenthesis denote amino acid number; METB and META are the initiating methionines for PR-Band PR-A respectively. Panel B. Assuming coexpression of PR-B and -A within the same cell, the possible PR homo-and hetero-dimers that can occur are denoted. Each PR dimer type would be expected to elicit a different transactivationalresponse; PRE is an acronym for progesterone response element.

recent years demonstrating that while both the Aand B protein bind progesterone and the same DNAelements, they interact differently with some coacti-vators and have different promoter and cell specifictranscription activation properties [reviewed in (6)].These findings have significant implications with re-gard to the complexity of progesterone signaling in

vivo. If expressed in the same cells, the A and Bproteins can dimerize and bind to DNA as threespecies: A:A or B:B homodimers or A:B heterodim-ers (Fig. 1, panel B). The specific contribution of eachof these species to the regulatory effects of progester-one will depend on the differential transactivationproperties contributed to these complexes by the B

Progesterone Action in the Mammary Gland 327

specific domain. Based on the distinct transactivationproperties of PR-A and -B proteins observed in vitro,the relative expression of these isoforms in the mam-mary gland is likely to have a significant impact onthe functional responses to progesterone.

In this review, we will first summarize the essen-tial role of steroid hormones in maintaining postnataldevelopmental plasticity of the mammary gland. Wewill then describe the recent advances in our under-standing of the specific roles of progesterone in regu-lating mammary gland morphogenesis and tumori-genesis that have emerged through genetic ablationof PR expression in mice and mammary gland trans-plantation experiments. Finally, we will discuss howthe different functional activities of the PR-A and-B proteins observed in vitro are likely to contributeto the complexity of progesterone signaling withinthe gland.

OVARIAN STEROIDS AND MAMMARYGLAND DEVELOPMENT

Despite over fifty million years of divergent evo-lution between the human and rodent, mammarygland development in both mammals is remarkablysimilar; thus within this phylogenetic Class, the ro-dent model has been the experimental system ofchoice to define the general principles of mammarygland development. In particular, our recent abilityto genetically manipulate the mouse has providedunparalleled opportunities to investigate the molecu-lar and cellular mechanisms that underlie normalmammary gland development as well as mammarytumorigenesis, in an in vivo context.

For all eutherian mammals, mammary gland de-velopment occurs predominantly postnatally, and isdirected by a complex signal transduction interplaybetween endocrine hormones and locally actinggrowth factors [reviewed in (7,8)]. The adult mam-mary gland consists primarily of a secretory epithe-lium consisting of a branching ductal architectureembedded in an adipose stromal compartment (alsoknown as the fat pad) that provides an inductiveenvironment (from the embryonic stage to the adult)in which the epithelium develops and functions.Mammary gland morphogenesis occurs in distinct de-velopmental stages depending on the age and repro-ductive state of the animal. In the prepubertal mouse,as with the human, the mammary gland contains arudimentary primary epithelial duct with limited sidebranching that extends a short distance from the nip-

ple into the mammary fat pad. Following puberty,cells localized in the bulbous shaped terminal endbuds (TEB) at the distal ends of the mammary ductsundergo extensive mitotic activity that results in boththe elongation and bifurcation of mammary ducts tothe periphery of the fat pad. Once the ductal networkextends to the limits of the mammary fat pad, theTEB regress and mammary epithelial ductal growthis arrested until pregnancy. The second round ofmammary epithelial proliferation occurs in responseto the hormones of pregnancy, and subsequentepithelial differentiation is manifested by the devel-opment of secretory alveolar structures that progres-sively occupy the remaining fat pad during preg-nancy, parturition and the onset of lactation.Following removal of the suckling stimulus at wean-ing, milk protein-secreting alveoli undergo apoptic-mediated reductive remodeling termed involution,and by four weeks of this post lactational epithelialregression, the mammary gland developmental path-way is essentially complete.

The entire developmental program is controlledby the combined action of ovarian steroids and pep-tide hormones, such as prolactin [reviewed in (7,8)].Progesterone and estrogen are the principle steroidhormones involved in normal breast developmentand tumorigenesis (9). Early ductal outgrowth ob-served postpuberty is strongly controlled by the cyclicrise in ovarian estrogen. Deletion of this hormonein mice by a null mutation of the aromatase generesponsible for its synthesis [the aromatase knockout(ARKO) mouse, (10)] results in a rudimentary hypo-plastic epithelium that is typical of a prepubertalmammary phenotype. Null mutations of each of thetwo receptor genes [estrogen receptor-� (ER-�) andestrogen receptor-� (ER-�), (11,12)] known to medi-ate the genomic effects of estrogen have demon-strated that the ER-� receptor alone is both necessaryand sufficient to mediate the morphogenic effectsof estrogen on the mammary epithelium. Further,embryonic tissue recombination experiments usingwild type and ER-� knock-out (ERKO) mice suggestthat loss of ER-� in the embryonic stromal but notepithelial compartment results in inhibition of ductaloutgrowth and a rudimentary ductal structure thatlacks terminal end buds (13). This study suggests thatthe role of ER-� in early ductal proliferation andbranching morphogenesis may involve a paracrinesignaling system initiated by estrogen dependentstromal derived signals acting on epithelial cells topromote their proliferation. However, because thisinvestigation was limited to embryonic tissue, extrap-

328 Lydon, Sivaraman, and Conneely

olation of these suggestions on ER-� action to themammary gland of the pubescent and adult mousemust be tempered.

The studies summarized have underscored theessential role of both estrogen and ER-� in ductalproliferation and branching that occurs during pu-berty. The PR, however, is a downstream moleculartarget for ER action (discussed later), and as such,the ERKO and ARKO mouse models were unableto define the specific role of progesterone in thisorgan system. The adoption of a similar genetic ap-proach to specifically ablate expression of the PR inmice has allowed us to delineate the specific effectsof progesterone signaling on mammary gland devel-opment.

