J. Dinny Graham† and Christine L. Clarke.


Westmead Institute for Cancer Research, University of Sydney, Westmead Hospital, Westmead NSW 2145, Australia.


Address reprint requests to: Christine L. Clarke, Ph.D., Westmead Institute for Cancer Research, University of Sydney, Westmead Hospital, Westmead NSW 2145, Australia.


* Supported by grants from the National Health and Medical Research Council (NHMRC) of Australia, the New South Wales Cancer Council, and the Westmead Hospital Research Institute. J. D. Graham was an NHMRC Dora Lush Biomedical Research Scholar and is now an NHMRC C. J. Martin Fellow.


† Present address: Division of Endocrinology, University of Colorado Health Sciences Center, Campus Box B151, 4200 East Ninth Avenue, Denver, Colorado 80262.



Introduction


The steroid hormone progesterone is a key component in the complex regulation of normal female reproductive function. Broadly speaking, the major physiological roles of progesterone in the mammal are 1) in the uterus and ovary: release of mature oocytes, facilitation of implantation, and maintenance of pregnancy, by promotion of uterine growth and suppression of myometrial contractility; 2) in the mammary gland: lobular-alveolar development in preparation for milk secretion and suppression of milk protein synthesis before parturition; and 3) in the brain: mediation of signals required for sexually responsive behavior. Recent evidence also supports a role for progesterone in modulation of bone mass (Table 1). The recent description of a mouse model carrying a null mutation of the progesterone receptor (PR) gene (1) has served to answer many of the complex questions of progesterone action in vivo and has confirmed the importance and diversity of roles of progesterone in normal female development and reproduction.


Table 1. Physiological functions of progesterone



The pathways of progesterone action in target tissues are not well defined and, in many respects, distinctions remain to be made between the direct downstream targets of progesterone action and indirect consequences of progesterone regulation. Furthermore, as the effects of progesterone are mediated by its receptor and as PR is induced by estrogen in most target tissues, the delineation of specific progesterone effects, as distinct from those of estrogen, is similarly not clear. A number of reviews have described the molecular mechanisms of PR action (2-4), progesterone and progesterone antagonist effects on cellular proliferation (5, 6), and regulation of the concentration of progesterone-responsive proteins (7,8). This review does not seek to replicate existing reviews of the molecular biology of PR action but will provide an overview of the physiological action of progesterone and its regulation of gene expression in target tissues.



Synthesis and Secretion of Progesterone


The ovary is the major site of synthesis and secretion of estrogen and progesterone in the mammal and gives rise to cyclical fluctuations in the levels of these hormones in the circulation (reviewed in Ref. 9). Primary follicles play a dual role in secreting both hormones as well as being responsible for the release of the ovum during the normal cycle. Before ovulation, granulosa cells in the follicle biosynthesize and secrete estrogen. After follicle rupture and release of the ovum, these granulosa cells mature to form the corpus luteum, which is responsible for secretion of progesterone and estrogen in the latter part of the cycle. In the human, if fertilization does not occur within 1 to 2 days, the corpus luteum will continue to enlarge for 10-12 days followed by regression of the gland and concomitant cessation of estrogen and progesterone release. If fertilization occurs, the corpus luteum will continue to grow and function for the first 2 to 3 months of pregnancy. After this time it will slowly regress as the placenta assumes the role of hormonal biosynthesis for the maintenance of pregnancy.


The release of progesterone from the corpus luteum is influenced by a number of hormones. Primary among these is LH, the activity of which is mediated via its intracellular effects on cAMP (10, 11). FSH, PRL, prostaglandins, and β-adrenergic agents also play a role in the control of progesterone secretion (9). Intermediates such as activin, which is stimulated by FSH and inhibits progesterone secretion by granulosa cells, and follistatin, which is synthesized by granulosa cells and is able to bind activin, contribute to a complex pattern of regulation of progesterone secretion. At the time of implantation of the blastocyst in the rat uterus, increased progesterone synthesis is accompanied by induction of ovarian follistatin gene expression, which appears to help in maintaining progesterone secretion (12). However, it is not clear whether follistatin is induced by progesterone to prevent local inhibition of progesterone effects by activin in the uterus or whether follistatin prevents down-regulation of progesterone secretion from the corpus luteum.


Once released, progesterone is carried in the blood by transcortin (corticosteroid-binding globulin) in many species including humans. In the uterine fluid of the rabbit, between days 3 and 12 of pregnancy, an additional progesterone carrier, uteroglobin, is present. Uteroglobin has a postulated role in protection of the embryo during pregnancy (discussed in Section IV), by mechanisms that are still not clear (13,14). A specific progesterone binding plasma protein has also been described in the pregnant guinea pig, which has significantly higher affinity for progesterone than corticosteroid binding globulin (15,16) and is strongly induced from days 15 to 20 of pregnancy, remaining elevated until parturition at approximately day 65 (17). During this time it represents the major progesterone-binding protein in the guinea pig and is specifically synthesized by the placenta (17,18). Synthesis of this protein appears to be under progesterone control since ovariectomy of pregnant animals, or parturition, causes a fall in progesterone binding protein levels concomitant with decreased progesterone levels (19).



The Progesterone Receptor


Progesterone effects are mediated by its nuclear receptor. Receptor proteins that specifically bind progesterone, and are induced by estrogen, were initially characterized in the mammalian uterus and chick oviduct in the early 1970s (20 23). O'Malley and co-workers showed that chick oviduct PR was a dimer composed of two receptor proteins, PR A and PR B, which each bound progesterone (24). In human breast cancer the PR A and B proteins, characterized in vitro (25) and in vivo (26), are detected with molecular masses of approximately 81 kDa and 115 kDa, respectively. The two PR proteins are encoded by a single gene in the human (Fig. 1), under the control of distinct promoters, each of which gives rise to a distinct subgroup of PR mRNA species (27). In contrast, only one PR protein has been described in the rabbit, which has high homology to PR B in the human (28, 29).



Figure 1. The human PR cDNA and PR A and B proteins. The arrows indicate the translation start sites for PR B and PR A (27). DBD, DNA-binding domain; HBD, hormone-binding domain.


PR is a member of a large family of ligand-activated nuclear transcription regulators, which includes receptors for steroids, retinoids, thyroid hormones, and vitamin D. The genes are characterized by organization into specific functional domains that are conserved, to differing degrees, between species and family members. The most highly conserved region between receptor genes is a region in the center of the gene [Fig. 1, DBD (DNA-binding domain)] encoding two "zinc finger" DNA-binding motifs (30). Binding of progestins to the carboxyl-terminal ligand-binding domain of PR [Fig. 1, HBD (hormone-binding domain)] causes association of the progestin-complexed PR dimer with specific progestin response elements (PREs) in target genes, resulting in modulation of transcription of those genes (reviewed in Refs. 3 and 4).


Although both PR A and PR B bind progestins and interact with PREs, there is increasing evidence that they are functionally different. In transfection studies the two proteins have different abilities to activate progestin responsive promoters; these differences are promoterand cell-specific (3134), suggesting that cellular responsiveness to progestins may be modulated via alterations in the ratio of PR A and B expression. While PR B tends to be a stronger activator of target genes, PR A can act as a dominant repressor of PR B (33, 34), suggesting that high PR A expression may result in reduced progestin responsiveness and that PR A and PR B may thus be, respectively, an activator and repressor of progestin action. The repressor role of PR A extends beyond that on PR B, as PR A has been shown to diminish the response of other hormone receptors such as the androgen, glucocorticoid, mineralocorticoid, and estrogen receptors to their appropriate ligands (35-37).


There are interspecies differences in the relative expression of PR A and B in normal tissues. Approximately equimolar expression of PR A and B is observed in chick oviduct (38) and human uterus (39), and a similar ratio of expression is seen in cultured human breast cancer cells (25). In the rodent, PR A expression predominates over PR B in a ratio of 3:1 (40, 41). Alterations in the ratio of PR forms in the chick oviduct during late winter, or in aged nonlaying animals, results in a measurable decrease in PR functional activity (42, 43). In human breast tumors, the ratio of expression of PR A and B proteins differs markedly between patients (44). The biological importance of these different ratios of PR expression has not been extensively explored. Little is known of whether relative PR A and B expression is modulated in vivo, although in PR-positive breast cancer cells in culture PR B is preferentially stimulated by estradiol, resulting in a significant decrease in PR A to B ratio (45). Given the functional differences between the two PR proteins demonstrated in vitro, this suggests that the relative expression of PR A and B may influence cellular responsiveness to progesterone.


PR expression and regulation

PR expression has been described in tissues known to be progesterone responsive such as the uterus [mammalian endometrium (46-51) and myometrium (51, 52)]; the ovary [luteinizing granulosa cells and corpus luteum (53), preovulatory granulosa cells (54)]; and the chick oviduct (24) and bursa of Fabricius (55). Specific progestin binding has been described in other reproductive tissues such as testes (56) and vaginal tissue (57). PR has been described in normal and neoplastic breast (58-61), and in the brain, in the pituitary, ventromedial hypothalamus, and preoptic areas (62, 63). PR has also been described in other tissues where the action of progesterone is less well defined, including vascular endothelium (64) and rat thymus (65). Specific progestin binding has been detected in rabbit lung (66), rat pancreatic islets (67), and human osteoblast-like cells (68). A summary of the tissues and cell types in which PR has been detected is shown in Table 2.


Table 2. Tissues and cell types expressing PR



The expression of PR, and therefore sensitivity to progestins, is under the control of estrogen, which increases, and progesterone, which decreases PR expression in most target tissues. PR protein is increased during proestrus or by exogenous estrogen administration in the mammalian uterus (52, 69-74). During the second half of the cycle, as serum progesterone levels increase, total PR levels in the uterus decrease, and this decrease can be brought on prematurely by treatment with progesterone (73). Furthermore, progesterone treatment can oppose the estrogen induction of PR (71). Although progesterone decreases total PR expression in the uterus, down-regulation is not observed in all cell types. During the follicular phase of the cycle, high levels of PR are present in the nuclei of epithelial and stromal cells of the human endometrium and in myometrial smooth muscle cells. In the midand late luteal phase, detection of PR in the luminal and glandular epithelium declines markedly, to undetectable levels; on the other hand, stromal and myometrial cells continue to express high levels of PR despite high circulating progesterone and absent estrogen receptor (ER) (48, 49,51). By contrast with the uterus, PR levels do not decrease in the breast between the follicular and luteal phases (75-78).



Progesterone Regulation of Gene Expression in the Uterus, Ovary, and Chick Oviduct


Progesterone has a central role in reproduction, being involved in ovulation, implantation, and pregnancy. Associated with this is the involvement of progesterone in regulation of uterine function during the menstrual cycle, by control of cyclical changes in proliferation and decidualization. Progesterone is essential for the development of decidual tissues, and if fertilization occurs, high circulating progesterone levels are important not only for facilitating implantation, but also for maintaining pregnancy by stimulating uterine growth and opposing the actions of factors involved in myometrial contraction. The mechanisms underlying these diverse and complex actions of progesterone are yet to be fully defined, but it is clear that progesterone can both stimulate and inhibit cell proliferation in the uterus, depending on the cell type and physiological context, and that it also plays a role in differentiated function. Some of the functions influenced by progesterone include the stimulation of glycogenesis (79, 80), cyclic nucleotide metabolism (8), protein synthesis and secretion (8), and cell cycle regulation (5), although the molecular mechanisms of these effects of progesterone are not known. Intracellular proteins known to be regulated by progesterone, and which therefore may be involved in mediation of its effects, include ER, estrogen metabolizing enzymes, a-fucosidase, and type II cAMP-dependent kinase. Secreted proteins include enzymes for protein, carbohydrate and prostaglandin metabolism, hydrolases, phosphatases, prostaglandins, plasminogen activator, and PRL (8).


Progesterone effects on proliferation and decidualization in the uterus during the menstrual cycle

Changes in proliferative activities of the glandular epithelium and stromal elements of the human endometrium can be correlated directly with circulating levels of estrogen and progesterone (5), with estrogen stimulating proliferation and progesterone opposing the effects of estrogen and causing inhibition of proliferation. Potential mechanisms through which progesterone opposes estrogen action during the menstrual cycle and maintains the balance between the cyclical influences of estrogen and progesterone are discussed in Section VIII. The estrogen-stimulated follicular phase of the cycle is associated with high proliferative activity in both the epithelial and stromal cells (81). This is followed by a decline in proliferation in the first half of the progesterone-dominated luteal phase of the cycle. In the late luteal phase, while proliferative activity remains low in the epithelium, a second peak of proliferation, consistent with decidual changes, is seen in the stromal elements, associated with high serum levels of progesterone and presumably mediated by the continued expression of PR in those cells (48, 49, 51).


Specificity of the proliferative effects of progesterone in decidualization may be due in part to cell type-specific expression and regulation of growth factor receptors and their peptide ligands. Heparin binding epidermal growth factor (EGF)-like growth factor mRNA is specifically induced by progestins in uterine stromal cells (and is a mitogen in these cells) but, in contrast, is repressed by progesterone treatment in luminal and glandular epithelium (82).


