Elsevier

Steroids

Volume 65, Issues 10–11, October–November 2000, Pages 571-577
Steroids

Progesterone receptors in reproduction: functional impact of the A and B isoforms

https://doi.org/10.1016/S0039-128X(00)00115-XGet rights and content

Abstract

Progesterone (P) is a key regulator of female reproductive activity. The effects of P are mediated by two progesterone receptor (PR) proteins, termed A and B, that arise from a single gene and act as ligand-activated transcription factors to regulate the expression of reproductive target genes. Null mutation of the PR gene in mice (PRKO) leads to pleiotropic reproductive abnormalities. This paper will review the reproductive functions of PRs delineated using the PRKO mouse. Further, we will summarize the structure and functional properties of PRs and discuss how functional differences between the PR-A and PR-B isoforms are likely to impact on the overall physiological role of the receptor in reproductive systems.

Introduction

Progesterone is an essential regulator of the reproductive events associated with the establishment and maintenance of pregnancy, including ovulation, uterine and mammary gland development, and the neurobehavioral expression associated with sexual responsiveness [1], [2]. The physiological effects of progesterone are mediated by interaction with specific intracellular proteins termed progesterone receptors (PRs). Recently, we demonstrated that the PR is absolutely essential for the female reproductive activities associated with progesterone by generating null mutant mice in which the expression of the PR gene was ablated [2]. These mice displayed pleiotropic reproductive abnormalities that underscored the critical role of PR in mammalian reproduction.

The PR is a transcription factor and a member of a large family of structurally related gene products known as the nuclear receptor superfamily [3], [4], [5], [6]. The PR is composed of two protein isoforms, termed A and B. Both isoforms are expressed from a single gene in rodents and humans as a result of transcription from two alternative promoters and translation initiation at two different AUG codons [7], [8]. The selective physiological roles of the two isoforms of PR are predicted to differ based on different structural and functional properties of the individual proteins observed using in vitro assay systems. In the following sections, we will summarize the structural and functional significance of the PR isoforms and will highlight the functional differences between the two proteins observed in vitro. Then, we will review the physiological role of PRs in reproduction and discuss how the functional differences between the two PR isoforms observed in vitro are likely to impact on the physiological role of the progesterone in reproductive systems.

Section snippets

Structure and functional properties of PRs

Binding of progesterone to PRs induces a significant conformational change on the proteins [9], [10] that results in: 1) homodimerization of two ligand receptor complexes [11], [12], [13], 2) increased receptor phosphorylation [14], [15]; 3) binding of receptor dimers to specific hormone responsive DNA elements located in the promoter regions of target genes [4], [16]; and 4) interaction of the receptor complex with specific coactivator proteins and general transcripton factors [17], [18] to

Structural and functional significance of the PR-A and -B protein isoforms

The PR-A and -B isoforms are expressed in a number of vertebrate species, including humans and rodents [43], [44], [45], [46], [47]. Two distinct promoters have been identified that are independently regulated and give rise to the A and B isoforms [7], [8]. The absolute levels of PR as well as the stoichiometric ratio of PR-A to PR-B in reproductive tissues vary as a consequence of developmental and hormonal status. The conservation of these two receptor isoforms and the complex genomic

Physiological role of PRs

To directly address the role of PRs in reproductive function, we previously generated a mouse model (PRKO) in which both isoforms of the PR were ablated by PR gene targeting. This model has provided definitive proof that PRs are essential coordinators of all reproductive events that together culminate in preparation for and maintenance of pregnancy [2]. In the following summary of the physiological role of PRs, we will focus on those aspects of progesterone-dependent reproductive function that

PRs and ovarian function

Evidence for a direct role of PRs in ovarian function has been supported in recent years by the demonstration that 1) the antiprogestin RU486 inhibits ovulation [56] and 2) that luteinizing hormone (LH), the primary signal for rupture of preovulatory ovarian follicles, can stimulate transient expression of PR mRNAs and both protein isoforms in granulosa cells isolated form preovulatory follicles [57], [58], [59]. Definitive proof that PRs are essential mediators of ovulation has been provided