PROGESTERONE RECEPTORKNOCK-OUT MOUSE

Delineating the Role of Progesterone

To directly address the physiological importanceof PR function in the murine mammary gland as wellas gain insight into progesterone’s functional inter-relationship not only with estrogen, but also withprolactin and locally acting growth factors known toinfluence mammary gland development, a progester-one receptor knockout (PRKO) mouse model wasgenerated in which the functional activity of bothforms of the PR (PR-A and PR-B) were simultane-ously ablated through gene targeting techniques (14).

Initial mammary gland whole mount analysis re-vealed that the ductal architecture of the adult PRKOmammary gland was similar to that of the age-matched wild type virgin (Fig. 2; panels A and B).Importantly, the virgin PRKO mammary gland wasnot a phenocopy of the ERKO or ARKO mammarydefect, demonstrating the importance of estrogenrather than progesterone in mammary gland develop-ment that occurs between the stages of puberty andthe adult virgin.

However, in response to exogenous estrogen andprogesterone treatment, comparative whole mountanalysis revealed that the adult PRKO mammarygland failed to develop the typical pregnancy-associ-ated epithelial ductal morphogenesis that consists ofextensive side-branching with attendant interductallobuloalveolar development (Fig. 2; panels C–F).These initial gross morphological investigations, inaddition to recent molecular analysis (15), unequivo-cally demonstrated both a proliferative and a differ-

entiative involvement for progesterone and its recep-tor in this tissue. To address the argument that thissimple steroid treatment was insufficient to rescuethe PRKO mammary phenotype, recent mammarygland transplantation experiments were performedin which the PRKO mammary gland was trans-planted in toto into a wild type mouse (16). In concor-dance with our initial observations, the PRKO mam-mary defect was not rescued during pregnancy,despite the exposure of the transplanted gland to thefull spectrum of pregnancy hormones.

Because epidemiological and experimental stud-ies have demonstrated that an early first pregnancylowers breast cancer risk whereas nulliparity or a latefirst pregnancy increases this risk (17,18), under-standing the cellular and molecular mechanisms bywhich progesterone induces mammary gland prolifer-ation and differentiation during pregnancy is a majorfocus of mammary gland research.

TOWARDS A CELLULAR MECHANISM OFACTION FOR PROGESTERONE IN THEMAMMARY GLAND

The PRKO mammary phenotype represents acritical in vivo validation of the importance of proges-terone in the induction of mammary gland epithelialproliferation and differentiation that is required forthe formation of ductal and alveolar structures duringpregnancy. In the embryo and the adult, normalmammary epithelial ontogenesis is dependent on areciprocal molecular dialogue between the epithelialand stromal cellular compartments (19). Similar tothe prostate and uterus (20,21), the existence of epi-thelial-stromal interactions in the mammary glandhas potentially important clinical implications for cer-tain disease states like breast cancer. The aberrantcellular proliferation and loss of steroid hormonalregulation that often occurs in mammary cancerscould conceivably be associated with a change or aloss in normal regulatory interactions between mam-mary stromal and epithelial cells (20). Elucidatingthese reciprocal cellular interactions can be facili-tated by a clear understanding of the spatiotemporalexpression of PRs in the mammary gland and by thegland’s intrinsic ability to recapitulate all the ductaland alveolar structures from a fragment of primaryductal epithelium transplanted into a isogenic stroma:the mammary gland transplantation technique (22).

Two different approaches to study the spatio-temporal expression of PR in the mammary gland

Progesterone Action in the Mammary Gland 329

Fig. 2. Ablation of progesterone receptor function manifests as a defect in mammary gland ductalbranching and alveologenesis. Adult virgin wild type (panel A) and PRKO (panel B) mammary glandsshow a similar ductal morphology; scale bar in panel A is 5 mm. The PRKO mammary phenotype isclearly evident when wild type (panel C) and PRKO (panel D) are treated for three weeks withestrogen and progesterone. Note the absence of extensive side branching and alveologenesis in thehormone-treated PRKO (panel D) as compared to wild type (panel C). Panels E and F are highermagnifications of C and D respectively; scale bar in panel E is 500 �m.

have provided conflicting results: Early binding stud-ies using R5020 demonstrated progestin binding sitesin both the epithelial and stromal compartments thatwere distinct with respect to their biochemistry andontogenesis (23,24). However, more recent immuno-histochemical analyses support an exclusively epithe-lial distribution of receptors that are undetectable inthe stromal compartment (25–28). The discrepancy

in PR localization observed using these two tech-niques could be attributed to either poor specificityof the R5020 binding method or to insufficient sensi-tivity of the anti-PR antibodies. However, recentanalysis of PR expression in newly generated PRknock-in mice using Lac-Z as a reporter gene demon-strate an exclusively luminal epithelial localizationfor PR that is in complete agreement with data

330 Lydon, Sivaraman, and Conneely

observed from immunohistochemical studies (J. P.Lydon, unpublished observations). The epithelial lo-calization of PR in the mammary gland is surprisingin light of its regulation by estrogen and the previousdemonstration that ER-� is localized to both the stro-mal and epithelial compartments of the gland (13).Taken together, these observations suggest that theregulation of PR by estrogen may be compartmentspecific in the mammary gland.

Most importantly, support for the functionalinvolvement of epithelial rather than stromal derivedPRs in mediating progestin dependent mammarymorphogenesis has been obtained through the useof the PRKO mouse in combination with mammarygland transplantation approaches to produce mam-mary gland recombinants that were devoid of PR ineither the epithelial, stromal, or both compart-ments (16).

To determine whether PR mediated responseswithin the mammary stroma contribute to ductal sidebranching and alveologenesis, wild type epitheliumwas transplanted into the PRKO stroma, previouslydivested of PRKO epithelium. Following the preg-nancy of the host animal, whole mount analysis re-vealed that this mammary tissue recombinant devel-oped normally, suggesting mammary stroma was notthe primary target for progesterone-initiated prolif-erative and differentiative responses in the mammarygland (Fig. 3; panels A and B).

