1-s2.0-s1084952107000717-main

Upload: ayluy

Post on 03-Jun-2018

219 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/12/2019 1-s2.0-S1084952107000717-main

    1/11

  • 8/12/2019 1-s2.0-S1084952107000717-main

    2/11

    322 S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331

    progesterone withdrawal and estrogen activation initiate parturi-

    tion by transforming the myometrium to a highly contractile and

    excitable state. This review focuses on recent progress in under-

    standing of how progesterone maintains myometrial relaxation

    for most of pregnancy and how estrogens promote a contractile

    state. We also examine recent data regarding the mechanism for

    progesterone withdrawal and estrogen activation in human par-

    turition and how these key events are mediated, coordinated and

    controlled.

    2. The relaxatory actions of progesterone

    Progesterone affects myometrial contractility through

    genomic and non-genomic pathways. Genomic pathways func-

    tion by altering the expression of specific contraction associated

    genes to modulate the long-term contractile phenotype. Non-

    genomic pathways in contrast are more rapid and directly affect

    the contractile machinery by modulating intracellular signal

    transduction pathways.

    2.1. Genomic actions of progesterone

    Genomic actions of progesterone are mediated by the classic

    nuclear progesterone receptors (nPRs) that function as ligand-

    activated transcription factors. The human nPR gene encodes

    two major products, the full-length PR-B and the truncated (by

    164 N-terminal amino acids) PR-A, under the control of two

    separate promoters [25]. Other proteins generatedby exondele-

    tions and intronic insertions have also been reported (see[6]for

    review); however, their physiological roles are uncertain. Mul-

    tiple in vitro studies suggest that PR-B is the principal mediator

    of genomic progesterone actions. PR-A also modulates the tran-scription of some genes; however, it mainly acts to repress the

    transcriptional activity of PR-B [79]. Theextent to which PR-A

    decreases PR-B activity depends on its amount relative to PR-

    B. Thus, genomic progesterone responsiveness is determined

    by the dual and opposing actions of PR-A and PR-B, and is

    inversely related to the PR-A/PR-B ratio.

    The induction of labor and delivery by treatment with

    nPR antagonists such as RU486, reflects the importance of

    nPR-mediated progesterone actions for the maintenance of

    myometrial relaxation during pregnancy. The principal genomic

    mechanism by which progesterone represses myometrial con-

    tractility is by modulating the expression of genes encoding

    contraction-associated proteins (CAPs). Some important CAPsinclude the oxytocin receptor (OTXR) and the prostaglandin

    (PG)-F2 (PGF2) receptor (FP), the gap-junction protein

    connexin-43 (Cx43) and the PG-metabolizing enzyme 15-

    hydroxy-PG-dehydrogenase (PGDH). Progesterone decreases

    myometrial OT and PGF2responsiveness by inhibiting OTXR

    and FP expression, respectively [1014]. In pregnant rats,

    removal of endogenous progesterone by ovariectomy or inhi-

    bition of progesterone action with RU486 treatment increases

    myometrial OTXR expression and OT responsiveness. Pro-

    gesterone decreases myometrial OTXR levels indirectly by

    inhibiting estrogen-induced OTXR expression [15,16]. Inter-

    estingly, progesterone also inhibits stretch-induced myometrial

    OTXR expression in rats [17,18], suggesting that it represses

    multiple pathways that induce OTXR expression. Adminis-

    tration of RU486 [19] or epostane [20,21] (a progesterone

    synthesis inhibitor) to pregnant women at early and mid-

    gestation increases the effectiveness of PG treatment to induce

    labor, indicating that progesterone represses PG responsive-

    ness. Importantly, progesterone also increases the inactivation

    of PGs by increasing expression of PGDH in the myometrium

    and chorion[2225].

    Progesterone also decreases the development of coordinated

    uterine contractions by inhibiting expression of Cx43, a major

    component of myometrial gap-junctions that serve to synchro-

    nize contractions over the entire uterus. Expression of Cx43 in

    the human pregnancy myometrium increases with the onset of

    labor[26], and in human myometrial cell cultures its expres-

    sion is up-regulated by estrogen and inhibited by progesterone

    [27,28]. Studies in rats showed that progesterone not only

    decreases expression of Cx43 but also its translocation through

    the Golgi and its assembly into functional gap-junctions at the

    plasma membrane[29].Thus, progesterone decreases contrac-tile capacity by inhibiting Cx43 expression and gap-junction

    formation.