PRL is likely to be involved in decidualization. PRL is secreted by both the endometrial stroma (83) and myometrium (84) during the normal cycle in the human and appears to be under the control of progesterone. Synthesis of PRL by the stroma is greatest during the midto late luteal phase (83) and can be induced, in vitro, in follicular phase endometrial tissue by treatment with progesterone (85). Furthermore, progesterone treatment of both follicular and luteal phase tissue explants results in PRL induction, which coincides with early morphological changes resembling the decidualization of cells during early pregnancy (86). The progesterone-mediated increase in PRL secretion from stromal cells is the result of increased PRL mRNA expression and is additive to the stimulatory effects of estrogen and relaxin (87). In contrast, progesterone inhibits PRL mRNA transcription by myometrial cells (84).


Progesterone may influence uterine proliferation and differentiation during the menstrual cycle by regulation of proteases or matrix proteins. Progestins suppress expression of stromelysins in endometrial stromal cells and induce transforming growth factor-β (TGFβ) in these cells, resulting in down-regulation of matrilysin expression in endometrial epithelium in stromal-epithelial cocultures (88, 89). Thrombospondin-1, an extracellular matrix glycoprotein that is expressed in vascular endothelium and inhibits angiogenesis, is induced by progestins in endometrial stromal cells (90). Furthermore, expression of thrombospondin-1 in endometrial stroma in vivo is correlated with stage of the menstrual cycle, with strongest expression seen in the luteal phase, suggesting that progesterone-mediated induction of thrombospondin-1 influences cyclical regulation of vascular formation and differentiation in this tissue.


Progesterone regulation of insulin-like growth factor (IGF) pathways in the endometrium

The inhibitory effects of progesterone on estrogen-mediated cell proliferation in the endometrium during the menstrual cycle may be mediated by opposition of estrogen action, as discussed in Section VII, but modulation of growth factor pathways may also play a role. The proliferative effects of IGFs are specifically controlled by progesterone, principally through regulation of IGF-binding protein I (IGFBP-I). In humans, IGFBP-I is expressed in a cyclical fashion in endometrial stromal cells, with the highest expression seen in the midto late luteal phase (91). IGFBP-I may act in a paracrine fashion to prevent epithelial cell proliferation during the late luteal phase, since progestins increase IGFBP-I secretion from endometrial stromal cells both in vitro (92) and in vivo (93). It is postulated that by binding to IGF-I, IGFBP-I prevents binding of the growth factor to its receptor, resulting in decreased cellular responsiveness to IGF-I (91, 94). Progesterone and IGFBP-I may also form an autocrine loop controlling stromal cell proliferation at the end of the luteal phase, since IGFBP-I treatment blocks the proliferative effects of both IGF-I and progestins on stromal cells in culture (95). Alternatively, it has been postulated that IGFBP-I may play a role in tissue remodeling toward the end of the cycle, by binding a5β1-integrin, a specific cellular receptor for the extracellular matrix protein fibronectin, and thus altering cell motility (96, 97).


Control of ovulation

The presence of PR in most follicular cell types and in the corpus luteum of the human ovary (98) suggests that the process of ovulation is regulated by progesterone, an interpretation confirmed by studies on PR null mice, which fail to ovulate despite the presence of mature preovulatory follicles (1). Relaxin is increased in the endometrium of nonpregnant women during mid to late secretory phase and is postulated to be progesterone-dependent (99). Studies with rat granulosa cells in culture suggest that the increase in relaxin may facilitate follicle rupture by increasing the secretion of plasminogen activator, collagenase, proteoglycanase, and β-glucuronidase (100). This suggestion is supported by reports in mice that treatment with epostane, which inhibits 3β-hydroxysteroid dehydrogenase, resulting in decreased serum progesterone levels, inhibits the activities of serine proteases, kallikrein, and plasminogen activator and suppresses ovulation (101). Treatment with progesterone relieves the suppression, just as treatment with the antiprogestin RU 38486 suppresses ovulation (102), an inhibition associated with decreased protease activity (103), implying that progesterone is responsible for their induction.


The formation of the corpus luteum represents a distinct intraovarian process and appears to be progesterone-dependent. Expression of PR is induced by LH in granulosa cells of mature preovulatory follicles (104), and PR is detectable in the primate corpus luteum despite high local progesterone concentrations (53, 105). Granulosa cells from mature preovulatory follicles of PR null mice show an inability to luteinize correctly despite prolonged exposure to gonadotropins (1).


Implantation, uterine proliferation, and early pregnancy

Progesterone has a major role in the endometrium in preparation for implantation of a fertilized ovum, and in many species a decrease in circulating progesterone at the time of fertilization is sufficient to delay implantation (106). Progesterone is important in promoting and maintaining implantation through effects on both the maternal uterus and on the developing blastocyst. Progesterone facilitates implantation by stimulating the synthesis of enzymes responsible for lysis of the zona pellucida. However, while progesterone is known to be essential for implantation to occur, lysis of the zona is not the crucial step in this process, suggesting that other essential progesterone-mediated events are yet to be described in the initiation of implantation (106). PRL plays a role in implantation, and this is supported by recent observations that female mice that are PRL receptor null have complete failure of embryonic implantation, leading to sterility (107).


The induction of uterine cell proliferation in early pregnancy may be mediated by locally produced growth factors, many of which are under progesterone control. Furthermore, cell type-specific expression of growth factor receptors controls cellular sensitivity to the autocrine/paracrine effects of growth factors. Progesterone induction of growth factor secretion from the luminal and glandular epithelium in the mouse endometrium promotes proliferation of the EGF receptor-positive blastocyst trophectoderm to facilitate implantation (108). In early pregnancy, EGF receptor mRNA is also induced in the stroma of the maternal uterus by progesterone, but not in the luminal or glandular epithelium (108). It has been suggested that the hemopoietic growth factor, colony stimulating factor-I, exerts a paracrine influence on the growth and differentiation of the placental trophoblast, and its secretion from the luminal and glandular epithelium in the mouse is regulated by estrogen and progesterone (109). In the first 2 days of pregnancy in the mouse, IGF-I is secreted from the luminal and glandular epithelium of the uterus under estrogen stimulation and may contribute to effects on the blastocyst. After this time, secretion from the epithelium declines and significantly greater synthesis and secretion of IGF-I by the stroma are induced by progesterone, resulting in increased proliferation and enlargement of the uterus (110). It is postulated that increased growth factor receptor expression in the stroma mediates the effects of EGF, TGFα, and heparin-binding EGF-like growth factor from the epithelium and IGF-I from the stroma, resulting in tissuespecific stimulation of proliferation.


The molecular mechanisms of progesterone action during pregnancy have been studied intensively in the rabbit uterus. In particular, uteroglobin, which is transcriptionally regulated by progesterone, has been well characterized at both the cellular and molecular levels. Uteroglobin is expressed between days 3 and 12 of pregnancy in the rabbit uterus. It is a dimer of two identical 8-kDa subunits and incorporates two FeII ions into its normal structure. Although it shows acid phosphatase activity, its primary role has been suggested to be the binding and transport of progesterone and its metabolite 5-a-pregnane-3,20-dione, which protects the blastocyst from the high levels of circulating progesterone required for maintenance of pregnancy (13). Alternatively, it has been hypothesized that uteroglobin may protect the embryo from maternal immune and inflammatory response during implantation by contributing to the inhibition of phospholipase A2 activity—a key point in the regulation of these response pathways (14).


Expression of uteroglobin is almost exclusively confined to the rabbit uterus, the exception being the lung where its expression is constitutive and is not regulated by progesterone (13). The uteroglobin gene is encoded by three exons (111), and progesterone regulation of the gene is mediated via binding of PR to specific regulatory elements in the 5'flanking region of the gene 2 to 3 kb upstream of the start of transcription (112). Other progesterone-stimulated proteins are also postulated to bind the uteroglobin gene in positions more proximal to the promoter and to function as transacting factors in progesterone regulation of the gene (113). The binding of these proteins may be a mechanism by which the strict tissue specificity of uteroglobin expression is maintained (114). Protein binding to the regulatory region of the uteroglobin gene is also modulated by other pregnancyassociated proteins. PRL, acting through its receptor, augments progesterone effects by increasing protein binding to the uteroglobin promoter (115). Uteroglobin is expressed only in the rabbit, although when transgenically expressed in the mouse, the uteroglobin promoter is specifically regulated by progesterone in the uterus (116). A homolog of uteroglobin, the Clara cell 10-kDa protein (CC10), has been described in the human. However, it is expressed primarily in the lung, and the 5'-flanking regions of the gene, which correspond to the PREs described in rabbit uteroglobin, are only partially conserved, resulting in a lack of progesterone responsiveness (117).


Myometrial contractility

Progesterone suppresses myometrial contractility during pregnancy, and a number of mechanisms exist whereby this may be mediated, including progesterone effects on intracellular calcium concentration, and levels of prostaglandins, relaxin, and oxytocin. Increases in free intracellular calcium, if unopposed, lead to myometrial contraction. Induction and secretion of calcitonin, a peptide hormone involved in calcium homeostasis, are postulated to lower free calcium levels in the uterus, thereby preventing contraction (118). In the rat uterus, expression of calcitonin is induced in glandular epithelial cells during early pregnancy. This effect can also be achieved by progesterone treatment after estrogen priming, suggesting that progesterone is primarily responsible (118). It is also postulated that suppression of gene expression of the calcium transporter calbindin-D9k prevents increases in intracellular calcium and therefore contributes to prevention of myometrial contraction. Calbindin-D9k mRNA expression in the pregnant rat uterus decreases significantly with increasing endogenous progesterone levels, and this decrease can be blocked by the antagonist RU 38486, suggesting that the effect is PR-mediated (119). Furthermore, calbindinD9k levels in rat uterus are lowest during the progesteronedominated diestrus phase of the cycle, and estrogen induction of calbindin-D9k mRNA can be blocked by the progesterone agonist R5020 (120).


Progesterone inhibits prostaglandin synthesis and activity in the pregnant sheep and therefore decreases myometrial contractility. This inhibition is mediated by a number of pathways that include blocking prostaglandin action, decreasing prostaglandin synthesis, and increasing its rate of inactivation. Progesterone is thought to stimulate prostaglandin 15-dehydrogenase, which catalyzes prostaglandin oxidation and results in inactivation (121). Progesterone opposes the effects of prostaglandins in the human uterus, during pregnancy, and in the luteal phase of the cycle by decreasing the levels of prostaglandins F2α and E in the endometrium. Furthermore, estrogen stimulation of prostaglandin F2α expression in the luteal phase of the cycle in the human endometrium is inhibited by progesterone (122). A fall in progesterone levels at the end of pregnancy is associated with increased prostaglandin synthase activity and prostaglandin F2α production, leading to parturition (121). The antiprogestin RU 38486 antagonizes all the actions of progesterone on prostaglandin synthesis and catabolism and stimulates prostaglandin production, resulting in its abortifacient effect (123).


Prostaglandin effects are mediated by prostaglandin receptors and indirectly via oxytocin receptors, proteins that are also regulated by steroid hormones. Oxytocin receptors are decreased by progesterone in uteri of ovariectomized ewes (124). Oxytocin receptor levels are also inhibited in the human uterus by blocking PGF2α production; conversely, PGF2α induction of luteolysis results in decreased plasma progesterone and a parallel increase of oxytocin receptors (125). Angiotensin II receptors are increased in the rabbit uterus by estrogen priming, resulting in increased contractile sensitivity. This effect is blocked by progesterone treatment, and progesterone alone markedly decreases angiotensin II receptor expression (126). Similarly, atrial natriuretic factor receptors are decreased by progesterone in the rat myometrium during pregnancy, resulting in refractoriness to the tocolytic effects of atrial natriuretic factor on the uterus. It has been postulated that this is mediated by abrogation of estrogen induction of these receptors (127).


During pregnancy the adrenergic system is involved in myometrial quietening. Progesterone increases transcription of β-adrenergic receptors in myometrium from late pregnant rats, resulting in increased sensitivity to adrenergic agents (128). Relaxin is also important in inhibiting spontaneous or prostaglandin-induced myometrial contraction, contributing to the maintenance of implantation and early pregnancy by increasing the collagen framework and distensibility of the uterus (106). The corpus luteum, placenta, and decidua are major relaxin-containing tissues during pregnancy, and progesterone has been shown to be responsible for maintaining relaxin levels (99).


In summary, progesterone has diverse roles in the uterus and ovary at every stage of reproductive function (Table 1). Modulation of cyclical proliferation during the menstrual cycle, regulation of ovulation, stromal growth and decidual formation, promotion and maintenance of implantation, uterine growth, and prevention of myometrial contractility are all dependent upon specific gene regulation by progesterone. It is apparent that transcriptional regulation by progesterone is central to cell-specific growth regulation and involves the coordination of growth factors and their receptors in a complicated array of autocrine and paracrine effects. Similarly, the local signals controlling prostaglandin effects on myometrial contraction involve gene regulation by progesterone at many distinct levels, from regulation of oxytocin signaling to control of prostaglandin synthesis and promotion of calcium homeostasis. Current understanding of the involvement of progesterone in these processes is fragmentary, and the interrelationships between the many regulatory steps largely remain to be described.