The converse experiment was performed inwhich PRKO epithelium was transplanted into wildtype stroma. Despite a full-term pregnancy, wholemount analysis showed that the absence of PR in theepithelium resulted in the typical PRKO mammaryphenotype (Fig. 3; panels C and D). This result un-equivocally demonstrated that the epithelial cellularcompartment is the primary target for progesterone

Fig. 3. The mammary epithelial cellular component is the primary target for progesterone receptor action. In the PRKO stroma, wildtype epithelium can undergo normal epithelial proliferation and differentiation in response to pregnancy hormones (panel A). Panel Bis a positive control in which wild type epithelium is transplanted into wild type stroma. Wild type mice consisted of the ROSA 26(�-galactosidase�) mouse line. Transplantation of PRKO epithelium into wild type stroma (panel C) could not rescue the PRKO de-fect despite exposure to pregnancy hormones. Panel D is a positive control in which wild type epithelium is transplanted into wildtype stroma; note the pregnancy-induced ductal branching and alveologenesis. The mere coexistence of fragments of wild type andPRKO epithelium within the same wild type stroma could not rescue the PRKO phenotype following pregnancy (panels E and F).The wild type epithelium is blue (ROSA 26 (�-galactosidase�)) whereas the PRKO epithelium is red; scale bars in panels E andF are 2 mm and 200 �m respectively. Following pregnancy, PRKO mammary epithelial cells (ROSA 26 (�-galactosidase�)) cancontribute to alveologenesis when mixed with wild type epithelial cells; panel G (scale bar: 200�m). This result suggests that (throughparacrine effects) PR positive cells can affect neighboring PR negative (PRKO) cells to contribute to alveologenesis and ductal sidebranching. Adapted from Brisken et al. (16).

action in the mammary gland. Recent reciprocalmammary gland transplantation approaches appliedto the CAAT enhancer binding protein beta (CEBP-�), cyclin D1 and prolactin receptor knockout mousemodels have revealed that the mammary epitheliumis also the primary target for these important regula-tory proteins (29–32). In apparent contrast to theseobservations, mammary transplantation experimentsusing the ERKO mouse have demonstrated that thestromal rather than epithelial derived ER populationis necessary for ductal proliferation in the neonatalgland (13); however, the adult gland was not exam-ined in this study.

These PRKO mammary gland transplantationexperiments provided a unique insight into the func-tional exclusivity of epithelial-derived PRs in preg-nancy-associated mammary morphogenesis. How-ever, these investigations did not address the questionof whether all epithelial cells are required to expressthe PR as a prerequisite for normal ductal side-branching and alveologenesis. This question wasprompted by data emerging from immunohistochem-ical experiments that revealed a nonuniform expres-sion pattern for PR in the luminal epithelial cellularcompartment of the mammary gland of the adultmurine virgin (25–28).

To answer this question, mammary epithelialcells derived from the PRKO mouse were mixed withequivalent cells from the wild type (16). To differenti-ate between wild type and PRKO epithelial contribu-tions, the wild type or the PRKO was back-crossedto the ROSA26 mouse in which the lacZ gene isexpressed in all epithelial cells; ROSA26 derived cellsstain blue with X-gal, a chromogenic substrate for �-galactosidase. The resultant chimeric epithelial cellmixture was injected into a cleared stromal compart-ment of the wild type gland followed by transplanta-

332 Lydon, Sivaraman, and Conneely

tion of the recombinant gland to a wild type host.Following pregnancy, whole mount and immunohis-tochemical analysis revealed that PRKO epithelialcells could contribute to alveolar structures (Fig. 3;panel G), and functioned normally as judged by theexpression of the milk protein: �-casein. Thus, PRKOderived epithelial cells can contribute to both theproliferative and differentiative responses to proges-terone when placed in close apposition with PR posi-tive cells.

The following conclusions can be drawn fromthese observations. First, at least two distinct luminalepithelial cell lineages exist in the mammary gland:cells that score positive or negative for PR expression.Indeed, apart from the adult mouse, mammary epi-thelial cells, expressing PR, have been identified inthe embryonic and prepubertal stages of develop-ment, suggesting that the fate of these cells may bespecified early for specialized functions later in theadult (25) and (J. P. Lydon, unpublished observa-tions); these functions may consist of initiating alveo-logenesis and ductal side branching. This proposal issupported by the fact that although PR expressionoccurs early in mammary development, loss of PRfunction as in the PRKO does not impact normalmammary morphogenesis until pregnancy. Second,because only PR negative cells in close associationwith PR positive cells can contribute to ductal sidebranching and alveologenesis (16), paracrine-signal-ing pathways between these cell types is hypothesizedto occur. Third, although lacking PR-positive cells,the PRKO mammary gland still retains those PRnegative cells that are responsive to PR-mediatedparacrine signaling, again suggestive of distinct epi-thelial cell populations that have evolved separatelyto either express PR or to be receptive to paracrinesignals induced by PR.

These conclusions have been recently substanti-ated and further extended by studies on human andrat mammary tissue that revealed a segregation be-tween those epithelial cells expressing both ER andPR and those cells undergoing proliferation (33–35),and Fig. 4, panel A. Close scrutiny of the immunohis-tochemical results revealed that the majority of pro-liferating cells were distinct from, but in close apposi-tion to steroid receptor positive cells, again hinting ata paracrine action between steroid receptor positive‘sensor’ cells and a sub-population of steroid receptornegative cells, but mitotically competent, ‘effector’cells. These observations do not discount the possibil-ity that steroid receptor positive cells may eventuallycommit to cell division with attendant loss of steroid

receptor expression (temporal segregation); the re-verse scenario could also be contemplated.

The fact that this spatial organization for ERand PR positive cells has been conserved betweenthe human and rodent mammary gland strongly sup-ports an evolutionarily conserved cellular mechanismof action by which cells expressing these receptorsimpact the functional activity of neighboring cells toinduce ductal morphogenesis. How and why such acellular patterning evolved in the mammary epithe-lium is currently a matter of conjecture; however,perturbations of such an important cellular arrange-ment would be predicted to have adverse conse-quences for normal mammary gland development.Indeed, it has been reported that in many breasttumors, the majority of ER and PR expressing cellsalso undergo proliferation (33), clearly at odds withthe earlier paracrine signaling pathways that are op-erative in the normal gland. Further, they suggestthat the development of breast cancer may involvea switch in steroid dependent regulation of prolifera-tion from a paracrine to intracrine/autocrine mecha-nism. The possible modes of PR action within theluminal epithelial cell are summarized in Fig. 4,panel C.