    Progesterone also augments activity of the cAMP/protein

    kinase-A (PK-A) signaling cascade in myometrial cells. The

    cAMP/PK-A pathway promotes smooth muscle relaxation in

    part by PK-A-mediated inhibition of the phospholipase C

    (PLC)/Ca2+ pathway (for reviewsee [30]; seealso Lopez Bernal,

    this issue;Sanborn,this issue). Activityof PK-A is modulatedby

    its association with A-kinase anchoring proteins (AKAPs) that

    localize PK-A to specific intracellular compartments and facili-

    tate its capacity to phosphorylate specific targets, especially PLC

    [3133]. In rats, labor is preceded by a decrease in PK-A associ-ation with the AKAP complex[34],and progesterone treatment

    prevents the parturition-related PK-A/AKAP decline [35]. Thus,

    progesterone, through its effects on the PK-A/AKAPinteraction,

    augments PK-A-mediated inactivation of PLC. This opposes

    the capacity for stimulatory uterotonins such as OT and PGF2to increase intracellular Ca2+ levels, via the PLC/Ca2+ path-

    way. That this action of progesterone was inhibited by RU486

    suggests that it is nPR-mediated[35].

    Thus, progesterone via its interactions with nPRs, maintains

    myometrial relaxation by: (1) directly inhibiting CAP expres-

    sion; (2) decreasing estrogen-induced CAP expression, and (3)

    increasing the effectiveness of PK-A to inhibit PLC activity. In

    vitro studies suggest that these actions are most likely medi-ated by PR-B in the human pregnancy myometrium[3638].

    However, in PR-B-knockout mice, PR-A alone is sufficient to

    mediate the pro-gestational actions of progesterone [39,40], sug-

    gesting that species diversity exists in the role of the two nPRs

    in pregnancy and parturition.

    2.2. Non-genomic actions of progesterone

    Non-genomic actions of progesterone are characterized by:

    (1) a rapid time-course for response with a latency of min-

    utes, rather than hours; (2) no requirement for nPR activity

    or occupancy; (3) no requirement for RNA or protein synthe-

  • 8/12/2019 1-s2.0-S1084952107000717-main

    3/11

    S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331 323

    sis; and (4) occurrence in response to conjugated progesterone

    (e.g., progesterone-BSA) that cannot enter the target cell. These

    effects are potentially mediated by the interaction of proges-

    terone with specific mPRs that are coupled to intracellular

    signaling pathways; activation of Src/MAPK cascade intracel-

    lular signaling pathways by ligand activated nPRs, and/or by

    progesterone interaction with neurotransmitter and peptide hor-

    mone receptors (e.g., GABAAand OTXR).

    Studies examining the rapid effects of progesterone on iso-

    lated myometrial strips from various species generally showed

    that it inhibits OT-induced contractions and that it uncouples

    the excitationcontraction process (for review see [41]).How-

    ever, in vitro studies using human pregnancy myometrium

    yielded mixed results with some investigators reporting a rapid

    relaxatory effect of progesterone and progesterone metabolites

    [4246]while others reporting that progesterone augments con-

    traction frequencybut decreases duration, amplitude and activity

    area in term myometrial strips[4749].The reason for this vari-

    ability is not readily apparent but could be due to difference in

    theprogestins used andhow they were prepared,and thecontrac-tile state of the tissue before it was mounted on the myograph.

    Nonetheless, the studies clearly demonstrated that progesterone

    has a rapid non-genomic affect on myometrial contractility.

    Several in vivo studies support the hypothesis that proges-

    terone non-genomically influences myometrial contractility. In

    one of the first clinical trials of progestin tocolysis, Hendricks

    et al. [50] found that administration of a large bolus of pro-

    gesterone into the amniotic fluid of women at term decreased

    the frequency of spontaneous contractions and attenuated OT

    responsiveness. The tocolytic effect of intra-amniotic proges-

    terone therapy was rapid and in some women persisted for

    several days. However, the number of subjects was small andthe data were somewhat anecdotal. In the mid-1960s Pinto et al.