Chick oviduct

In the chick oviduct, progesterone induces the synthesis of egg-white proteins, including ovalbumin, conalbumin, lysozyme, and ovomucoid, and it is postulated that induction is mediated by binding of PR to the genes encoding these proteins (129). Hormonal induction of ovalbumin, in particular, has been well described in this system, and its cDNA was one of the first to be fully sequenced (130, 131). The ovalbumin gene 5'-flanking region contains several PR-binding regions, as well as regions that are postulated to bind other proteins and thus influence progesterone and estrogen regulation of the gene (129,132,133). Two regions within the ovalbumin 5'-flanking sequences mediate induction of a reporter gene by progesterone in transfection studies (31, 134). Estrogen is also able to activate transcription through these regions (31, 135) and in some cells acts additively with progesterone-bound PR A, but not PR B, to induce ovalbumin gene transcription (31). It is not clear whether PR and ER act through overlapping or distinct motifs in these regions. Estrogen action on the proximal steroid-responsive region of the ovalbumin gene involves interaction of the fos-jun complex with this region (136). It has been reported that progestins down-regulate c-jun transcription in estrogen-withdrawn chick oviduct (137), although the implications of this regulation for estrogen control of ovalbumin expression have not been explored.



Progesterone Action in the Breast


The major developmental role of progesterone in the normal breast has been postulated to be the formation of lobularalveolar structures during pregnancy (138). This is supported by the observation that mammary glands in PR null mice develop ductal structures that are relatively similar to the wild type but fail to form an interductal lobular-alveolar structure upon exposure to estrogen and progesterone (1). The influence of progesterone is likely to be proliferative in this process, mediated by progesterone regulation of cell cycle genes, growth factors, and growth factor receptors. Progesterone also exerts a differentiating effect on the breast through its role in lactation. The role of progesterone in differentiated function at other times has not been extensively explored.


In comparison with the uterus, there is less known of the mechanisms through which progesterone exerts its effect in the breast, primarily because of the difficulty of obtaining normal breast tissue and the relative paucity of models of progesterone action in the normal breast. Breast cancer cells have been used extensively as models to examine the role of growth factors and growth factor receptors in mediating progesterone effects. However, the limitation of studying progesterone regulation of gene expression in malignant cells, often derived from metastatic lesions, is the difficulty in extrapolating results to the normal breast. An illustration of this is the difference in progesterone effects on the PRL receptor in breast cancer cells and normal mammary gland. In T-47D and MCF-7 cells, progestins increase PRL receptor levels (139), whereas in the normal mammary gland of pseudopregnant rabbits, progestins antagonize PRL induction of PRL receptors (140). Another example is the demonstrated decrease in PR associated with exposure to progestins in breast cancer cells (141-143), which contrasts with the persistence of PR in the breast in the luteal phase of the cycle (75-78).


Effect of progesterone on proliferation of the normal breast

In addition to the developmental role of progesterone in formation of lobular-alveolar structures, there is an increasing body of in vivo evidence that supports a role for progesterone in the induction of cyclical proliferation in the breast. A number of studies have examined the effects of cyclical hormonal changes during the menstrual cycle on DNA synthesis in normal breast epithelium, and there is general agreement between studies that an increase in DNA synthesis is seen in the late luteal phase of the natural cycle (144-148). The increase in DNA synthesis is consistent with the observation of a cyclical increase in the number of epithelial mitoses, which peaks toward the end of the luteal phase and is followed by an increase in apoptotic activity (149,150). These in vivo data are further supported by the observation that high circulating progesterone levels during pregnancy are responsible for inducing marked lobular-alveolar development of the breast in preparation for lactation (151).


In contrast to this, a recent study examined proliferation in breast tissue from patients who had received percutaneous estrogen and progesterone administration to the breast before surgery (152). Epithelial mitoses and expression of proliferating cell nuclear antigen were lowest in progesterone-treated samples, compared with both untreated controls and those receiving estrogen or estrogen plus progesterone (152). However, these data should be interpreted with caution: while this is clearly an in vivo study, the duration of hormone administration, the percutaneous route of administration, and the levels of hormones applied were likely to result in tissue levels of hormones different from those found during the menstrual cycle, with attendant difficulty in extension of these effects on proliferative parameters to those observed in the breast during natural cycles.


In vitro studies of the involvement of progesterone in breast epithelial proliferation have produced inconsistent results (Table 3 and Ref. 5). Although estrogen consistently increases proliferation of normal breast epithelium in vitro, the progesterone effects either alone or combined with estrogen have been variable. Progesterone has been found to increase DNA synthesis in normal mammary epithelium in organ culture (153). However, progesterone either decreases, or has no effect on, the proliferation of normal breast epithelium explanted into nude mice (154, 155). Gompel et al (156), who examined the effects of estrogen and the progestin R5020 on the growth of cultured normal breast epithelial cells, found that estrogen and progesterone had opposing effects, estrogen increasing and progesterone decreasing cell proliferation.


Table 3. Effects of estrogen and progesterone on cell proliferation in the breast



It is not known why the in vivo evidence in support of a role for progesterone in cell proliferation in the breast is difficult to reproduce in vitro, but a contributing factor may be the limitations inherent in attempts to reproduce the physiological environment of breast tissue, including paracrine effects from surrounding stroma on epithelial cell proliferation. Further studies on determinants of cell proliferation in the breast and the involvement of ovarian hormones in this process are required to reconcile the discrepancies in the published data on the role of progesterone in cell proliferation. The effects of estrogen and progesterone on proliferation in the breast, in the in vitro and in vivo models used, are summarized in Table 3.


Progesterone regulation of genes associated with cell cycle progression

Insights into the mechanisms underlying proliferative effects of progesterone in the breast have been obtained from studying the effects of progestins on the cell cycle, primarily in breast cancer cells in culture. A transient increase in cell cycle progression is seen in PR-positive T-47D breast cancer cells after administration of progesterone, which is correlated with a short-lived induction of genes associated with cell cycle progression (157-159). This can be demonstrated by progestin treatment of cells that have been growth arrested in G1 phase by serum deprivation and then released by treatment with insulin, which is a strong mitogen in these cells. Under these conditions, the cells are stimulated into a single round of synchronized progression through the cell cycle, an effect accompanied by transient increases in expression of cell cycle-regulatory genes, such as cyclins and cyclin-dependent kinases, and of protooncogenes associated with proliferative activity, such as c-myc and c-fos (158,159). The progestin induction of cell cycle progression reflects an increased rate of progression of cells already in transit through the cycle, rather than increased numbers of cells entering S phase. Furthermore, while progestins potentiate the insulin-mediated increase in cyclin D1 mRNA levels, the timing of the cyclin D1 induction remains the same as seen with insulin alone (159). Therefore, although the increases in cyclin D1 and c-myc expression resulting from progestin treatment can be blocked by RU 38468, demonstrating that the progestin effects are PR-mediated, it is unlikely that cell cycle genes are direct targets of progesterone action. The temporal relationship between progestin regulation of cell cycle progression and expression of cell cycle genes is summarized schematically in Fig. 2.



Figure 2. Relationship between progestin regulation of cell cycle progression, cell cycle gene expression, and growth factor gene expression in breast cancer cell lines. Schematic representation of progestin regulation of cell cyclerelated genes (middle panel) and growth factor and growth factor receptor genes (lower panel), and comparison to timing of progestin-induced cell cycle progression into S phase (upper panel). EGF, Epidermal growth factor; EGFR, EGF receptor; TGFα, transforming growth factor-α; TGFβ, transforming growth factor-β. [Redrawn from information contained in Refs. 157-159,161,164, and 231].


In addition to stimulating expression of genes associated with cell cycle progression, progestins may act by inhibiting the expression of genes responsible for suppression of cell growth. Expression of the tumor suppressor protein p53 is decreased by progestins in T-47D breast cancer cells (160), suggesting that progestins may stimulate proliferation of these cells by removing the inhibitory effects of this protein. Furthermore, loss of progestin responsiveness, resulting in constitutive down-regulation of p53, may be a mechanism by which increased, unregulated proliferation may occur.


Progesterone regulation of growth factors and growth factor receptors in the breast

Growth factors and growth factor receptors have been proposed as candidate mediators of progesterone effects on cell proliferation. EGF mRNA (161) and EGF receptor protein (162) and mRNA (163) are elevated by progestins in T-47D breast cancer cells. Progestins also increase expression of TGFα mRNA and modestly decrease that of TGFβ in T-47D breast cancer cells (164). The effect is time- and dose-dependent and can be inhibited by RU 38486. The implications of these effects were at first unclear, since it is believed that EGF and TGFα stimulate and TGFβ inhibits the growth of breast epithelial cells, and yet progesterone inhibits T-47D cell growth. However, more recent data suggest that progestins have both stimulatory and inhibitory effects on breast cancer cell growth (158), and that increased growth factor expression may be associated with the transient growth stimulation of these cells by progestins, before growth inhibition. It has also been suggested that overexpression of EGF and TGFα may represent one mechanism by which breast cancer cells acquire progestin resistance, as the development of resistance by a subline of T-47D-5 cells coincided with elevated levels of these factors (165). The above data must be interpreted with caution as in many cases regulation of mRNA expression only has been demonstrated, and increased mRNA levels may not necessarily lead to increased concentrations of biologically active signal or receptor. It is also noteworthy that the timing of the modulation in growth factor and growth factor receptor levels is not consistent with these factors having a primary role in mediating progesterone actions on cell proliferation: the increased expression of growth factors and their receptors occurs after the changes in cell cycle gene expression and is not associated closely with the increase in S phase distribution. The timing of growth factor gene regulation by progestins is summarized schematically in Fig. 2.


Progesterone also increases insulin receptor expression in both T-47D cells (166) and the subline T-47Dco (167). Although progesterone alone inhibits growth of these cells, cotreatment with progestin and insulin resulted in a synergistic induction of T-47D cell growth, suggesting that the progesterone-mediated increase in insulin receptor expression may result in greater sensitivity to the mitogenic effects of insulin (166, 168). This is consistent with the ability of progestins to potentiate insulin effects on synchronously growing breast cancer cell cultures (159). These effects have been postulated to have negative implications for the therapeutic use of progestins, since growth-stimulatory effects may be seen in breast tumors that express elevated levels of insulin receptors and IGF receptors.


In contrast to the effects of progestins on EGF and insulin receptor pathways, which are postulated to lead to increased cell proliferation, progestins are generally believed to inhibit the mitogenic effects of IGFs in breast cancer cells. This is in keeping with the observations in the uterus (see Section IV) and may be a mechanism through which progesterone-mediated inhibition of cell proliferation in breast cancer cells takes place. However, while in the uterus IGFBP1 is likely to be involved in progesterone modulation of IGF action, in breast cancer cells IGFBP1 is not widely expressed (169,170). Nevertheless, if IGFBP1 is expressed, it has been postulated to inhibit the mitogenic effects of IGF-I in breast tumors (171, 172), similar to its action in the endometrium. Progestin inhibition of the mitogenic effects of IGFs in breast cancer cells is likely to occur by modulation of IGF receptor concentrations. While breast cancer cells express type I IGF receptors, few express IGF-I; in vivo this is thought to be contributed by surrounding stromal cells, producing a paracrine mitogenic effect on tumor growth (173). Progestins decrease expression of IGF receptors in T-47D cells and increase IGFII, which produces a further down-regulation of IGF receptor (168, 174).


In summary, although there is evidence in support of a role for progesterone in cell proliferation in the breast, the underlying mechanisms are not clear, and the lack of appropriate models has slowed progress in this area. Much of the data on mechanisms of progestin action in the breast are derived from studies on breast cancer cells in culture: although these provide important information, the validity of extending this information to the normal breast has yet to be fully tested. Modulation of the cell cycle is associated with progesterone effects on cell proliferation: it remains to be determined whether direct PR involvement or, more likely, some other progesterone-regulated factor or factors underlies these effects. Furthermore, while progestins increase the expression of EGF and its receptor, the timing of these effects argues against their having a causative role in mediating progestin effects on cell proliferation. However, although the role of EGF pathways in mediating progesterone effects are unclear, progestins have been shown consistently to inhibit the mitogenic activity of the IGF pathways in both breast and uterus.


Markers of progestin action in the breast

Genes that are independently regulated by progestins in the mammary gland and, therefore, may act as markers of progestin responsiveness have been sought for their potential therapeutic or prognostic value in breast cancer. Two such candidate genes are those encoding the enzymes fatty acid synthetase and alkaline phosphatase.