Based on these studies, it will be interesting todetermine whether a nonuniform spatial organizationis adopted by mammary epithelial cells that expressother important modulators of mammary morpho-genesis and function. Indeed, the steroid receptorcoactivator-1 (SRC-1), a coactivator for certain mem-bers of the nuclear receptor superfamily (36), wasshown to be expressed in mammary epithelial cellsin a nonuniform spatial arrangement; these cells,however, were distinct from those cells expressingER and PR (37). Considering that SRC-1 has beenshown to be a coactivator for ER and PR, this resultwas surprising, and suggests that at least in the normalmammary gland, ER and PR do not require thiscoactivator to exert their effects. Recent character-ization of the SRC-1 knockout mouse revealed amammary defect (38), indicating that SRC-1 mayeither interact with other nuclear receptors and/orparticipate in novel regulatory pathways to elicitmammary epithelial ductal branching and alveolo-genesis.

Although mammary gland transplantation andimmunohistochemical experiments have uncoveredan unanticipated mechanism of action for PR in themammary gland, these investigations have also raisedquestions for future studies. First, do mammary epi-thelial cells that express the PR comprise a stem cell

Fig. 4. In the normal mammary gland, the majority of epithelial cells that express PR are segregated from proliferating cells.Panel A: epithelial cells within the mammary gland of a 45 day old rat that express PR and/or incorporate 5-bromo-2-deoxyuridine(BrdU) were detected by indirect immunofluorescence; cells in S-phase of the cell cycle incorporate BrdU. Epithelial cellsexpressing PR are red (secondary antibody: Texas Red conjugated goat anti-rabbit antibody) whereas cells incorporating BrdUare green (anti-BrdU antibody, conjugated with fluorescein-isothiocyanate (FITC)). Proliferating cells that also express PR areshown in yellow (arrow). Panel B: in parallel, all nuclei in this field were detected with 6-diamidino-2-phenylindole (DAPI),and appear blue. Magnification is 400X. Panel C: possible pathways by which progesterone-initiated signaling can influence PRnegative mammary epithelial cells to proliferate. Pathway 1 denotes a paracrine-signaling pathway through which PR positiveepithelial cells communicate with juxtaposed PR negative epithelial cells; this pathway predicts the existence of paracrine factors(possibly secretory) that deliver the progesterone signal to neighboring cells. Pathway 2 is similar to 1 except that the stromalcellular compartment is a necessary mediator of the progesterone paracrine signal. Pathway 3 represents an autocrine or intracrinepathway by which the progesterone signal would be directed back to the cell of origin to elicit proliferation. This scenario mightoccur in a minority of cells in the normal gland (perhaps a preneoplastic cell population) (panel A (arrow)) and/or in themajority of PR positive cells in breast cancer (33).

334 Lydon, Sivaraman, and Conneely

class? This question may be answered, in part, byexamining these cells using electronmicroscopy todetermine whether this cell type exhibits the cellularcharacteristics for stem cells that have recently beendescribed using this technique (39). If mammary epi-thelial cells expressing PR were identified as stemcells, the spatial organization of these cells (affector/sensor cells) and their close juxtaposition to prolifer-ating cells (effector cells) would draw parallels to asimilar cellular organization that has been reportedfor other self renewing tissues, such as the epidermisand hair follicle (40–42).

Considering the evolutionary importance of thiscellular organization for epithelial cells that expressPR in the adult virgin gland, how this cellular organi-zation is reinstated following pregnancy, lactation,and involution will be a challenging question for thefuture. A further question will be to address the issueof whether embryonic, pre- and pubertal stages ofmammary gland development also manifest the non-uniform organization of PR-expressing epithelialcells as observed in the adult virgin. If this cellularorganization is shown to be exclusive to the adultvirgin, it may suggest that at the onset of pregnancy,this cellular arrangement has specifically evolvedto ensure that extensive side branching and atten-dant alveologenesis occurs in a spatially orderedmanner.

Finally, a key question to understanding PRsmolecular mechanism of action in the mammarygland will be to identify the genes involved in theparacrine signaling pathway(s) between mammaryepithelial cells positive and negative for PR expres-sion. Recent elegant studies by Brisken and cowork-ers have provided compelling evidence that wnt-4may play a pivotal paracrine role in dispatching theprogesterone signal in the mouse mammary gland(43). Since the arrival of functional genomics, withthe availability of knockout mouse models, and anincreasing number of gene discovery approaches, weshould be confident that more of these paracrine-signaling molecules will be identified in the not toodistant future.

PROGESTERONE RECEPTOR’SINVOLVEMENT IN MAMMARYTUMORIGENESIS

In addition to its role in normal mammary glanddevelopment, the proposed involvement of proges-terone in mammary gland tumorigenesis has

formented much discussion. Epidemiological studieshave revealed a close correspondence between breastcancer risk and the exposure of the mammary glandto cyclical levels of ovarian sex steroids that occursduring the reproductive years of premenopause [re-viewed in (44)]. This correlation is further supportedby clinical studies in which inhibition of such steroidalexposure, for example after bilateral oophorectomy,markedly reduced breast cancer risk (45,46); an earlymenopause and a late menarche have also beenshown to exhibit similar beneficial effects (17,47).Based on these observations, the increase in breastcancer observed with advancing age is hypothesizedto result from ovarian sex steroid-induced prolifera-tion of the mammary epithelial cell which, through-out the reproductive years, provides a temporal win-dow of opportunity for the accumulation of geneticchanges resulting in breast cancer in later life. Be-cause a significant measure of breast cancer risk islinked with the cyclical exposure of the mammaryepithelial cell to ovarian sex steroids, breast cancerprevention treatments based on ablating ovarian ste-roidogenesis have been considered (17).