PR and uterine development

The uterus is composed of heterogeneous cell types that undergo continuous synchronized waves of proliferation and differentiation in response to the cyclical rise and fall of estrogen and progesterone. PRs are expressed in the epithelial, stromal, and myometrial compartments of the uterus, and their spatiotemporal expression in these compartments is controlled by estrogen [1], [61], [62]. The primary proliferative stimulus in the epithelial compartment is estrogen [63], [64], [65], while

PR and mammary gland development

Progesterone and estrogen are the principle steroid hormones involved in normal breast development and tumorigenesis [71], [72]. Early ductal outgrowth observed postnatally is strongly controlled by estrogen acting through the ERα receptor localized in the stromal compartment of the mammary gland [73]. During pregnancy, increased branching, alveolar proliferation, and differentiation of alveolar lobules is controlled primarily by progesterone. Studies with the PRKO mouse [2] have confirmed

PR and sexual behavior

The preoptic areas, ventromedial nucleus (VMNH) of the hypothalmus and amygdala, have long been associated with a female sexual response exhibited by rodents, termed lordosis, and these areas express PRs [87], [88]. The lordosis response is characterized by a concave arching of the back with rump and head elevated to facilitate copulation by the male. The VMNH is primarily responsible for coordinating this sexual behavior with reproductive condition in order to optimize the chances for

Summary

Analysis of the reproductive phenotypes of the PRKO mouse has provided unequivocal evidence of the central role of the PR proteins in the coordination of all aspects of reproductive function. Our recent generation of new mutant strains of mice, in which the PR-A or -B proteins have been selectively ablated, now permits us to address the specific contributions of each PR isoform to the overall physiological role of progesterone. Preliminary phenotypic analysis of these mice has already provided

References (91)

  • S.E. Fawell et al.

    Characterization and colocalization of steroid binding and dimerization activities in the mouse estrogen receptor

    Cell

    (1990)
  • E. Vegeto et al.

    The mechanism of RU486 antagonism is dependent on the conformation of the carboxy-terminal tail of the human progesterone receptor

    Cell

    (1992)
  • W.B. Pratt et al.

    A region in the steroid binding domain determines formation of the non-DNA binding GS glucocorticoid receptor complex

    J Biol Chem

    (1988)
  • C.A. Sartorius et al.

    Antagonist-occupied human progesterone receptors bound to DNA are functionally switched to transcriptional agonists by cAMP

    J Biol Chem

    (1993)
  • W.T. Schrader et al.

    Progesterone-binding components of chick oviduct IV. Characterization of purified subunits

    J Biol Chem

    (1972)
  • D.P. McDonnell et al.

    RU486 exerts antiestrogenic activities through a novel progesterone receptor A form- mediated mechanism

    J Biol Chem

    (1994)
  • W.L. Kraus et al.

    Determinants for the repression of estrogen receptor transcriptional activity by ligand-occupied progestin receptors

    J Steroid Biochem Mol Biol

    (1997)
  • V.C. Jordan et al.

    Effect of estradiol benzoate, tamoxifen and monohydroxytamoxifen on immature rat uterine progesterone receptor synthesis and endometrial cell division

    J Steroid Biochem

    (1979)
  • P. Sicinski et al.

    Cyclin D1 provides a link between development and oncogenesis in the retina and breast

    Cell

    (1995)
  • R.Q.J. Schaapveld et al.

    Impaired mammary gland development and function in mice lacking LAR receptor-like tyrosine phosphate activity

    Dev Biol

    (1997)
  • C.A. Lange et al.

    Convergence of progesterone and epidermal growth factor signaling in breast cancer. Potentiation of mitogen-activated protein kinase pathways

    J Biol Chem

    (1998)
  • J.K. Richer et al.

    Convergence of progesterone with growth factor and cytokine signaling in breast cancer

    J Biol Chem

    (1998)
  • R.L. Sutherland et al.

    Regulation of breast cancer cell cycle progression by growth factors, steroids and steroid antagonists

    J Steroid Biochem Mol Biol

    (1992)
  • N.J. Maclusky et al.