Tissue-unspecific isoforms of alkaline phosphatase are detectable in the normal breast and breast milk and are induced by progestins in rat endometrial cells (175). In contrast, placental-type alkaline phosphatase activity is induced by estrogen but not progestins in the Ishikawa endometrial carcinoma cell line (176). Di Lorenzo and co-workers (177,178) reported that tissue-unspecific alkaline phosphatase activity was induced by progestins in T47D breast cancer cells, and that this induction was accompanied by the acquisition of a differentiated, secretory phenotype. The increase in activity was due to increased expression of alkaline phosphatase mRNA, resulting in new alkaline phosphatase protein synthesis (178) rather than increased activity of existing enzyme. The progestin induction of alkaline phosphatase activity has not been characterized in vivo, and the physiological significance of its activity, the type of isoforms expressed in breast tumors, and the clinical significance of alkaline phosphatase activity in breast cancer remain to be determined.


Fatty acid synthetase was cloned from the MCF-7 breast cancer cell line by a subtractive hybridization strategy designed to detect progestin-regulated genes (179). It was subsequently shown (180) that the fatty acid synthetase protein had also been identified previously by [35S]methionine labeling as a progestin-responsive protein in both MCF-7 and T-47D breast cancer cells (181). Progestins rapidly induce fatty acid synthetase both transcriptionally and posttranscriptionally, with an increase in gene transcription detectable as early as 15 min after hormone treatment, and a concomitant stabilization of mRNA (182). Furthermore, the progestin antagonist RU 38486 decreased basal transcription, stabilized existing mRNA, and blocked progestin induction of fatty acid synthetase, demonstrating that the effect was PR-mediated. Fatty acid synthetase catalyzes the conversion of acetyl-CoA and malonyl-CoA into fatty acid, and its induction in breast cancer cells is accompanied by increased lipid synthesis and the accumulation of lipid droplets (183). The enzyme is postulated to be a marker of differentiation and progestin responsiveness in breast cancer (184). As in the case of alkaline phosphatase, the clinical significance of fatty acid synthetase expression in breast cancer remains to be demonstrated. Studies have revealed that fatty acid synthetase mRNA levels in breast cancer tissues, measured by in situ hybridization, are not correlated to ER or PR levels or to node involvement. However, the enzyme may act as a marker of proliferation in benign mastopathies, since its expression is higher in cysts and lobules than in ducts (184).


Progesterone effects on lactation

In the normal breast, progesterone acts synergistically with estrogen and PRL during pregnancy to prepare for lactation by promoting lobuloalveolar development (185). Progesterone also acts as an anti-PRL by preventing the synthesis of milk proteins in midto late pregnancy (7) and by inhibiting PRL secretion in women expressing abnormally high circulating PRL (186). In pseudopregnant rabbits, progesterone antagonizes PRL induction of PRL receptors (140). A sudden fall in circulating progesterone accompanies parturition and is associated with a concurrent increase in PRL secretion and the onset of lactation.


α-Lactalbumin, part of the lactose synthetase complex, is involved in lactogenesis after parturition and is induced by PRL, insulin, and glucocorticoids. Glucocorticoid induction of α-lactalbumin synthesis is specifically blocked by progestins in midpregnant rat mammary gland explants (187). The progesterone effect is seen at low concentrations, suggesting that it is PR-mediated. Several putative glucocorticoid/progesterone response elements have been identified in the α-lactalbumin gene (188), and PR is postulated to compete with GR for binding of these elements, resulting in antagonism of glucocorticoid effects (187, 189). cAMP levels are increased by progesterone during pregnancy and are also able to block hormonal stimulation of α-lactalbumin (138). In the final days of pregnancy and during lactation, α-lactalbumin synthesis increases significantly and is insensitive to induction by glucocorticoids or suppression by progestins (190). In the mouse, the lack of effect of progesterone on α-lactalbumin during lactation is attributed to a specific suppression of PR expression, which is refractory to stimulation by exogenous estrogen (191). However, this does not explain the observed loss of α-lactalbumin suppression during late pregnancy, when PR remains high (190). Another major milk protein, β-casein, is also under progesterone control. β-Casein mRNA expression is blocked by progesterone during pregnancy (192), presumably through the binding of pregnancy-specific factors to the casein gene promoter (193). This repressor effect is lost upon parturition, and β-casein mRNA expression is greatly increased during lactation.


In summary, studies of the physiological actions of progesterone in the breast have been focused primarily on the roles of this hormone in cell proliferation and milk protein regulation (Table 1). While the involvement of progesterone in the regulation of some milk protein genes has been defined and there are data in vitro describing genes likely to be implicated in control of cell proliferation, on the whole the molecular action of progesterone in vivo in the breast is poorly understood, there are few experimental models of normal breast physiology available, and genes that are direct targets of progesterone action have not been described in the normal breast in vivo.



Progesterone Effects in the Brain


Estrogen and progesterone control specific brain functions involved in reproductive behavior. In guinea-pigs and rats, sequential secretion of estrogen and progesterone during the estrous cycle induces the sexually receptive lordosis response that coincides with ovulation. Induction of PR expression in the ventromedial hypothalamus and preoptic area, by estrogen or by sequential exposure to estrogen and progesterone, follows a time course that is correlated with the observed increase in lordosis response (194-196). Furthermore, while sequential treatment with estrogen and progesterone initially results in an even greater induction of PR than with estrogen alone (195,196), receptor levels are later downregulated, and this is accompanied by a parallel decrease in lordosis behavior (197, 198). A second progesterone treatment is ineffective on both PR and behavior (198). These data suggest the direct involvement of PR and progesterone in the regulation of sexual behavior. This is supported by the finding that sexual behavior can be abrogated by the direct delivery of antisense PR oligonucleotides to the cerebral ventricle of the rat brain (199). Sequential administration of estrogen and progesterone to these animals elicited a reduced or absent lordosis response, and the suppression was dependent on the oligonucleotide dose. Furthermore, PR was significantly lower in the hypothalamus of these animals. Similarly, PR null mice show a complete lack of lordosis response after hormone administration (1), demonstrating an absolute requirement for PR expression.


The mechanisms by which progesterone acts in the brain are not fully defined; however, progesterone is known to affect the expression of a number of proteins. Progesterone stimulates y-aminobutyric acid (GABA) signaling pathways in specific areas of the brain. Progesterone-mediated increases in GABAA receptor binding sites in a number of regions of the brain, including some areas where PR expression is low or absent, are postulated to contribute to stimulation of lordosis behavior in rats and hamsters, suppression of aggressive behavior, and induction of the release of GnRH (200 -202). Part of this effect may be mediated by direct interaction between 5a-reduced progesterone metabolites and GABAA receptor complexes in PR-negative regions of the brain (Refs. 202 and 203 and references therein) as well as by PR in areas such as the hypothalamus. Sequential estrogen and progesterone treatment, but not estrogen alone, potentiates oxytocin induction of norepinephrine release from the ventromedial hypothalamus (204), which in turn mediates hormone-dependent sexual behavior via noradrenergic projections. Progesterone and estrogen may also regulate behavior by affecting synthesis of POMC, the precursor of β-endorphin, in the ventromedial hypothalamus; estrogen down-regulates the synthesis of this peptide, and preliminary data suggest that progesterone prevents this down-regulation (205). β-Endorphin decreases pituitary secretion of LH and FSH. Adenylate cyclase activity and cAMP levels are rapidly increased (206) and serotonin turnover is down-regulated (207) by progesterone in the ventromedial hypothalamus-preoptic area in rats, and both are implicated in increased sexual receptivity.


Progesterone also affects gene expression in areas of the brain not involved in sexual behavior. PR is detectable in the cortex, hypothalamus, and pituitary within the first few days of postnatal life in rats and in the cortex may play a role in early learning patterns (63). Furthermore, progesterone treatment increases β-adrenergic receptors in the rat cortex, postulated to be involved in modulation of emotional activity (208). A recent report suggested that the rat PR isoforms have different functions in different areas of the brain. The study found that PR A is more highly expressed in the hypothalamus-preoptic area, whereas PR B predominates in the cortex (209). Rat PR A and B have been demonstrated in vitro to be differently inducible by estrogen (210, 211), suggesting that hormone regulation of PR expression may differ between the hypothalamus and cortex.


In summary, progesterone regulates signals in the brain involving sexually responsive behavior. The most well defined aspect of progesterone effects on this process are PRmediated effects in the hypothalamus and preoptic area. Progesterone effects in the brain may also be mediated by nonclassic mechanisms of action such as direct interaction of progesterone metabolites with other receptors, such as GABAA receptors. Furthermore, the relative expression of PR A and B may be important in determining progesterone effects in specific sites in the brain.



Progesterone Effects on Bone


Expression of both ER and PR in normal human osteoblastlike cells has been reported (68, 212), and several lines of evidence support a role for steroid hormones in regulating the expression and function of matrix proteins and metalloproteinases involved in bone remodeling and resorption. Estrogen down-regulation of mRNA for bone matrix-associated proteins has been reported to decrease bone resorption and formation, resulting in a net slowing of the rate of loss of bone mass (213, 214) and thereby supporting the use of estrogen agonists in the prevention of osteoporosis in postmenopausal women.


Progesterone may have a role in bone matrix regulation, via its effect on metalloproteinases. As mentioned earlier, progestins regulate proteinase activity in the uterus, suppressing expression of stromelysins in endometrial stromal cells and inducing TGFβ, resulting in down-regulation of matrilysin expression in the endometrial epithelium component of stromal-epithelial cocultures (88, 89). The demonstration that a sequence contained in the 5'-flanking region of the mouse gene encoding the bone matrix protein, osteonectin, can act as a PRE in vitro suggests that progestins may also regulate this protein in vivo (215).


It has been suggested that progesterone regulation of bone remodeling may also be indirectly facilitated by the ability of progesterone to act as a ligand for the glucocorticoid receptor. Glucocorticoids have been implicated in the process of bone loss through their ability to block 1,25-(OH)2-vitamin D-induced osteocalcin synthesis (216) and to prevent attachment of osteoblasts to matrix proteins, including osteonectin, possibly through down-regulation of β1-integrin and other cell surface attachment factors (217). Glucocorticoids also increase bone sialoprotein mRNA levels in rat osteosarcoma cells, an effect that can be blocked by 1,25-(OH)2-vitamin D and has been postulated to contribute to acceleration in maturation of preosteoblasts and ultimately to contribute to bone loss (218). Progesterone has been postulated to antagonize glucocorticoid-mediated effects in bone, resulting in abrogation of glucocorticoid-induced bone loss (219).


In summary, although the data are preliminary at this stage, progesterone appears to modulate bone remodeling, resulting in protection against bone loss. This effect appears to be mediated by PR expression in osteoblasts, as well as through binding to glucocorticoid receptor and perhaps reducing the influence of glucocorticoids.



Antiestrogen Action of Progesterone


Many of the effects of progesterone are thought to be due to its ability to oppose the action of estrogen, particularly in the uterus. Progesterone abrogates estrogen induction of many of the known hormone-responsive genes, and this effect is mediated by down-regulation of cytoplasmic and nuclear ER protein concentrations, decreasing the active estrogen concentration (reviewed in Ref. 5) and antagonizing the action of ER at the molecular level. The importance of this balance between estrogenand progesterone-mediated effects is demonstrated by the extensive proliferation of the luminal and glandular epithelium, consistent with unopposed estrogen effects, in the uterus of ovariectomized PR null mice (1).


Inhibition of ER expression

The mechanism of progesterone action on ER was initially elucidated in the mammalian uterus. Uterine ER levels were decreased by administration of progesterone to estrogentreated rats (220). Progesterone also antagonized estrogen induction of ER in the rat myometrium and in whole rat uterus (221,222). Furthermore, administration of a synthetic progestin, medroxyprogesterone acetate, to women undergoing curettage during the follicular phase of the menstrual cycle resulted in decreased endometrial ER levels (223). A decrease in ER in hamster decidual cells, due to progesterone-mediated shortening of the ER protein half-life, suggested direct destabilization of ER by progestins (224). The progesterone-mediated decrease in ER protein has been shown more recently in breast cancer cells to result from decreased cellular ER mRNA levels (225), likely to reflect decreased transcription of the ER gene, since the effect was seen rapidly without shortening of the ER mRNA half-life (226).


B. Progesterone inhibition of the molecular action of ER

As well as directly reducing ER concentration, progesterone opposes ER-mediated gene-regulatory events, although the molecular mechanisms of this antagonism are not clear. In terms of this effect, the best defined model is the regulation of PR itself. Progestins inhibit and estrogens stimulate rabbit PR gene expression through the same region in the rabbit PR promoter (29), although this effect is mediated without binding of PR to this region, suggesting that PR may sequester transcription factors that are essential for estrogen action (227). By contrast, progesterone and estrogen effects on human PR appear to be distinct, since estrogen primarily regulates the PR B promoter (45), whereas progestins regulate both PR isoforms in breast cancer cells.