Based on this hypothesis, to investigate breastcancer etiology, without considering the involvementof progesterone, would be a failure in perspective.However, progress in our understanding of breastcancer in relation to progesterone exposure has beenstymied due to our inability to mechanistically dissectin vivo the individual and integrative roles of estrogenand progesterone in this cancer. The contentiousnessof the issue has been further exacerbated by the nu-merous conflicting reports concerning the influenceof synthetic progestins on mammary tumorigenesisin rodents (48–50). Nevertheless, a number of studieshave linked progesterone to the progression of cer-tain carcinogen induced and transplantable rat mam-mary tumors, these studies also suggest that proges-terone may play a role in spontaneous tumors of themurine mammary gland (50–55). However, proges-terone can be both stimulatory and inhibitory de-pending on the time of progesterone exposure inrelation to carcinogen treatment suggesting a degreeof caution in ascribing a role for progesterone inmammary gland tumorigenesis.

Studies with cultured human breast cancer cellshave shown that depending on the dosage and dura-tion of treatment, progesterone can exert bothgrowth stimulatory and inhibitory effects [reviewedin (56)]. Because recent in vitro studies have identi-fied a cross-communication pathway between proges-terone and growth factor/cytokine family members

Progesterone Action in the Mammary Gland 335

(57), it has been hypothesized that, following oneround of proliferation, the initial pulse of progester-one acts as a primer for the actions for secondaryfactors involved in either proliferative, differentia-tive, or apoptotic pathways. The commitment to anyone of these subsequent pathways would be influ-enced by the type of cross-talk between progesteroneand growth/cytokine signaling pathways which inturn would be determined by the dosage and durationof subsequent progesterone exposures.

Recently, the PRKO mouse in combination withthe chemical carcinogen-treated mammary tumormodel was used to evaluate the functional relevanceof progesterone-initiated intracellular signaling inmammary gland tumorigenesis (27). Carcinogen-treated PRKO mice exhibited a significant reductionin mammary tumor incidence as compared to wildtypes; mammary tumors arose in 12 (60%) of 20 wildtype mice compared with 3 (15%) of 20 PRKO miceby 44 weeks after the initial carcinogen treatment.Despite the complexity of progesterone’s involve-ment in mammary tumorigenesis, these results under-scored the specific importance of the PR (as distinctfrom ER) as an obligate mediator for those intracel-lular signaling pathways that are essential for theinitiation of murine mammary tumors induced bychemical carcinogens.

Collectively, these studies have prompted a re-valuation in our understanding of progesterone’s par-ticipation in mammary tumor progression that mayhave consequences not only for the current use ofprogestins in contraception and postmenopausal hor-mone replacement, but also in the design of diagnos-tic approaches and/or therapies for the future treat-ment and prevention of breast cancer.

Delineating the Specific Role of the PR-A and -B

While the PRKO mouse provides a powerfultool to define the role of progesterone in mammaryepithelial cell proliferation, differentiation and tu-morigenesis, the specific contribution of the PR-Aand -B isoforms to these activities remains to be ad-dressed. As indicated in the introduction, significantevidence has accumulated indicating that the PR-Aand -B proteins are functionally distinct when exam-ined in vitro. First, when expressed individually intissue cultured cells, PR-A and -B display differenttransactivation properties that are specific to bothcell type and target gene promoter used [reviewedin (6)]. These findings suggest that A and B may

regulate different physiological target genes in re-sponse to progesterone and that each protein maydisplay different transactivation capabilities in differ-ent target tissues. Second, when the A and B proteinsare coexpressed in cultured cells in cell and promotercontexts in which agonist bound PR-A is inactive,the protein can act as a dominant repressor of PR-B activity (58,59). These findings suggest that PR-A has the ability to diminish overall progesteroneresponsiveness of specific target genes in specific tis-sues. This repressor capability, which appears to bea selective property of the A protein, extends notonly to PR-B but also to other steroid receptors.Thus, PR-A has been shown to inhibit estrogen, glu-cocorticoid and mineralocorticoid receptor depen-dent gene activation presumably through competi-tion for common limiting coactivators (60). Third,the A and B proteins also respond differently to pro-gestin antagonists [reviewed in (6)]. While antagonistbound PR-A is inactive, antagonist bound PR-B canbe converted to a strongly active transcription factorby modulating intracellular phosphorylation path-ways (61–63). Finally, transrepression of ER activityhas been observed in the presence of either proteinwhen bound to antagonists (59,60,64,65).

Both the PR-A and -B proteins are expressedin the mammary glands of rodents and humans. Inthe case of the mouse, the expression of PR-A pre-dominates over PR-B by a 2:1 ratio in the virgin glandand throughout pregnancy (32,66). In normal humanbreast tissue, the ratio of PR-A and -B is equimolarwhile this ratio is significantly altered in some breasttumors (67). It is unclear at the present time whetheralterations in the ratio of PR-A to PR-B can contrib-ute to an altered susceptibility of these cells to carci-nogenesis. Conceivably, such alterations could havea dramatic effect on the cellular response to proges-terone agonists and antagonists as well as the growthfactors and proto-oncogenes regulated by proges-terone.

Most importantly, recent reports have providedthe first in vivo demonstration that disruption ofPRA/B ratios by overexpression of either the PR-Aor PR-B protein in the mammary gland of transgenicmice results in impaired mammary gland develop-ment. As a consequence of PR-A overexpression,mammary glands exhibited increased ductalbranching and hyperplasia and most interestingly,an abnormal disruption of the organization of thebasement membrane and decreased cell-cell adhe-sion (68). Interestingly, PR-B overexpression re-sulted in limited ductal elongation and branching;

336 Lydon, Sivaraman, and Conneely

alveolar growth was unaffected (69). These findingsprovide strong evidence that a regulated expressionof these receptor isoforms is critical for the mammarygland to respond appropriately to progesterone.However, in light of our recent studies and those ofothers, the mammary defects observed in PR-A and-B transgenic mice could also be explained by inap-propriate targeting of PR-A and -B expression toepithelial subtypes that normally would not expressPR, but may be competent to proliferate. Thus, theindiscriminate targeting of these receptor isoformsto the mammary gland would breach the cellular seg-regation rules that apply to normal epithelial cellgrowth, resulting in a scenario reminiscent of theinappropriate co-localization of steroid receptor ex-pression and proliferation observed in cells ofbreast tumors.