    [51]re-addressed the issue and reported that large amounts of

    progesterone(100200 mg bolus iv; this is almost as much as the

    placenta produces in 24 h at term) administeredto women in term

    labor inhibited the frequency and intensity of uterine contrac-

    tions within minutes. In one woman, progesterone completely

    silenced the laboring uterus within 10 minutes of administra-

    tion. In two other women, progesterone inhibited spontaneous

    contractions but failed to decrease the intensity of contractions

    elicited by OT. Those data demonstrated direct non-genomic

    relaxatory actions of progesterone at high doses; however, study

    cohorts were small. Importantly, Pinto and colleagues later

    found the same effects of progesterone on isolated myometrialstrips[46].

    Two recent clinical studies reported that administration of

    moderate doses of progesterone (100 mg daily by vaginal

    suppository) or 17-hydroxyprogesterone caproate (250 mg intra-

    muscular injection in oil weekly) starting at 16 weeks gestation

    to women at high risk for preterm birth reduced the incidence

    of preterm birth and improved neonatal outcome[52,53].The

    mechanism by which progestin supplementation decreased the

    rate of preterm birth is uncertain. Interestingly, da Fonseca

    et al. [52] reported that women receiving progesterone via

    vaginal suppositories and who presented with preterm labor

    responded more favorably than women in the placebo group

    to tocolytic treatment with -mimetics. It appeared that pro-

    longed exposure of the myometrium to exogenous progestin

    improved the effectiveness of tocolytic therapy. This was also

    observed by Chanrachakul et al. [42] who reported that in

    isolated term myometrial strips, progesterone, albeit at high

    doses, decreased contractility and augmented the capacity for

    ritodrine, a -mimetic commonly used for tocolytic therapy,

    to block OT-induced contractions. They concluded that the

    mechanism for this action is through a non-genomic path-

    way, as it occurred soon after progesterone exposure. However,

    using a similar approach, Sexton et al. [54] reported that 17-

    hydroxyprogesterone caproate had no effect on spontaneous

    or OT-induced contractions in myometrial strips. They sug-

    gested that 17-hydroxyprogesterone-caproate affects the rate

    of preterm birth via long-term genomic affects rather than by

    direct non-genomic mechanisms. Clearly, further studies are

    needed to determine the mechanism by which progestin treat-

    ment decreases the incidence of preterm birth and in particular

    whether this occurs through genomic or non-genomic pathways.

    Several groups have reported that progesterone interacts withthe OTXR and that this interaction decreases contractility by

    inhibitingOT-induced inositol triphosphate production and Ca2+

    mobilization [5557]. However, this effect was species-specific;

    Grazzini et al. [57]reported that progesterone interacted with

    the rodent but not with the human OTXR. The human OTXR

    instead interacted with 5-dihydroprogesterone, a 5 reduced

    progesterone metabolite. This observation is consistent with

    myograph studies showing that 5-dihydroprogesterone, but

    not progesterone or 5-reduced progesterone metabolites, is a

    potent myometrial relaxant that decreases basal and OT-induced

    contractile activity[45,5860].Thus, progesterone could relax

    the myometrium in an intracrine manner through its conversionto 5-dihydroprogesterone. Sheehan et al. [61] found that in

    human pregnancy, circulating levels of 5-dihydroprogesterone

    and expression of the 5-reductase enzyme in the placenta

    and myometrium decrease in association with the onset of

    labor. Mitchell et al.[62]also found that the human pregnancy

    myometrium expresses the 5-reductase enzyme and has the

    capacity to generate 5-dihydroprogesterone.However,whether

    5-dihydroprogesterone interacts with the human OTXR is con-

    troversial, as others [63,64] could not confirm the outcome

    reported by Grazzini et al. [57] and instead reported that the

    human OTXR does not bind 5-dihydroprogesterone. This sug-

    gests that 5-reduced metabolites of progesterone relax the

    myometrium by interacting with other receptors. One possibilityis that the gamma butyric acid-A (GABAA) receptor is involved.

    5-Dihydroprogesterone binds to the GABAAreceptor and this

    interaction is in part responsible for the anesthetic effect of

    these steroids [65]. Putnam et al. [66] found that in the rat

    myometrium, a GABAA-specific antagonist blocked inhibition

    of contractility by progesterone and its 5-reduced metabo-

    lites suggesting that the relaxatory actions of those steroids

    were mediated by the GABAA receptor. Importantly, GABAAreceptors have been identified in the human uterus [67].Thus,

    progesterone metabolites, especially-reduced forms,may non-

    genomically relax the myometrium by interacting directly with

    the OTXR or with the GABAA receptor.