Other recent demonstrations that PR can inhibit transcriptional activation by ER of estrogen-responsive promoters without binding to DNA support the view that PR may act by sequestering transcription factors required for ER activity (36, 37, 228, 229). However, the repressive effects of progesterone appear to be promoterand cell-specific, and there is considerable variability between reports. McDonnell and Goldman (36) reported that PR A but not PR B, in the presence of either progesterone or antiprogestins, lessened the ability of estrogen to induce an estrogen-responsive reporter when the two constructs were transfected into CV-1 or HS578T cells, but not HepG2 cells. PR A had similar antiestrogenic effects on endogenous ER activation of a minimal estrogen-responsive reporter in MCF-7 breast cancer cells in the presence of RU 38486 (37). However, when the estrogenresponsive region of the pS2 gene was used as a reporter in MCF-7 cells, PR B and not PR A repressed activation of the reporter by estrogen (229). The reason for this variability is not yet known, but it is possible that specific accessory proteins involved in transcriptional activation by ER and PR differ between cell types or are expressed at different concentrations, resulting in variable effects. PR A has been demonstrated to have similar repressive effects on other members of the nuclear receptor family, including those for androgens, mineralocorticoids, and glucocorticoids (35-37), although the physiological significance of this observation remains to be determined.



Summary and Conclusion


Examination of the regulation of gene expression by progesterone has revealed the complexity of the diverse roles of this hormone in the control of female reproductive function. The actions of progesterone have been characterized in the most detail in the uterus, where progesterone participates in the cyclical control of proliferation and differentiation during the menstrual cycle and plays a key role in pregnancy, being involved in ovulation, implantation, uterine growth, and maintenance of pregnancy. In the uterus, progesterone both stimulates and inhibits cell proliferation. It promotes differentiated function and, although the mechanisms underlying progesterone opposition of estrogen action have been investigated, most of the mechanisms underlying the diverse roles of progesterone in the uterus still require elucidation. In the breast, progesterone is primarily responsible for development of the lobular-alveolar structures during puberty and pregnancy, but also plays a role in cyclical control of proliferation during the menstrual cycle by mechanisms that are poorly understood. In the brain, the effects of progesterone on sexually receptive behavior have been established in animal studies, but information on the role of progesterone in the human brain is limited. Similarly, although progesterone is likely to play a role in bone remodeling, there is a paucity of information on this at present.


The molecular mechanisms of progesterone action have been described only for a small number of well defined target genes, examples being the uteroglobin and ovalbumin genes, which have been studied in detail. However, progesterone is clearly involved in the regulation of a considerable number of genes about which little is known. Furthermore, the distinction between direct and indirect targets of progesterone action in cellular processes is largely yet to be made. By separating gene-regulatory events, which are central to the physiological effects of progesterone, from the secondary consequences of progesterone action, it may be possible to define the determinants of response to progesterone in normal and malignant cells.



Future Directions


Although significant progress has been made in understanding the physiological actions of progesterone in the mammalian reproductive system and the molecular structure and function of PR, there are still marked gaps in knowledge. There is a great deal to be learned about the biology and hormone responsiveness of the normal breast, and the relative paucity of models has been a limitation in this regard. Importantly, there is a need for more information on the human breast, both normal and malignant, to provide baseline information that will be beneficial in model development, as well as in a better understanding of the physiology of hormone action in the breast.


More information is needed on the significance of PR A and PR B expression. While PR involvement in regulating a host of physiological events has been described, there is little known of the individual roles of the PR proteins PR A and PR B in mediating these effects. While in vitro work suggests that these proteins have different functions and that PR A may be a repressor of PR B in particular, and more generally of other members of the nuclear receptor family, there are no data on the relative activities of PR A and B in vivo. Furthermore, it is not known whether PR A and B are expressed in all target tissues, and within target tissues whether cells express one or both of these proteins. The relative expression of the two PR proteins has not been examined to date in most tissues of the reproductive system. Addressing these questions, which rely on single cell analysis, poses significant technical challenges that will necessitate continued development and refinement of immunohistochemical, in situ, and other methodologies.


The mechanism of PR action may also depend on an array of other proteins, such as the recently described nuclear receptor coactivators and corepressors. Nuclear receptors interact with coregulatory proteins, which may function as intermediates in transcription (230). If they play a role in the transcriptional activity of PR, it is likely that they will be expressed in progesterone target tissues, although this has yet to be described. The role of coregulatory proteins in progesterone action needs further investigation to clarify whether progesterone regulates coregulatory protein expression and whether tissue levels of coregulatory proteins play a role in modulation of progesterone action.


Progesterone has both proliferative and differentiating actions, and identification of genes whose transcription is directly modulated by progesterone is an essential first step in understanding progesterone effects on these complex processes. The number of described genes that are directly modulated by progesterone is small, and many of the effects of progesterone described in this review are likely to be indirect consequences of the action of as yet undescribed gene products. New information on genes directly regulated by progesterone is urgently required, and techniques such as differential display PCR and similar approaches by which to identify progesterone-regulated transcripts, despite their limitations, are likely to yield important new knowledge in the near future.




References


Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR, Montgomery Jr CA, Shyamala G, Conneely OM, O'Malley BW 1995 Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities. Genes Dev 9:2266-2278 

Clarke CL 1993 Ovarian steroid hormone receptors and their mechanisms of action. In: Rice GE, Brennecke SP (eds) Molecular Aspects of Placental and Fetal Autacoids. CRC Press, Inc., Boca Raton, FL, pp 27-54 

Gronemeyer H 1991 Transcription activation by estrogen and progesterone receptors. Annu Rev Genet 25:89-123 

Tsai M-J, O'Malley BW 1994 Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem 63:451-486 

Clarke CL, Sutherland RL 1990 Progestin regulation of cellular proliferation. Endocr Rev 11:266-301 

Horwitz KB 1992 The molecular biology of RU486: is there a role for antiprogestins in the treatment of breast cancer? Endocr Rev 13:146-163 

Savouret JF, Misrahi M, Milgrom E 1988 Molecular action of progesterone. Oxf Rev Reprod Biol 10:293-347 

Savouret JF, Misrahi M, Milgrom E 1990 Molecular action of progesterone. Int J Biochem 22:579-594 

Norman AW, Litwack G 1987 Hormones. Academic Press, Orlando, FL Marsh JM, Butcher RW, Savard K, Sutherland EW 1966 The stimulatory effect of luteinizing hormone on adenosine 3',5'-monophosphate accumulation in corpus luteum slices. J Biol Chem 241: 5436-5440 

Jordan III AW, Caffrey JL, Niswender GD 1978 Catecholamineinduced stimulation of progesterone and adenosine 3',5'-monophosphate production by dispersed ovine luteal cells. Endocrinology 103:385-392 

Mercado M, Shimasaki S, Ling N, DePaolo L 1993 Effects of estrous cycle stage and pregnancy on follistatin gene expression and immunoreactivity in rat reproductive tissues: progesterone is implicated in regulating uterine gene expression. Endocrinology 132:1774-1781 

Savouret JF, Guiochon-Mantel A, Milgrom E 1984 The uteroglobin gene, structure and interaction with the progesterone-receptor. Oxf Surv Eukaryot Genes 1:192-214 

Miele L, Cordella-Miele E, Mukherjee AB 1987 Uteroglobin: structure, molecular biology, and new perspectives on its function as a phospholipase A2 inhibitor. Endocr Rev 8:474-490 

Milgrom E, Atger M, Baulieu E-E 1970 Progesterone binding plasma protein (PBP). Nature 228:1205-1206 

Burton RM, Harding GB, Rust N, Westphal U1971 Steroid protein interactions. XXIII. Nonidentity of cortisol-binding globulin and progesterone-binding globulin in guinea pig serum. Steroids 17: 1-16 

Perrot M, Milgrom E 1978 Immunochemical studies of guinea pig progesterone-binding plasma protein. Endocrinology 103:1678-1685 

Perrot-Applanat M, David-Ferreira JF 1982 Immunocytochemical localization of progesterone-binding protein (PBP) in guinea pig placental tissue. Cell Tissue Res 223:627-639 

Evans JJ, Sin IL, White FJ 1982 Progesterone-binding globulin and progesterone in guinea-pigs after ovariectomy, abortion and parturition. J Steroid Biochem 16:171-173 

Leavitt WW, Blaha GC 1972 An estrogen-stimulated, progesterone-binding system in the hamster uterus and vagina. Steroids 19:263-274 

Milgrom E, Baulieu E-E 1970 Progesterone in uterus and plasma. I. Binding in rat uterus 105,000 g supernatant. Endocrinology 87: 276-286 

Milgrom E, Atger M, Baulieu E-E 1970 Progesterone in uterus and plasma. IV Progesterone receptor(s) in guinea pig uterus. Steroids 16:741-754 

Sherman MR, Corvol PL, O'Malley BW 1970 Progesterone-binding components of chick oviduct. I. Preliminary characterization of cytoplasmic components. J Biol Chem 245:6085-6096 

Spelsberg TC, Steggles AW, Chytil F, O'Malley BW 1972 Progesterone-binding components of chick oviduct. V. Exchange of progesterone-binding capacity from target to nontarget tissue chromatins. J Biol Chem 247:1368-1374 

Lessey BA, Alexander PS, Horwitz KB 1983 The subunit structure of human breast cancer progesterone receptors: characterization by chromatography and photoaffinity labeling. Endocrinology 112: 1267-1274 

Horwitz KB, Alexander PS 1983 In situ photolinked nuclear progesterone receptors of human breast cancer cells: subunit molecular weights after transformation and translocation. Endocrinology 113: 2195-2201 

Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H, Chambon P 1990 Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J 9:1603-1614 

Loosfelt H, Logeat F, Vu Hai MT, Milgrom E 1984 The rabbit progesterone receptor. Evidence for a single steroid-binding subunit and characterization of receptor mRNA. J Biol Chem 259: 14196-14202 

Savouret JF, Bailly A, Misrahi M, Rauch C, Redeuilh G, Chauchereau A, Milgrom E 1991 Characterization of the hormone responsive element involved in the regulation of the progesterone receptor gene. EMBO J 10:1875-1883 

Evans RM 1988 The steroid and thyroid hormone receptor superfamily. Science 240:889-895 

Tora L, Gronemeyer H, Turcotte B, Gaub MP, Chambon P 1988 The N-terminal region of the chicken progesterone receptor specifies target gene activation. Nature 333:185-188 

Meyer M-E, Quirin-Stricker C, Lerouge T, Bocquel M-T, Gronemeyer H 1992 A limiting factor mediates the differential activation of promoters by the human progesterone receptor isoforms. J Biol Chem 267:10882-10887 

Tung L, Mohamed MK, Hoeffler JP, Takimoto GS, Horwitz KB Antagonist-occupied human progesterone B-receptors activate transcription without binding to progesterone response elements and are dominantly inhibited by A-receptors. Mol Endocrinol 7:1256-1265 

Vegeto E, Shabaz MM, Wen DX, Goldman ME, O'Malley BW, McDonnell DP 1993 Human progesterone receptor A form is a celland promoterspecific repressor of human progesterone receptor B function. Mol Endocrinol 7:1244-1255 

McDonnell DP, Shahbaz MM, Vegeto E, Goldman ME 1994 The human progesterone receptor A-form functions as a transcriptional modulator of mineralocorticoid receptor transcriptional activity. J Steroid Biochem Mol Biol 48:425-432 

McDonnell DP, Goldman ME 1994 RU486 exerts antiestrogenic activities through a novel progesterone receptor A form-mediated mechanism. J Biol Chem 269:11945-11949 

Wen DX, Xu Y-F, Mais DE, Goldman ME, McDonnell DP 1994 The A and B isoforms of the human progesterone receptor operate through distinct signaling pathways within target cells. Mol Cell Biol 14:8356-8364 

Schrader WT, O'Malley BW 1972 Progesterone-binding components of chick oviduct. Characterization of purified subunits. J Biol Chem 247:51-59 

Feil PD, Clarke CL, Satyaswaroop PG 1988 Progestin mediated changes in progesterone receptor forms in the normal human endometrium. Endocrinology 123:2506-2513 

Ilenchuk TT, Walters MR 1987 Rat uterine progesterone receptor analyzed by [3H]R5020 photoaffinity labelling: evidence that the A and B subunits are not equimolar. Endocrinology 120:1449-1456 

Schneider W, Ramachandran C, Satyaswaroop PG, Shyamala G Murine progesterone receptor exists predominantly as the 83-kilodalton 'A' form. J Steroid Biochem Mol Biol 38:285-291 

Boyd PA, Spelsberg TC 1979 Seasonal changes in the molecular species and nuclear binding of chick oviduct progesterone receptor. Biochemistry 18:3685-3690 

Boyd-Leinen PA, Fournier D, Spelsberg TC 1982 Nonfunctioning progesterone receptors in the developed oviducts from estrogenwithdrawn immature chicks and in aged nonlaying hens. Endocrinology 111:30-36 

Graham JD, Yeates C, Balleine RL, Harvey SS, Milliken JS, Bilous AM, Clarke CL 1995 Characterization of progesterone receptor A andB expression in human breast cancer. Cancer Res 55:5063-5068 

Graham JD, Roman SD, McGowan E, Sutherland RL, Clarke CL 1995 Preferential stimulation of human progesterone receptor B expression by estrogen in T-47D human breast cancer cells. J Biol Chem 270:30693-30700 