CONCLUSIONS

Studies during the past five years have led tosignificant advances in our understanding of thespecific role of progesterone in the mammary gland.The combined use of improved detection methods(PR specific antibodies) to detect PRs in situ, geneticmanipulation of PR expression in mice and classicalmammary gland transplantation technologies havebegun to expose the progesterone signaling pathwayto molecular dissection. Together, these approacheshave defined a specific role for progesterone inpregnancy associated ductal epithelial proliferationand lobuloalveolar differentiation within the normalmammary gland as well as tumorigenesis in responseto carcinogen challenge. Further, they have demon-strated that PRs are expressed in the adult mam-mary gland in a nonuniform subset of epithelialcells, most of which are nonproliferative. Thesereceptors appear to regulate both epithelial cellproliferation and differentiation in the normal glandby a paracrine mechanism in which proliferationand differentiation of PR negative cells is controlledby paracrine factors released from neighboring PRpositive cells. The hierarchical organization of PRswithin epithelial cells appears to be a key conservedfeature of rodent and human mammary epithelialcells that may underscore normal progesterone de-pendent regulation of pregnancy associated develop-mental plasticity of the gland. The observation thata high number of human breast tumors show adirect association between PR and ER expressionand proliferation suggests that disruption of the

normal paracrine relationship between receptor pos-itive and proliferating cells may contribute to abnor-mal steroid dependent signaling and may be animportant feature of tumorigenesis. A key objectivein future studies will be to gain insight into howthe cellular pattern of PR positive cells is specifiedand maintained during mammary gland develop-ment and to identify the paracrine effectors ofprogesterone dependent epithelial morphogenesis.

Finally, despite a wealth of in vitro evidencesupporting a distinct role of the PR-A and -Bproteins in mediating transcriptional responses toprogesterone, our knowledge of the individual con-tributions of each of these isoforms to progesteronephysiology is still in its infancy. We have recentlybegun to address this question by producing asecond generation of PR knockout mice in whicheither the PR-A or -B isoforms have been selectivelyablated. Our initial analysis of these mice providesdefinitive evidence that the PR-A and -B proteinscontribute to the reproductive functions of proges-terone in a different tissue specific manner (manu-script in preparation). These models promise toprovide exciting new information on the selectivecontribution of PR-A and -B to mammary glanddevelopment as well as to mammary tumorigenesis.Further, information derived from these analysesmay ultimately lead to novel tissue specific chemo-therapeutic approaches to the treatment ofbreast cancer.

ACKNOWLEDGMENTS

For the studies described herein, the authorsgreatly appreciate the technical expertise of Jie Li,Jeannie Zhong, Mary Ge, and Francis Kittrell. Wewould like to acknowledge our collaborators: Drs.Bert W. O’Malley, Franco J. DeMayo, Daniel Me-dina, Jeffery M. Rosen, Robert A. Weinberg, andCathrin Brisken. This work was supported in part byNIH grants CA-77530 awarded to John P. Lydon andHD-32007 to Orla M. Conneely.

REFERENCES

1. M.-J. Tsai and B. W. O’Malley (1994). Molecular mechanismsof action of steroid/thyroid receptor superfamily members.Ann. Rev. Biochem. 63:451–486.

2. D. J. Mangelsdorf, C. Thummel, M. Beato, et al. (1995). Thenuclear receptor superfamily: The second decade. Cell83:835–839.

Progesterone Action in the Mammary Gland 337

3. P. Kastner, A. Krust, B. Turcotte, et al. (1990). Two distinctestrogen-regulated promoters generate transcripts encodingthe two functionally different human progesterone receptorforms A and B. EMBO. J. 9:1603–1614.

4. W. L. Kraus, M. M. Montano, and B. S. Katzenellenbogen(1993). Cloning of the rat progesterone receptor gene 5�-regionand identification of two functionally distinct promoters. Mol.Endocrinol. 7:1603–1616.

5. D. R. Schott, G. Shyamala, W. Schneider, et al. (1991). Molecu-lar cloning, sequence analyses, and expression of complemen-tary DNA encoding murine progesterone receptor. Biochemis-try 30:7014–7020.

6. P. H. Giangrande and D. P. McDonnell (1999). The A and Bisoforms of the human progesterone receptor: Two function-ally different transcription factors encoded by a single gene.Recent Prog. Horm. Res. 54:291–313.

7. Y. J. Topper and C. S. Freeman (1980). Multiple hormoneinteractions in the developmental biology of the mammarygland. Physiol. Rev. 60:1049–1056.

8. L. Hennighausen and G. W. Robinson (1998). Think globally,act locally: The making of a mouse mammary gland. GenesDev. 12:449–455.

9. C. W. Daniel and G. B. Silberstein (1987). Postnatal develop-ment of the rodent mammary gland. In M. C. Neville and C. W.Daniel (eds.), The Mammary Gland: Development, Regulationand Function, Plenum Press, New York, pp. 3–36.

10. C. R. Fisher, K. H. Graves, A. F. Parlow, et al. (1998). Charac-terization of mice deficient in aromatase (ArKO) because oftargeted disruption of the cyp19 gene. Proc. Natl. Acad. Sci.U.S.A. 95:6965–6970.

11. J. H. Krege, J. B. Hodgin, J. F. Couse, et al. (1998). Generationand reproductive phenotypes of mice lacking estrogen receptorbeta. Proc. Natl. Acad. Sci. U.S.A. 95:15677–15682.

12. J. F. Couse and K. S. Korach (1999). Estrogen receptor nullmice: What have we learned and where will they lead us?Endocrine Rev. 20:358–417.

13. G. R. Cunha, P. Young, Y. K. Hom, et al. (1997). Elucidationof a role for stromal steroid hormone receptors in mammarygland growth and development using tissue recombinants.J. Mam. Gland Biol. Neoplasia 2:393–402.

14. J. P. Lydon, F. J. DeMayo, C. R. Funk, et al. (1995). Micelacking progesterone receptors exhibit pleiotropic reproduc-tive abnormalities. Genes Dev. 9:2266–2278.