  • 8/12/2019 1-s2.0-S1084952107000717-main

    4/11

    324 S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331

    Non-genomic actions of progesterone can also be mediated

    by nPRs. PR-A and PR-B have proline-rich motifs in their N-

    termini that, upon ligand binding, interact with the Src tyrosine

    kinaseat the plasmamembrane to activate the Ras/Raf-1/MAPK

    pathway [68,69]. Src is a key intermediate that couples hormone

    signals at the plasma membrane with intracellular transduction

    pathways involved in regulating a variety of cellular processes

    including proliferation, differentiation, adhesion, migration, and

    apoptosis[68].This may be an important mechanism by which

    progesterone exerts tropic action on the pregnancy myometrium.

    Whether this pathway affects contractility is uncertain. Putnam

    et al. [66] found that RU486 reversed the rapid inhibition of

    spontaneous contractility by progesterone in myometrial strips

    from non-pregnant rats, suggesting that non-genomic relaxatory

    actions of progesterone involve nPRs.

    A number of unique membrane associated PRs (mPRs) have

    recently been identified and characterized. Progesterone recep-

    tor membrane component-1 and -2 (PGRMC1 and PGRMC2)

    have single transmembrane spanning domains and interact with

    plasminogen activator inhibitor of RNA binding-1 (PAIRBP1),which recruits the mPRs to form a multimeric progesterone-

    binding complex on the plasmamembrane [7076]. In granulosa

    cells, binding of progesterone to the PGRMCPAIRBP1 com-

    plex activates protein kinase G and decreases intracellular Ca2+

    levels [77]. The role of these mPRs in the onset/process of

    labor and whether the human pregnancy myometrium expresses

    PAIRBP1 is not known at this stage.

    mPR,mPR and mPR are structurally related to G-protein

    coupled receptors, having a typical seven-transmembrane

    domain structure [78,79]. mPR and mPR are coupled

    to inhibitory G-proteins and therefore, upon ligand binding

    decrease intracellular cAMP levels [80]. Several studies haveshown that mPR and - are expressed in the human pregnancy

    myometrium, whereas mPR expression is barely detectable

    [8082]. Karteris et al. [80] found that ligand activation of

    mPR and mPRin primary cultures of human term myome-

    trial cells decreased cAMP levels and increased phosphorylation

    of myosin. They proposed that these effects (decreased cAMP

    and activation of myosin) augment contractility. However, they

    also found that mPRand mPRincreased the transcriptional

    activity of ligand-activated PR-B, which would be expected

    to decrease contractility via the genomic pathway. To recon-

    cile those opposing activities, they proposed that for most of

    pregnancy the genomic actions of PR-B dominate to relax the

    myometrium, and that mPR and mPR augment this pathwayby augmenting PR-B activity. At parturition, functional proges-

    terone withdrawal (see below) negates PR-B actions, allowing

    non-genomic actions mediated by mPR and - to prevail,

    and therefore increase contractility by decreasing cAMP and

    increasing myosin phosphorylation. This model proposes that

    for most of pregnancy non-genomic and genomic actions of

    progesterone conspire to relax the myometrium, whereas non-

    genomic pathways mediated by mPRand mPRprevail after

    functional progesterone withdrawal and promote contraction.

    This may explain reports of progesterone increasing contractil-

    ity of isolated strips of term human myometrium[49,83].This

    would occur if the tissue was procured after genomic proges-

    terone withdrawal. The idea of functional cross-talk between

    the genomic and non-genomic pathways and the notion that

    progesterone switches from promoting relaxation to enhancing

    contraction are novel concepts that warrant further investigation.

    However,Krietschet al. [84] haverecentlysuggested thatmPR,

    -and -are not activated by progesterone and do not localize

    to the plasma membrane. Clearly, further studies are needed to

    confirm the model proposed by Karteris et al. and determine the

    roles of mPRand -in the pregnancy myometrium.