    Progestin receptors in the developing rat brain and pituitary

    Brain Res

    (1980)
  • A.H. Lauber et al.

    Steroid control of higher brain functionsgene expression for estrogen and progesterone receptor mRNAs in rat brain and possible relations to sexually dimorphic functions

    J Steroid Biochem Mol Biol

    (1991)
  • C.L. Clarke et al.

    Progestin regulation of cellular proliferation

    Endocrine Rev

    (1990)
  • J.P. Lydon et al.

    Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities

    Genes Dev

    (1995)
  • R.M. Evans

    The steroid and thyroid hormone receptor superfamily

    Science

    (1988)
  • M.-J. Tsai et al.

    Molecular mechanisms of action of steroid/thyroid receptor superfamily members

    Annu Rev Biochem

    (1994)
  • B.W. O’Malley et al.

    Minirevieworphan receptors: in search of a unifying hypothesis for activation

    Mol Endocrinol

    (1992)
  • P. Kastner et al.

    Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B

    EMBO J

    (1990)
  • W.L. Kraus et al.

    Cloning of the rat progesterone receptor gene 5′-region and identification of two functionally distinct promoters

    Mol Endocrinol

    (1993)
  • G.F. Allan et al.

    Hormonal induced conformational changes in the progesterone receptor are required for events following binding to DNA

    Proc Natl Acad Sci USA

    (1992)
  • N.L. Weigel

    Receptor phosphorylation

  • N.L. Weigel et al.

    Phosphorylation and progesterone receptor function

  • H. Gronemeyer

    Transcription activation by estrogen and progesterone receptors

    Annu Rev Genet

    (1991)
  • S.A. Onate et al.

    Sequence and characterization of a coactivator for the steroid hormone receptor superfamily

    Science

    (1995)
  • L. Tora et al.

    The N-terminal region of the chicken progesterone receptor specifies target gene activation

    Nature (London)

    (1988)
  • C.A. Sartorius et al.

    A third transactivation function (AF3) of human progesterone receptors located in the unique N-terminal segment of the B-isoform

    Mol Endocrinol

    (1994)
  • B.F. Luisi et al.

    Crystallographic analysis of the interaction of the glucocortioid receptor with DNA

    Nature (London)

    (1991)
  • L.P. Freedman

    Anatomy of the steroid receptor zinc finger region

    Endocrine Rev

    (1992)
  • D. Picard et al.

    Two signals mediate hormone-dependent nuclear localization of the glucocorticoid receptor

    EMBO J

    (1987)
  • L.A. Denner et al.

    Regulation of progesterone receptor-mediated transcription by phosphorylation

    Science

    (1990)
  • S.M. Aronica et al.

    Progesterone receptor regulation in uterine cellsstimulation by estrogen, cyclic adenosine 3′,5′-monophosphate, and insulin-like growth factor I and suppression by antiestrogens and protein kinase inhibitors

    Endocrinology

    (1991)
  • S.M. Aronica et al.

    Stimulation of estrogen receptor-mediated transcription and alteration in the phosphorylation state of the rat uterine estrogen receptor by estrogen, cyclic adenosine monophosphate, and insulin-like growth factor-1

    Mol Endocrinol

    (1993)
  • Cited by (186)

    • Progesterone control of myometrial contractility

      2023, Journal of Steroid Biochemistry and Molecular Biology
    • Progress in progestin-based therapies for neurological disorders

      2021, Neuroscience and Biobehavioral Reviews
    • Progesterone and breast

      2020, Best Practice and Research: Clinical Obstetrics and Gynaecology
    • SAR based in-vitro anticholinesterase and molecular docking studies of nitrogenous progesterone derivatives

      2020, Steroids
      Citation Excerpt :

      Although progesterone is predominantly a female sex hormone that plays a key role in menstrual cycle and maintenance of pregnancy. It has been shown to influence spermiogenesis, sperm capacitation/acrosome reaction and testosterone biosynthesis in the Leydig cells [1,2]. Many experimental models showed that progesterone exerts protective effects in pathogenic aspects of brain dysfunction, e.g. Alzheimer’s disease.

    View all citing articles on Scopus
    View full text