15. T. K. Said, O. M. Conneely, D. Medina, et al. (1997). Progester-one, in addition to estrogen, induces cyclin D1 expression inthe murine mammary epithelial cell, in vivo. Endocrinology138:3933–3939.

16. C. Brisken, S. Park, T. Vass, et al. (1998). A paracrine rolefor the epithelial progesterone receptor in mammary glanddevelopment. Proc. Natl. Acad. Sci. U.S.A. 95:5076–5081.

17. B. E. Henderson, R. K. Ross, and M. C. Pike (1993). Hormonalchemoprevention of cancer in women. Science 259:633–638.

18. J. Russo and I. H. Russo (1995). The etiopathogenesis of breastcancer prevention. Cancer Lett. 90:81–89.

19. G. R. Cunha and Y. K. Yom (1996). Role of mesenchymal-epithelial interactions in mammary gland development.J. Mam. Gland Bol. Neoplasia 1:5–19.

20. G. R. Cunha (1994). Role of mesenchymal-epithelial interac-tions in normal and abnormal development of the mammarygland and prostate. Cancer 74:1030–1044.

21. T. Kurita, P. Young, J. R. Brody, et al. (1999). Stromal proges-terone receptors mediate the inhibitory effects of progesterone

on estrogen-induced uterine epithelial cell deoxyribonucleicacid synthesis. Endocrinology 139:4708–4713.

22. K. B. DeOme, L. J. Faulkin, A. Bern, et al. (1958). Develop-ment of mammary tumors from hyperplastic alveolar nodulestransplanted into gland-free mammary pads of female C3Hmice. Cancer Res. 19:515–519.

23. S. Z. Haslam and G. Shyamala (1981). Relative distributionof estrogen and progesterone receptors among the epithelial,adipose, and connective tissue components of the normalmammary gland. Endocrinology 108:825–830.

24. S. Z. Haslam (1988). Acquisition of estrogen-dependent pro-gesterone receptors by normal mouse mammary gland. Ontog-eny of mammary progesterone receptors. J. Steroid Bio-chem. 31:9–13.

25. G. B. Silberstein, K. Van Horn, G. Shyamala, et al. (1996).Progesterone receptors in the mouse mammary duct: Distribu-tion and developmental regulation. Cell Growth Differ.7:945–952.

26. G. Shyamala, M. H. Barcellos-Hoff, D. Toft, et al. (1997). Insitu localization of progesterone receptors in normal mousemammary glands: Absence of receptors in the connective andadipose stroma and a heterogeneous distribution in the epithe-lium. J. Steroid Biochem. Mol. Biol. 63:251–259.

27. J. P. Lydon, G. Ge, F. S. Kittrell, et al. (1999). Murine mammarygland carcinogenesis is critically dependent on progesteronereceptor function. Cancer Res. 59:4276–4284.

28. T. N. Seagroves, J. P. Lydon, R. C. Hovey, et al. (2000).C/EBP� (CCAAT/enhancer binding protein) controls cellfate determination during mammary gland development. Mol.Endocrinol. 14:359–368.

29. G. Robinson, P. F. Johnson, L. Hennighausen, et al. (1998).The C/EBP� transcription factor regulates epithelial cell pro-liferation and differentiation in the mammary gland. GenesDev. 12:1907–1916.

30. T. N. Seagroves, S. Krnacik, B. Raught, et al. (1998). C/EBP�,but not C/EBP�, is essential for ductal morphogenesis, lobu-loalveolar proliferation, and functional differentiation in themouse mammary gland. Genes Dev. 12:1917–1928.

31. C. Brisken, S. Kaur, T. E. Chavarria, et al. (1999). Prolactincontrols mammary gland development via direct and indirectmechanisms. Dev. Biol. 210:96–106.

32. V. Fantl, P. A. Edwards, J. H. Steel, et al. (1999). Impairedmammary gland development in cyl-1�/� mice during preg-nancy and lactation is epithelial cell autonomous. Dev. Biol.212:1–11.

33. R. B. Clarke, A. Howell, C. S. Potten, et al. (1997). Dissociationbetween steroid receptor expression and cell proliferation inthe human breast. Cancer Res. 57:4987–4991.

34. E. Anderson, R. B. Clarke, and A. Howell (1997). Changesin the normal human breast throughout the menstrual cycle:Relevance to breast carcinogenesis. Endocrine Related Can-cer 4:23–33.

35. J. Russo, X. Ao, C. Grill, et al. (1999). Pattern of distributionof cells positive for estrogen receptor-� and progesterone re-ceptor in relation to proliferating cells in the mammary gland.Breast Cancer Res. Treat. 53:217–227.

36. S. A. Onate, S. Y. Tsai, M. J. Tsai, et al. (1995). Sequenceand characterization of a coactivator for the steroid hormonereceptor superfamily. Science 270:1354–1357.

37. W.-S. Shim, J. DiRenzo, J. A. DeCaprio, et al. (1999). Segrega-tion of steroid receptor coactivator-1 from steroid receptors

338 Lydon, Sivaraman, and Conneely

in mammary epithelium. Proc. Natl. Acad. Sci. U.S.A.96:208–213.

38. J. Xu, Y. Qiu, F. J. DeMayo, et al. (1998). Partial hormoneresistance in mice with disruption of the steroid receptor co-activator-1 (SRC-1) gene. Science 279:1922–1925.

39. G. Chepko and G. H. Smith (1999). Mammary epithelial stemcells: Our current understanding. J. Mam. Gland Biol. Neopla-sia 4:35–52.

40. P. H. Jones (1997). Epithelial stem cells. Bioessays 19:683–690.41. S. M. Jankovic and S. V. Jankovic (1998). The control of hair

growth. Dermatol. Online J. 4:2.42. F. M. Watt (1998). Epidermal stem cells: Markers, patterning

and the control of stem cell fate. Philos. Trans. R. Soc. Lond.B. Biol. Sci. 353:831–837.

43. C. Brisken, A. Heineman, T. Chavarria, et al. (2000) Essentialfunction of Wnt-4 in mammary gland development down-stream of progesterone signaling. Genes Dev. 14:650–654.