    3. Transformation to a contractile phenotype

    Transformation of the myometrium from a relaxed to a

    highly contractile state is an early and key event in the

    parturition process. The biochemical and physical changes

    include: increased Cx43 expression leading to increased cou-

    pling between myocytes so that contractions are synchronized

    across the whole uterus; increased sensitivity and contrac-

    tile responsiveness to stimulatory uterotonins such as OT and

    PGF2 due respectively to increased OTXR and FP expres-sion; increased production of PGs by the gestational tissues

    and decreased inactivation of PGs in the myometrium; low-

    ered threshold for myocyte excitability; and decreased capacity

    for the cAMP/PK-A signaling pathway to maintain relaxation.

    These events are controlled primarily by the combined effects

    of progesterone withdrawal and estrogen activation.

    3.1. Progesterone withdrawal

    In most animals the onset of labor is preceded by a fall in

    circulating maternal progesterone levels (i.e., a systemic pro-

    gesterone withdrawal). In some species this is due to decreasedplacental progesterone secretion (e.g., sheep), while in others

    (e.g., rabbit, mouse, rat) it is caused by regression of the corpus

    luteum (CL)[8588]. The mechanism by which progesterone

    withdrawal increases myometrial contractility is not clearly

    understood. As mentioned above, studies with the progesterone

    antagonist RU486 demonstrate that inhibition of nPR-mediated

    progesterone action induces the full parturition cascade. This

    suggests that withdrawal of genomic progesterone actions is a

    key parturition-triggering event. Whether withdrawal of non-

    genomic progesterone actions, including those mediated by

    nPRs, is also required for myometrial transformation is uncer-

    tain.

    Themechanism for progesterone withdrawal in human partu-rition is uncertain. Unlike most other species, labor and delivery

    in humans occur without a decrease in maternal, fetal and

    amniotic fluid progesterone levels[8991],suggesting that pro-

    gesterone withdrawal is not necessary. However, as RU486

    treatment induces labor at all stages of human pregnancy, it is

    generally considered that human parturition involves a form of

    progesterone withdrawal that does not depend on a decrease in

    circulating progesterone levels. Proposed mechanisms include:

    (1) sequestration of free active progesterone by a circulating

    progesterone binding protein; (2) intracrine inactivation of local

    progesterone bioactivity by myometrial cells; (3) production

    of an endogenous progesterone antagonist; and (4) decreased

  • 8/12/2019 1-s2.0-S1084952107000717-main

    5/11

    S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331 325

    myometrial progesterone responsiveness (i.e., a functional pro-

    gesterone withdrawal) mediated by changes in the levels of

    specific nPRs or nPR co-activator/co-repressors (for review see

    [92,93]). Indirect evidence for each of these mechanisms has

    been reported but their definitive roles in human parturition

    remain uncertain.

    Several research groups, including ours, have tested the

    hypothesis that functional progesterone withdrawal is mediated

    by specific changes in myometrial nPR expression. Studies of

    nPR protein[37,38,94]and mRNA[95]levels in myometrial

    biopsies indicated that human parturition involves an increase

    in the myometrial PR-A/PR-B ratio due to increased expression

    of PR-A. Functional studies in myometrial cells showed that

    an increase in the PR-A/PR-B ratio decreased genomic proges-

    terone responsiveness mediated by PR-B [37,38]. We found that

    PR-A and PR-B were exclusively expressed in myocytes in the

    human pregnancy uterus and that the PR-A/PR-B protein ratio

    was 0.5 (a PR-B-dominant state) at around 30 weeks, increased

    to 1.0 (equal amounts of PR-A and PR-B) at term before labor

    onset and increased further to around 3 (a PR-A-dominant state)in laboring myometrium[37].The pregnancy stage- and labor-

    associated increase in the PR-A/PR-B ratio was almost identical

    to data reported by Haluska et al. [94] in the rhesus monkey

    (Fig. 1),a species that also lacks a systemic progesterone with-

    drawal at parturition. Another truncated nPR, known as PR-C,

    that also represses PR-B activity, has been found to increase

    in fundal myometrium in association with labor at term [36].

    Thus, studies so far suggest that functional progesterone with-

    drawal in human parturition is mediated by an increase in the

    myometrial PR-A (or PR-C)/PR-B ratio and that regionalization

    exists in the uterus such that progesterone responsiveness is dif-

    ferentially regulated in fundal (by PR-C) and lower segment (byPR-A) myometrium.