Perrot-Applanat M, Logeat F, Groyer PM, Milgrom E 1985 Immunocytochemical study of mammalian progesterone receptor using monoclonal antibodies. Endocrinology 116:1473-1484 

Clarke CL, Zaino RJ, Feil PD, Miller JV, Steck ME, OhlssonWilhelm BM, Satyaswaroop PG 1987 Monoclonal antibodies to human progesterone receptor: characterization by biochemical and immunohistochemical techniques. Endocrinology 121:1123-1132 

Garcia E, Bouchard P, De Brux J, Berdah J, Frydman R, Schaison G, Milgrom E, Perrot-Applanat M 1988 Use of immunocytochemistry of progesterone and estrogen receptors for endometrial dating. J Clin Endocrinol Metab 67:80-87 

Lessey BA, Killam AP, Metzger DA, Haney AF, Greene GL, McCarty KSJ 1988 Immunohistochemical analysis of human uterine estrogen and progesterone receptors throughout the menstrual cycle. J Clin Endocrinol Metab 67:334-340 

Press MF, Greene GL 1988 Localization of progesterone receptor with monoclonal antibodies to the human progestin receptor. Endocrinology 122:1165-1175 

Press MF, Udove JA, Greene GL 1988 Progesterone receptor distribution in the human endometrium. Am J Pathol 131:112-124 

Janne O, Kontula K, Vihko R 1976 Progestin receptors in human tissues: concentrations and binding kinetics. J Steroid Biochem 7:1061-1068 

Duffy DM, Stouffer RL 1995 Progesterone receptor messenger ribonucleic acid in the primate corpus luteum during the menstrual cycle: possible regulation by progesterone. Endocrinology 136:1869-1876 

Parke-Sarge O-K, Parmer TG, Gu Y, Gibori G 1995 Does the rat corpus luteum express the progesterone receptor gene? Endocrinology 136:1537-1543 

Ylikomi T, Gasc J-M, Isola J, Baulieu E-E, Tuohimaa P 1985 Progesterone receptor in the chick bursa of Fabricius: characterisation and immunohistochemical localization. Endocrinology 117: 155-160 

Terner C 1977 Progesterone and progestins in the male reproductive system. Ann NY Acad Sci 286:313-320 

Batra S, Iosif S 1985 Progesterone receptors in human vaginal tissue. Am J Obstet Gynecol 153:524-528 

Pollow K, Sinnecker R, Schmidt-Gollwitzer M, Boquoi E, Pollow B 1977 Binding of [3H]progesterone to normal and neoplastic tissue samples from tumour bearing breasts. J Mol Med 2:69-82 

Horwitz KB, McGuire WL 1975 Specific progesterone receptors in human breast cancer. Steroids 25:497-505 

Perrot-Applanat M, Groyer-Picard M, Lorenzo F, Jolivet A, Vu Hai MT, Pallud C, Spyratos F, Milgrom E 1987 Immunocytochemical study with monoclonal antibodies to progesterone receptor in human breast tumors. Cancer Res 47:2652-2661 

Perrot-Applanat M, Groyer-Picard M, Vu Hai MT, Pallud C, Spyratos F, Milgrom E 1989 Immunocytochemical staining of progesterone receptor in paraffin sections of human breast cancers. Am J Pathol 135:457-468 

Kato J, Onouchi T, Okinaga S 1978 Hypothalamic and hypophysial progesterone receptors: estrogen-priming effect, differential localization, 5a-dihydroprogesterone binding, and nuclear receptors. J Steroid Biochem 9:419-427 

MacLusky NJ, McEwen BS 1980 Progestin receptors in the developing rat brain and pituitary. Brain Res 189:262-268 

Perrot-Applanat M, Cohen-Solal K, Milgrom E, Finet M 1995 Progesterone receptor expression in human saphenous veins. Circulation 92:2975-2983 

Pearce PT, Khalid BAK, Funder JW 1983 Progesterone receptors in rat thymus. Endocrinology 113:1287-1291 

Giannopoulos G, Phelps DS, Munowitz P1982 Heterogeneity and ontogenesis of progestin receptors in rabbit lung. J Steroid Biochem 17:503-510 

Green IC, Howell SL, El Seifi S, Perrin D 1978 Binding of 3Hprogesterone by isolated rat islets of Langerhans. Diabetologia 15:349-355 

Wei LL, Leach MW, Miner RS, Demers LM 1993 Evidence for progesterone receptors in human osteoblast-like cells. Biochem Biophys Res Commun 195:525-532 

Feil PD, Glasser SR, Toft DO, O'Malley BW 1972 Progesterone binding in the mouse and rat uterus. Endocrinology 91:738-746 

Milgrom E, Atger M, Perrot M, Baulieu E-E 1972 Progesterone in uterus and plasma. VI. Uterus progesterone receptors during the estrus cycle and implantation in the guinea pig. Endocrinology 90:1071-1078 

Milgrom E, Thi L, Atger M, Baulieu E-E 1973 Mechanisms regulating the concentration and the conformation of progesterone receptor(s) in the uterus. J Biol Chem 248:6366-6374 

Rao BR, Wiest WG, Allen WM 1973 Progesterone "receptor" in rabbit uterus. I. Characterization and estradiol-17β augmentation. Endocrinology 92:1229-1240 

Leavitt WW, Toft DO, Strott CA, O'Malley BW 1974 A specific progesterone receptor in the hamster uterus: physiologic properties and regulation during the estrous cycle. Endocrinology 94:1041-1053 

Leavitt WW, Chen TJ, Allen TC 1977 Regulation of progesterone receptor formation by estrogen action. Ann NY Acad Sci 286:210225 

Battersby S, Robertson BJ, Anderson TJ, King RJB, McPherson K Influence of menstrual cycle, parity and oral contraceptive use on steroid hormone receptors in normal breast. Br J Cancer 65: 601-607 

Ricketts D, Turnbull L, Ryall G, Bakhshi R, Rawson NSB, Gazet J-C, Nolan C, Coombes RC 1991 Estrogen and progesterone receptors in the normal female breast. Cancer Res 51:1817-1822 

Soderqvist G, von Schoultz B, Tani E, Skoog L 1993 Estrogen and progesterone receptor content in breast epithelial cells from healthy women during the menstrual cycle. Am J Obstet Gynecol 168:874879 

Joyeux C, Chalbos D, Rochefort H 1990 Effects of progestins and menstrual cycle on fatty acid synthetase and progesterone receptor in human mammary glands. J Clin Endocrinol Metab 70:1438-1444 

Demers LM, Feil PD, Bardin CW 1977 Factors that influence steroid induction of endometrial glycogenesis in organ culture. Ann NY Acad Sci 286:249-259 

Shapiro SS, Dyer RD, Colas AE 1980 Progesterone-induced glycogen accumulation in human endometrium during organ culture. Am J Obstet Gynecol 136:419-425 

Ferenczy A, Bertrand G, Gelfand MM 1979 Proliferation kinetics of human endometrium during the normal menstrual cycle. Am J Obstet Gynecol 133:859 -867 

Zhang Z, Funk C, Roy D, Glasser S, Mulholland J 1994 Heparinbinding epidermal growth factor-like growth factor is differentially regulated by progesterone and estradiol in rat uterine epithelial and stromal cells. Endocrinology 134:1089-1094 

Maslar IA, Riddick DH 1979 Prolactin production by human endometrium during the normal menstrual cycle. Am J Obstet Gynecol 135:751-754 

Gellersen B, Bonhoff A, Hunt N, Bohnet HG 1991 Decidual-type prolactin expression by the human myometrium. Endocrinology 129:158-168 

Daly DC, Maslar IA, Riddick DH 1983 Prolactin production during in vitro decidualization of proliferative endometrium. Am J Obstet Gynecol 145:672-678 

Maslar IA, Ansbacher R 1986 Effects of progesterone on decidual prolactin production by organ cultures of human endometrium. Endocrinology 118:2102-22108 

Huang JR, Tseng L, Bischof P, Janne OA 1987 Regulation of prolactin production by progestin, estrogen, and relaxin in human endometrial stromal cells. Endocrinology 121:2011-2017 

Osteen KG, Rodgers WH, Gaire M, Hargrove JT, Gorstein F, Matrisian LM 1994 Stromal-epithelial interaction mediates steroidal regulation of metalloproteinase expression in human endometrium. Proc Natl Acad Sci USA 91:10129-10133 

Bruner KL, Rodgers WH, Gold LI, Korc M, Hargrove JT, Matrisian LM, Osteen KG 1995 Transforming growth factor beta mediates the progesterone suppression of an epithelial metalloproteinase by adjacent stroma in the human endometrium. Proc Natl Acad Sci USA 92:7362-7366 

Iruela-Arispe ML, Porter P, Bornstein P, Sage EH 1996 Thrombospondin-1, an inhibitor of angiogenesis, is regulated by progesterone in the human endometrium. J Clin Invest 97:403-412 

Giudice LC, Lamson G, Rosenfeld RG, Irwin JC 1991 Insulin-like growth factor-II (IGF-II) and IGF binding proteins in human endometrium. Ann NY Acad Sci 626:295-307 

Bell SC, Jackson JA, Ashmore J,ZhuHH, Tseng L 1991 Regulation of insulin-like growth factor-binding protein-1 synthesis and secretion by progestin and relaxin in long term cultures of human endometrial stromal cells. J Clin Endocrinol Metab 72:1014-1024 

Pekonen F, Nyman T, Lahteenmaki P, Haukkamaa M, Rutanen E-M 1992 Intrauterine progestin induces continuous insulin-like growth factor-binding protein-1 production in the human endometrium. J Clin Endocrinol Metab 75:660 664 

Westley BR, May FEB 1994 Role of insulin-like growth factors in steroid modulated proliferation. J Steroid Biochem Mol Biol 51:1-9 

Frost RA, Mazella J, Tseng L 1993 Insulin-like growth factor binding protein-1 inhibits the mitogenic effect of insulin-like growth factors and progestins in human endometrial stromal cells. Biol Reprod 49:104-111 

Zhou J, Dsupin BA, Guidice LC, Bondy CA 1994 Insulin-like growth factor system gene expression in human endometrium during the menstrual cycle. J Clin Endocrinol Metab 79:1723-1734 

Jones JI, Gockerman A, Busby Jr WH, Wright G, Clemmons DR Insulin-like growth factor binding protein 1 stimulates cell migration and binds to the alpha 5 beta 1 integrin by means of its Arg-Gly-Asp sequence. Proc Natl Acad Sci USA 90:10553-10557 

Suzuki T, Sasano H, Kimura N, Tamura M, Fukaya T, Yajima A, Nagura H 1994 Immunohistochemical distribution of progesterone, androgen and oestrogen receptors in the human ovary during the menstrual cycle: relationship to expression of steroidogenic enzymes. Hum Reprod 9:1589-1595 

Yki-Jarvinen H, Wahlstrom T, Seppala M 1985 Human endometrium contains relaxin that is progesterone-dependent. Acta Obstet Gynecol Scand 64:663-665 

Too CKL, Bryant-Greenwood GD, Greenwood FC 1984 Relaxin increases the release of plasminogen activator, collagenase, and proteoglycanase from rat granulosa cells in vitro. Endocrinology 115:1043-1050 

Tanaka N, Espey LL, Stacy S, Okamura H 1992 Epostane and indomethacin actions on ovarian kallikrein and plasminogen activator activities during ovulation in the gonadotropin-primed immature rat. Biol Reprod 46:665-670 

Loutradis D, Bletsa R, Aravantinos L, Kallianidis K, Michalas S, Psychoyos A 1991 Preovulatory effects of the progesterone antagonist mifepristone (RU486) in mice. Hum Reprod 6:1238-1240 

Iwamasa J, Shibata S, Tanaka N, Matsuura K, Okamura H 1992 The relationship between ovarian progesterone and proteolytic enzyme activity during ovulation in the gonadotropin-treated immature rat. Biol Reprod 46:309-313 

Natraj V, Richards JS 1993 Hormonal regulation, localization, and functional activity of the progesterone receptor in granulosa cells of rat preovulatory follicles. Endocrinology 133:761-769 

Aladin Chandrasekher Y, Melner MH, Nagalla SR, Stouffer RL Progesterone receptor, but not estradiol receptor, messenger ribonucleic acid is expressed in luteinizing granulosa cells and the corpus luteum in rhesus monkeys. Endocrinology 135:307-314 

Rothchild 11983 Role of progesterone in initiating and maintaining pregnancy. In: Bardin CW, Milgrom E, Mauvais-Jarvis P (eds) Progesterone and Progestins. Raven Press, New York, pp 219-229 

Ormandy CJ, Camus A, Barra J, Damotte D, Lucas B, Buteau H, Edery M, Brousse N, Babinet C, Binart N, Kelly PA 1997 Null 

mutation of the prolactin receptor gene produces multiple reproductive defects in the mouse. Genes Dev 11:167-178 