44. I. H. Russo and J. Russo (1998). Role of hormones in mammarycancer initiation and progression. J. Mam. Gland Biol. Neopla-sia 3:49–61.

45. T. Hirayama and E. L. Wynder (1962). A study of epidemiol-ogy of cancer of the breast. II. The influence of hysterectomy.Cancer 15:28–38.

46. M. Feinleib (1968). Breast cancer and artificial menopause. J.Natl. Cancer Inst. 41:315–329.

47. D. Trichopoulos, B. MacMahon, and P. Cole (1972). Meno-pause and breast cancer. J. Natl. Cancer Inst. 48:605–613.

48. P. Pazos, C. Lanari, R. Meiss, et al. (1991). Mammary carcino-genesis induced by N-methyl-N-nitrosourea (MNU) andmedroxyprogesterone acetate (MPA) in Balb/c mice. BreastCancer Res. Treat. 20:133–138.

49. S. Li, C. Leveesque, C.-S. Geng, et al. (1995). Inhibitory effectsof medroxyprogesterone acetate (MPA) and the pure anties-trogen EM-219 on estrone (E1)-stimulated growth of dimeth-ylbenz(a)anthracene (DMBA)-induced mammary carcinomain the rat. Breast Cancer Res. Treat. 34:147–159.

50. C. M. Aldaz, Q. Y. Liao, M. La Bate, et al. (1996). Medroxy-progesterone acetate accelerates the development and in-creases the incidence of mouse mammary tumors induced bydimethylbenzanthracene. Carcinogenesis 17:2069–2072.

51. J. W. Jull (1954). The effects of oestrogens and progesteroneon the chemical induction of mammary cancer in mice of theIF strain. J. Pathol. Bact. 68:547–559.

52. A. G. Jabara and A. G. Harcourt (1971). Effects of progester-one, ovariectomy and adrenalectomy on mammary tumorsinduced by 7, 12-dimethylbenz(a)anthracene in Sprague-Dawley rats. Pathology 3:209–214.

53. C. W. Welsch (1985). Host factors affecting the growth ofcarcinogen-induced rat mammary carcinomas: A review andtribute to Charles Brenton Huggins. Cancer Res. 45:3415–3443.

54. S. P. Robinson and V. C. Jordan (1987). Reversal of antitumoreffects of tamoxifen by progesterone in the 7, 12-dimethylben-zanthracene-induced rat mammary carcinoma model. CancerRes. 47:5386–5390.

55. I. H. Russo and J. Russo (1991). Progestagens and mammary

gland development: Differentiation versus carcinogenesis.Acta Endocrinol. 125:7–12.

56. C. A. Lange, J. K. Richer, and K. B. Horwitz (1999). Hypothe-sis: Progesterone primes breast cancer cells for cross-talk withproliferative or antiproliferative signals. Mol. Endocrinol.13:829–836.

57. J. K. Richer, C. A. Lange, N. G. Manning, et al. (1998). Conver-gence of progesterone with growth factor and cytokine signal-ing in breast cancer. J. Biol. Chem. 273:31317–31326.

58. E. Vegeto, M. M. Shahbaz, D. X. Wen, et al. (1993). Humanprogesterone receptor A form is a cell and promoter specificrepressor of human progesterone receptor B function. Mol.Endocrinol. 7:1244–1255.

59. W. L. Kraus, K. E. Weis, and B. S. Katzenellenbogen (1995).Inhibitory cross-talk between steroid hormone receptors: Dif-ferential targeting of estrogen receptor in the repression of itstranscriptional activity by agonist- and antagonist-occupiedprogestin receptors. Mol. Cell. Biol. 15:1847–1857.

60. D. X. Wen, Y.-F. Xu, D. E. Mais, et al. (1994). The A and Bisoforms of the human progesterone receptor operate throughdistinct signaling pathways within target cells. Mol. Cell.Biol. 14:8356–8364.

61. C. A. Sartorius, L. Tung, G. S. Takimoto, et al. (1993). Antago-nist-occupied human progesterone receptors bound to DNAare functionally switched to transcriptional agonists by cAMP.J. Biol. Chem. 268:9262–9266.

62. E. A. Musgrove, J. A. Hamiliton, C. S. Lee, et al. (1993).Growth factor, steroid, and steroid antagonist regulation ofcyclin gene expression associated with changes in T47D humanbreast cancer cell cycle progression. Mol. Cell. Biol. 13:3577–3587.

63. C. A. Sartorius, S. D. Groshong, L. A. Miller, et al. (1994).New T47D breast cancer cell lines for the independent studyof progesterone B- and A-receptors: Only antiprogestin-occu-pied B-receptors are switched to transcriptional agonists bycAMP. Cancer Res. 54:3868–3877.

64. D. P. McDonnell, and M. E. Goldman (1994). RU486 exertsantiestrogenic activities through a novel progesterone receptorA form-mediated mechanism. J. Biol. Chem. 269:11945–11949.

65. W. L. Kraus, K. Weis, and B. S. Katzenellenbogen (1997).Determinants for the repression of estrogen receptor transcrip-tional activity by ligand-occupied progestin receptors. J. Ste-roid Biochem. Mol. Biol. 63:175–188.

66. G. Shyamala, W. Schneider, and D. Schott (1990). Develop-mental regulation of murine mammary progesterone receptorgene expression. Endocrinology 126:2882–2889.

67. J. D. Graham, C. Yeates, R. L. Balleine, et al. (1996). Progester-one receptor A and B protein expression in human breastcancer. J. Steroid Biochem. Mol. Biol. 56:93–98.

68. G. Shyamala, X. Yang, G. Silberstein, et al. (1998). Transgenicmice carrying an imbalance in the native-ratio of A to B formsof progesterone receptor exhibit developmental abnormali-ties in mammary glands. Proc. Natl. Acad. Sci. U.S.A. 95:696–701.

69. G. Shyamala, X. Yang, R. D. Cardiff, et al. (2000). Impact ofprogesterone receptor on cell-fate decisions during mammarygland development. Proc. Natl. Acad. Sci. U.S.A. 97:3044–3049.