Das SK, Tsukamura H, Paria BC, Andrews GK, Dey SK 1994 Differential expression of epidermal growth factor receptor (EGF-R) gene and regulation of EGF-R bioactivity by progesterone and estrogen in the adult mouse uterus. Endocrinology 134:971-981 

Pollard JW, Bartocci A, Arceci R, Orlofsky A, Ladner MB, Stanley ER 1987 Apparent role of the macrophage growth factor, CSF-I, in placental development. Nature 330:484-486 

Kapur S, Tamada H, Dey SK, Andrews GK 1992 Expression of insulin-like growth factor-I (IGF-I) and its receptor in the periimplantation mouse uterus, and cell-specific regulation of IGF-I gene expression by estradiol and progesterone. Biol Reprod 46: 208-219 

Bailly A, Atger M, Atger P, Cerbon MA, Alizon M, Vu Hai MT, Logeat F, Milgrom E 1983 The rabbit uteroglobin gene. Structure and interaction with the progesterone receptor. J Biol Chem 258: 10384-10389 

Jantzen K, Fritton HP, Igo KT, Espel E, Janich S, Cato AC, Mugele K, Beato M 1987 Partial overlapping of binding sequences for steroid hormone receptors and DNaseI hypersensitive sites in the rabbit uteroglobin gene region. Nucleic Acids Res 15:4535-4552 

Rider V, Bullock DW 1988 Progesterone-dependent binding of a trans-acting factor to the uteroglobin promoter. Biochem Biophys Res Commun 156:1368-1375 

Rider V, Peterson CJ 1991 Activation of uteroglobin gene expression by progesterone is modulated by uterine-specific promoterbinding proteins. Mol Endocrinol 5:911-920 

Kleis-SanFrancisco S, Hewetson A, Chilton BS 1993 Prolactin augments progesterone-dependent uteroglobin gene expression by modulating promoter-binding proteins. Mol Endocrinol 7:214-223 

DeMayo FJ, Damak S, Hansen TN, Bullock DW 1991 Expression and regulation of the rabbit uteroglobin gene in transgenic mice. Mol Endocrinol 5:311-318 

Wolf M, Klug J, Hackenberg R, Gessler M, Grzeschik K-H, Beato M, Suske G 1992 Human CC10, the homologue of rabbit uteroglobin: genomic cloning, chromosomal localization and expression in endometrial cell lines. Hum Mol Genet 1:371-378 

Ding Y-Q, Zhu L-J, Bagchi MK, Bagchi IC 1994 Progesterone stimulates calcitonin gene expression in the uterus during implantation. Endocrinology 135:2265-2274 

Krisinger J, Setoyama T, Leung PCK1994 Expression of calbindinD9k in the early pregnant rat uterus: effects of RU 486 and correlation to estrogen receptor mRNA. Mol Cell Endocrinol 102:15-22 

L'Horset F, Blin C, Brehier A, Thomasset M, Perret C 1993 Estrogen-induced calbindin-D9k gene expression in the rat uterus during the estrous cycle: late antagonistic effect of progesterone. Endocrinology 132:489-495 

Thorburn GD, Challis JRG 1979 Endocrine control of parturition. Physiol Rev 59:863-918 

Abel MH, Baird DT 1980 The effect of 17β-estradiol and progesterone on prostaglandin productionby human endometrium maintained in organ culture. Endocrinology 106:1599-1606 

Kelly RW, Healy DL, Cameron MJ, Cameron IT, Baird DT 1986 The stimulation of prostaglandin production by two antiprogesterone steroids in human endometrial cells. J Clin Endocrinol Metab 62:1116-1123 

Sheldrick EL, Flick-Smith HC 1993 Effect of ovarian hormones on oxytocin receptor concentrations in explants of uterus from ovariectomized ewes. J Reprod Fertil 97:241-245 

Lau TM, Kerton DJ, Gow CB, Fairclough RJ 1993 Role of progesterone in the control of endometrial oxytocin receptors at luteolysis in sheep. J Reprod Fertil 98:229-233 

Schirar A, Capponi A, Catt KJ 1980 Regulation of uterine angiotensin II receptors by estrogen and progesterone. Endocrinology 106:5-12 

Potvin W, Varma DR 1991 Down-regulation of myometrial atrial natriuretic factor receptors by progesterone and pregnancy and up-regulation by oestrogen in rats. J Endocrinol 131:259-266 

Vivat V, Cohen-Tannoudji J, Revelli J-P, Muzzin P, Giacobino J-P, Maltier J-P, Legrand C 1992 Progesterone transcriptionally regulates the |82-adrenergic receptor gene in pregnant rat myometrium. J Biol Chem 267:7975-7978 

Mulvihill ER, LePennec JP, Chambon P 1982 Chicken oviduct progesterone receptor: location of specific regions of high-affinity binding in cloned DNA fragments of hormone-responsive genes. Cell 28:621-632 

McReynolds L, O'Malley BW, Nisbet AD, Fothergill JE, Givol D, Fields S, Robertson M, Brownlee GG 1978 Sequence of chicken ovalbumin mRNA. Nature 273:723-728 

O'Malley BW, Roop DR, Lai EC, Nordstrom JL, Catterall JF, Swaneck GE, Colbert DA, Tsai M-J, Dugaiczyk A, Woo SLC 1979 The ovalbumin gene: organization, structure, transcription and regulation. Recent Prog Horm Res 35:1-46 

Pastorcic M, Bagchi MK, Tsai SY, Tsai M-J, O'Malley BW 1989 Multiple protein binding sites within the ovalbumin gene 5'-flanking region: isolation and characterization of sequence-specific binding proteins. Nucleic Acids Res 17:6693-6711 

Nordstrom LA, Dean DM, Sanders MM 1993 A complex array of double-stranded and single-stranded DNA-binding proteins mediates induction of the ovalbumin gene by steroid hormones. J Biol Chem 268:13193-13202 

Dean DC, Knoll BJ, Riser ME, O'Malley BW 1983 A 5'-flanking sequence essential for progesterone regulation of an ovalbumin fusion gene. Nature 305:551-554 

Dean DC, Gope R, Knoll BJ, Riser ME, O'Malley BW 1984 A similar 5 '-flanking region is required for estrogen and progesterone induction of ovalbumin gene expression. J Biol Chem 259:9967-9970 

Gaub M-P, Bellard M, Scheuer I, Chambon C, Sassone-Corsi P 1990 Activation of the ovalbumin gene by the estrogen receptor involves the fos-jun complex. Cell 63:1267-1276 

Subramaniam M, Harris SA, Rasmussen K, Spelsberg TC 1993 Rapid down-regulation of c-jun protooncogene transcription by progesterone in the avian oviduct. Endocrinology 133:2049-2054 

Topper YJ, Freeman CS 1980 Multiple hormone interactions in the developmental biology of the mammary gland. Physiol Rev 60: 1049-1106 

Murphy LJ, Murphy LC, Stead B, Sutherland RL, Lazarus L 1986 Modulation of lactogenic receptors by progestins in cultured human breast cancer cells. J Clin Endocrinol Metab 62:280-287 

Djiane J, Durand P 1977 Prolactin-progesterone antagonism in self-regulation of prolactin receptors in the mammary gland. Nature 266:641-643 

Nardulli AM, Katzenellenbogen BS 1988 Progesterone receptor regulation in T47D human breast cancer cells: analysis by density labeling of progesterone receptor synthesis and degradation and their modulation by progestin. Endocrinology 122:1532-1540 

ReadLD, Snider CE, Miller JS, Greene GL, Katzenellenbogen BS 1988 Ligand-modulated regulation of progesterone receptor messenger ribonucleic acid and protein in human breast cancer cell lines. Mol Endocrinol 2:263-271 

Alexander IE, Clarke CL, Shine J, Sutherland RL 1989 Progestin inhibition of progesterone receptor gene expression in human breast cancer cells. Mol Endocrinol 3:1377-1386 

Masters JRW, Drife JO, Scarsbrick JJ 1977 Cyclic variation of DNA synthesis in human breast epithelium. J Natl Cancer Inst 58:1263-1265 

Meyer JS 1977 Cell proliferation in normal human breast ducts, fibroadenomas, and other ductal hyperplasias measured by nuclear labeling with tritiated thymidine. Hum Pathol 8:67-81 

Anderson TJ, Battersby S, King RJB, McPherson K, Going JJ 1989 Oral contraceptive use influences resting breast proliferation. Hum Pathol 20:1139-1144 

Williams G, Anderson E, Howell A, Watson R, Coyne J, Roberts SA, Potten CS 1991 Oral contraceptive (OCP) use increases proliferation and decreases oestrogen receptor content of epithelial cells in the normal human breast. Int J Cancer 48:206-210 

Going JJ, Anderson TJ, Battersby S, Macintyre CCA 1988 Proliferative and secretory activity in human breast during natural and artifical menstrual cycles. Am J Pathol 130:193-204 

Anderson TJ, Ferguson DJP, Raab GM 1982 Cell turnover in the "resting" human breast: influence of parity, contraceptive pill, age and laterality. Br J Cancer 46:376-382 

Longacre TA, Bartow SA 1986 A correlative morphologic study of human breast and endometrium in the menstrual cycle. Am J Surg Pathol 10:382-393 

Vorherr H 1974 The Breast. Morphology, Physiology and Lactation. Academic Press, New York Chang K-J, Lee TTY, Linares-Cruz G, Fournier S, de Lignieres B Influences of percutaneous administration of estradiol and progesterone on human breast epithelial cell cycle in vivo. Fertil Steril 63:785-791 

van Bogaert LJ 1978 Effect of hormone on human mammary duct in vivo. Horm Metab Res 10:337-340 

McManus MJ, Welsch CW 1984 The effect of estrogen, progesterone, thyroxine, and human placental lactogen on DNA synthesis of human breast ductal epithelium maintained in athymic nude mice. Cancer 54:1920-1927 

Laidlaw IJ, Clarke RB, Howell A, Owen AWMC, Potten CS, Anderson E 1995 The proliferation of normal human breast tissue implanted into athymic nude mice is stimulated by estrogen but not progesterone. Endocrinology 136:164-171 

Gompel A, Malet C, Spritzer P, Lalardrie J-P, Kutten F, MauvaisJarvis P 1986 Progestin effect on cell proliferation and 17ß-hydroxysteroid dehydrogenase activity in normal human breast cells in culture. J Clin Endocrinol Metab 63:1174-1180 

Wong MSJ, Murphy LC 1991 Differential regulation of c-myc by progestins and antiestrogens in T-47D human breast cancer cells. J Steroid Biochem Mol Biol 39:39-44 

Musgrove EA, Lee CS, Sutherland RL 1991 Progestins both stimulate and inhibit breast cancer cell cycle progression while increasing expression of transforming growth factor a, epidermal growth factor receptor, c-fos, and c-myc genes. Mol Cell Biol 11:5032-5043 

Musgrove EA, Hamilton JA, Lee CS, Sweeney KJ, Watts CK, Sutherland RL 1993 Growth factor, steroid, and steroid antagonist regulation of cyclin gene expression associated with changes in T-47D human breast cancer cell cycle progression. Mol Cell Biol 13:3577-3587 

Hurd C, Khattree N, Alban P, Nag K, Jhanwar SC, Dinda S, Moudgil VK1995 Hormonal regulation of the p53 tumor suppressor protein in T47D human breast carcinoma cell line. J Biol Chem 270:28507-28510 

Murphy LC, Murphy LJ, Dubik D, Bell GI, Shiu RPC 1988 Epidermal growth factor gene expression in human breast cancer cells: regulation of expression by progestins. Cancer Res 48:4555-4560 

Murphy LJ, Sutherland RL, Stead B, Murphy LC, Lazarus L 1986 Progestin regulation of epidermal growth factor receptor in human mammary carcinoma cells. Cancer Res 46:728 -734 

Murphy LC, Murphy LJ, Shiu RPC 1988 Progestin regulation of EGF-receptor mRNA accumulation in T-47D human breast cancer cells. Biochem Biophys Res Commun 150:192-196 

Murphy LC, Dotzlaw H 1989 Regulation of transforming growth factor a and transforming growth factor ß messenger ribonucleic acid abundance in T-47D human breast cancer cells. Mol Endocrinol 3:611-617 

Murphy LC, Dotzlaw H, Wong MSJ, Miller T, Murphy LJ 1991 Mechanisms involved in the evolution of progestin resistance in human breast cancer cells. Cancer Res 51:2051-2057 

Papa V, Reese CC, Brunetti A, Vigneri R, Siiteri PK, Goldfine ID 1990 Progestins increase insulin receptor content and insulin stimulation of growth in human breast carcinoma cells. Cancer Res 50:7858-7862 

Horwitz KB, Freidenberg GR 1985 Growth inhibition and increase of insulin receptors in antiestrogen-resistant T47DCO human breast cancer cells by progestins: implications for endocrine therapies. Cancer Res 45:167-173 

Goldfine ID, Papa V, Vigneri R, Siiteri P, Rosenthal S 1992 Progestin regulation of insulin and insulin-like growth factor-I receptors in cultured human breast cancer cells. Breast Cancer Res Treat 22:69-79 

Owens PC, Gill PG, DeYoung NJ, Weger MA, Knowles SE, Moyse KJ 1993 Estrogen and progesterone regulate secretion of insulin-like growth factor binding proteins by human breast cancer cells. Biochem Biophys Res Commun 193:467-473 

Coutts A, Murphy LJ, Murphy LC 1994 Expression of insulin-like growth factor binding proteins by T-47D human breast cancer cells: regulation by progestins and antiestrogens. Breast Cancer Res Treat 32:153-164 

Figueroa JA, Yee D 1992 The insulin-like growth factor binding proteins (IGFBPs) in human breast cancer. Breast Cancer Res Treat 22:81-90 

Yee D 1994 The insulin-like growth factor system as a target in breast cancer. Breast Cancer Res Treat 32:85-95 

LeRoith D, Werner H, Beitner-Johnson D, Roberts CT 1995 Molecular and cellular aspects of the insulin-like growth factor I receptor. Endocr Rev 16:143-163 

Papa V, Hartmann KKP, Rosenthal SM, Maddux BA, Siiteri PK, Goldfine ID 1991 Progestins induce down-regulation of insulinlike growth factor-I (IGF-I) receptors in human breast cancer cells: potential autocrine role of IGF-II. Mol Endocrinol 5:709-717 

Lieberherr M, Acker GM, Grosse B, Pesty A, Balsan S 1984 Rat endometrial cells in primary culture: effects and interaction of sex hormones and vitamin D3 metabolites on alkaline phosphatase. Endocrinology 115:824-829 

Holinka CF, Hata H, Kuramoto H, Gurpide E 1986 Effects of steroid hormones and antisteroids on alkaline phosphatase activity in human endometrial cancer cells (Ishikawa line). Cancer Res 46:2771-2774 

Di Lorenzo D, Albertini A, Zava D 1991 Progestin regulation of alkaline phosphatase in the human breast cancer cell line T47D. Cancer Res 51:4470-4475 

Di Lorenzo D, Gianni M, Savoldi GF, Ferrari F, Albertini A, Garattini E1993 Progesterone induced expression of alkaline phosphatase is associated with a secretory phenotype in T47D breast cancer cells. Biochem Biophys Res Commun 192:1066-1072 

Chalbos D, Westley B, May F, Alibert C, Rochefort H 1986 Cloning of cDNA sequences of a progestin-regulated mRNA from MCF-7 human breast cancer cells. Nucleic Acids Res 14:965-982 

Chalbos D, Chambon M, Ailhaud G, Rochefort H 1987 Fatty acid synthetase and its mRNA are induced by progestins in breast cancer cells. J Biol Chem 262:9923-9926 

Chalbos D, Rochefort H 1984 A 250-kilodalton cellular protein is induced by progestins in two human breast cancer cell lines MCF-7 and T-47D. Biochem Biophys Res Commun 121:421-427 

Joyeux C, Rochefort H, Chalbos D 1989 Progestin increases gene transcription and messenger ribonucleic acid stability of fatty acid synthetase in breast cancer cells. Mol Endocrinol 4:681-686 

Chambon M, Rochefort H, Vial H, Chalbos D 1989 Progestins and androgens stimulated lipid accumulation in T47D breast cancer cells via their own receptors. J Steroid Biochem 33:915-922 

Chalbos D, Joyeux C, Galtier F, Rochefort H 1992 Progestininduced fatty acid synthetase in human mammary tumours: from molecular to clinical studies. J Steroid Biochem Mol Biol 43:223-228 

Houdbine L-M, Teyssot B, Devinoy E, Ollivier-Bousquet M, Djiane J, Kelly PA, Delouis C, Kann G, Fevre J 1983 Role of progesterone in the development and the activity of the mammary gland. In: Bardin CW, Milgrom E, Mauvais-Jarvis P (eds) Progesterone and Progestins. Raven Press, New York, pp 297-319 

Sitruk-Ware R, Varin C, Clair F, Maugis E-L, Fermanian J, Mauvais-Jarvis P 1985 In vivo effects of progestins on prolactin secretion. J Clin Endocrinol Metab 60:575-578 

Quirk SJ, Gannell JE, Fullerton MJ, Funder JW 1985 Progestins specifically suppress α-lactalbumin synthesis and secretion. J Steroid Biochem 23:901-905 

Qasba PK, Safaya SK 1984 Similarity of the nucleotide sequences of rat α-lactalbumin and chicken lysozyme genes. Nature 308:377380 

Quirk SJ, Funder JW 1988 Steroid receptors, and the generation of closely coupled/biphasic dose-response curves. J Steroid Biochem 30:9-15 

Quirk SJ, Gannell JE, Funder JW 1988 α-lactalbumin production by rat mammary gland: prepartum emergence of resistance to glucocorticoids and progestins. Mol Cell Endocrinol 58:183-189 

Haslam SZ, Shyamala G 1980 Progesterone receptors in normal mammary gland: receptor modulations in relation to differentiation. J Cell Biol 86:730-737 

Rosen JM, O'Neal DL, McHugh JE, Comstock JP 1978 Progesterone-mediated inhibition of casein mRNA and polysomal casein synthesis in the rat mammary gland during pregnancy. Biochemistry 17:290-297 

Lee CS, Oka T 1992 A pregnancy-specific mammary nuclear factor involved in the repression of the mouse β-casein gene transcription by progesterone. J Biol Chem 267:5797-5801 

Parsons B, MacLusky NJ, Krey L, Pfaff DW, McEwen BS 1980 The temporal relationship between estrogen-inducible progestin receptors in the female rat brain and the time course of estrogen activation of mating behaviour. Endocrinology 107:774-779 

DonCarlos LL, Greene GL, Morrell JI 1989 Estrogen plus progesterone increases progestin receptor immunoreactivity in the brain of ovariectomized guinea pigs. Neuroendocrinology 50:613-623 

Rainbow TC, McGinnis MY, Krey LC, McEwen BS 1982 Nuclear progestin receptors in rat brain and pituitary. Neuroendocrinology 34:426-432 

Blaustein JD, Turcotte JC 1990 Down-regulation of progestin receptors in guinea pig brain: new findings using an immunocytochemical technique. J Neurobiol 21:675-685 

Parsons B, McGinnis MY, McEwen BS 1981 Sequential inhibition of progesterone: effects on sexual receptivity and associated changes in brain cytosol progestin binding in the female rat. Brain Res 221:149-160 

Mani SK, Blaustein JD, Allen JM, Law SW, O'Malley BW, Clark JH 1994 Inhibition of rat sexual behavior by antisense oligonucleotides to the progesterone receptor. Endocrinology 135:1409-1414 

Canonaco M, O'Connor LH, Pfaff DW, McEwen BS 1989 Longer term progesterone treatment induces changes of GABAA receptor levels in forebrain sites in the female hamster: quantitative autoradiography study. Exp Brain Res 77:407-411 

Maggi A, Perez G 1984 Progesterone and estrogens in rat brain: modulation of GABA (y-aminobutyric acid) receptor activity. Eur J Pharmacol 103:165-168 

DeBold JF, Frye CA 1994 Genomic and non-genomic actions in the control of female hamster sexual behaviour. Horm Behav 28:445453 

El-Etr M, Akwa Y, Fiddes RJ, Robel P, Baulieu E-E 1995 A progesterone metabolite stimulates the release of gonadotropin-releasing hormone from GT1-1 hypothalamic neurons via the y-aminobutyric acid type A receptor. Proc Natl Acad Sci USA 92:37693773 

Vincent PA, Etgen AM 1993 Steroid priming promotes oxytocininduced norepinephrine release in the ventromedial hypothalamus of female rats. Brain Res 620:189-194 

Wilcox J, Roberts JL 1985 Estrogen decreases rat hypothalamic proopiomelanocortin messenger RNA levels. Endocrinology 117: 2392-2396 

Collado ML, Rodriguez-Manzo G, Cruz ML 1985 Effect of progesterone upon adenylate cyclase activity and cAMP levels on brain areas. Pharmacol Biochem Behav 23:501-504 

Gereau RW, Kedzie KA, Renner KJ 1993 Effect of progesterone on serotonin turnover in rats primed with estrogen implants into the ventromedial hypothalamus. Brain Res Bull 32:293-300 

Maggi A, Zucchi I, Perez J 1985 Progesterone in rat brain: modulation of β-adrenergic receptor activity. Pharmacol Res Commun 17:283-291 

Kato J, Hirata S, Nozawa A, Yamada-Mouri N 1994 Gene expression of progesterone receptor isoforms in the rat brain. Horm Behav 28:454-463 

Kraus WL, Montano MM, Katzenellenbogen BS 1993 Cloning of the rat progesterone receptor 5'-region and identification of two functionally distinct promoters. Mol Endocrinol 7:1603-1616 

Kraus WL, Montano MM, Katzenellenbogen BS 1994 Identification of multiple, widely spaced estrogen-responsive regions in the rat progesterone receptor gene. Mol Endocrinol 8:952-969 

Eriksen EF, Colvard DS, Berg NJ, Graham ML, Mann KG, Spelsberg TC, Riggs BL 1988 Evidence of estrogen receptors in normal human osteoblast-like cells. Science 241:84-86 

Turner RT, Colvard DS, Spelsberg TC 1990 Estrogen inhibition of periosteal bone formation in rat long bones: down-regulation of gene expression for bone matrix proteins. Endocrinology 127:1346-1351 

Turner RT, Backup P, Sherman PJ, Hill E, Evans GL, Spelsberg TC 1992 Mechanism of action of estrogen on intramembranous bone formation: regulation of osteoblast differentiation and activity. Endocrinology 131:883-889 

Nomura S, Hashmi S, McVey JH, Ham J, Parker M, Hogan BLM 1989 Evidence for positive and negative regulatory elements in the 5'-flanking sequence of the mouse spare (osteonectin) gene. J Biol Chem 264:12201-12207 

Beresford JN, Gallagher JA, Poser JW, Russell RGG 1984 Production of osteocalcin by human bone cells in vitro. Effects of 1,25(OH)2D3, parathyroid hormone, and glucocorticoids. Metab Bone Dis Relat Res 5:229-234 

Gronowicz GA, McCarthy M-B 1995 Glucocorticoids inhibit the attachment of osteoblasts to bone extracellular matrix proteins and decrease |81-integrin levels. Endocrinology 136:598-608 

Oldberg A, Jirskog-Hed B, Axelsson S, Heinegard D 1989 Regulation of bone sialoprotein mRNA by steroid hormones. J Cell Biol 109:3183-3186 

Prior JC 1990 Progesterone as a bone-trophic hormone. Endocr Rev 11:386-398 

Hsueh AJW, Peck Jr EJ, Clark JH 1975 Progesterone antagonism of the estrogen receptor and estrogen-induced uterine growth. Nature 254:337-339 

Mester I, Martel D, Psychoyos A, Baulieu E-E 1974 Hormonal control of estrogen receptor in uterus and receptivity for ovoimplantation in the rat. Nature 250:776-778 

Hsueh AJ, Peck Jr EJ, Clark JH 1976 Control of uterine estrogen receptor levels by progesterone. Endocrinology 98:438 -444 

Tseng L, Gurpide E 1975 Effects of progestins on estradiol receptor levels inhuman endometrium. J Clin Endocrinol Metab 41:402-404 

Takeda A, Leavitt WW 1986 Progestin-induced downregulation of nuclear estrogen receptor in uterine decidual cells: analysis of receptor synthesis and turnover by the density-shift method. Biochem Biophys Res Commun 135:98-104 

Read LD, Greene GL, Katzenellenbogen BS 1989 Regulation of estrogen receptor messenger ribonucleic acid and protein levels in human breast cancer cell lines by sex steroid hormones, their antagonists, and growth factors. Mol Endocrinol 3:295-304 

Alexander IE, Shine J, Sutherland RL 1990 Progestin regulation of estrogen receptor messenger RNA in human breast cancer cells. Mol Endocrinol 4:821-828 

Savouret JF, Rauch M, Redeuilh G, Sar S, Chauchereau A, Woodruff K, Parker MG, Milgrom E 1994 Interplay between estrogens, progestins, retinoic acid and AP-1 on a single regulatory site in the progesterone receptor gene. J Biol Chem 269:28955-28962 

Kraus WL, Weis KE, Katzenellenbogen BS 1995 Inhibitory crosstalk between steroid hormone receptors: differential targeting of estrogen receptor in the repression of its transcriptional activity by agonistand antagonist-occupied progestin receptors. Mol Cell Biol 15:1847-1857 

Chalbos D, Galtier F 1994 Differential effect of forms A and B of human progesterone receptor on estradiol-dependent transcription. J Biol Chem 269:23007-23012 

Horwitz KB, Jackson TA, Bain DL, Richer JK, Takimoto GS, Tung L 1996 Nuclear receptor coactivators and repressors. Mol Endocrinol 10:1167-1177 

Alkhalaf M, Murphy LC 1992 Regulation of c-jun and jun-B by progestins in T-47D human breast cancer cells. Mol Endocrinol 6:1625-1633