    Functional progesterone withdrawal could also be medi-

    ated by the inhibition of nPR interaction with target DNA.

    nPR binding to nuclear extracts of term human decidua is

    reduced in laboring compared with non-laboring tissue indi-

    cating that the onset of labor involves changes in the nPR

    Fig. 1. Comparison of the PR-A/PR-B protein ratio in human and rhesus mon-

    key pregnancy myometrium. *P < 0.001 (Adapted from Merlino et al.[37]and

    Haluska et al.[94]).

    transcriptional complex[96].In the myometrium, labor is asso-

    ciated with a decline in specific nPR co-activators, particularly

    cAMP-response element-binding protein-binding protein and

    steroid receptor coactivators-2 and -3 [97]. The reduction in

    co-activators may decrease histone acetylation that effectively

    closes chromatin around the progesterone response element,

    making it inaccessible to the nPR transcriptional complex. Such

    a scenario would explain the decrease in nPR binding to nuclear

    response elements in decidual cells[96].As a variation on this

    theme, Dong et al.[98]identified a protein known as polypirim-

    idine tract-binding protein-associated splicing factor (PTB) that

    specifically inhibits nPR transactivation and whoseexpression in

    rat myometrium increasesat term. They proposed that this factor

    contributes to functional progesterone withdrawal by acting as

    an additional nPR co-repressor. Interestingly, PTB also controls

    the splicing of myosin phosphatase targeting protein mRNAs,

    and therefore the functional activity of myosin phosphatase, a

    key determinant of smooth muscle contractility [99]. These data

    demonstrate the complexity that underlies the genomic actions

    of progesterone on myometrial contractility and the multiplelevels at which functional progesterone withdrawal could occur.

    3.2. Estrogen activation

    In 1967, Pinto et al. [46] examined the role of estrogens

    in human parturition by administrating a large amount of

    17-estradiol (200 mg intravenously in 1 h) to non-laboring

    pregnant women at term. They found that estradiol treatment

    increased uterine contractility and OT responsiveness within

    46 h and accelerated the time to delivery. Those findings were

    consistent with the stimulatory actions of estrogens on myome-

    trial contractility and showed that the progesterone block isnot absolute and can be overcome by estrogenic drive. Stud-

    ies in rats and sheep also demonstrated that treatment with

    estradiol at mid-gestation induces preterm labor [100102].

    Interestingly, Pinto et al.[46]also found that administration of

    progesterone rapidly (within 10 minutes) blocked the stimula-

    tory actions induced by 17-estradiol, supporting the hypothesis

    that progesterone acts non-genomically to promote relaxation.

    In the rhesus monkey, Nathanielsz et al. found that augment-

    ing placental estrogen production (by the administration of

    androstenedione, which is readily converted to estrogens by

    the placenta) increases myometrial contractility and OT respon-

    siveness and induces preterm birth [103,104]. Inhibition of

    aromatase activity eliminated the induction of parturition byandrostenedione, confirming that its conversion to estrogens was

    essential for the initiation of parturition[105].However, others

    found that estradiol treatment alone had no effect on parturition

    in the rhesus monkey even though circulating estradiol levels

    weremarkedlyelevated [106]. Those inconsistent outcomes sug-

    gest that local production of estrogens from androgen precursor

    is more important than circulating estrogen levels. Further stud-

    ies are needed to resolve this controversy. Nevertheless, data so

    far demonstrate the critical role of estrogenic drive (i.e., estrogen

    activation) for myometrial transformation to a contractile state.

    In most species estrogen activation is mediated by an increase

    in circulating estrogen levels and is coordinated with systemic

  • 8/12/2019 1-s2.0-S1084952107000717-main

    6/11

    326 S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331

    progesterone withdrawal[85].However, in humans (and higher

    primates) circulating estrogens increase at around mid-gestation

    and continue to rise gradually until birth[8991].This has led

    to the concept that estrogen activation in human parturition,

    as with progesterone withdrawal, is mediated at the functional

    level, by an increase in myometrial estrogen responsiveness.

    Thus, for most of pregnancy the human myometrium appears

    to be refractory to estrogens at least in terms of CAP gene

    expression, and parturition involves functional estrogen acti-

    vation whereby the myometrium becomes estrogen-responsive.

    Refractoriness of the myometrium to estrogens for most of

    pregnancy is likely due to very low levels of ER and ER.

    Importantly, we found that ER expression is low in non-

    laboring term myometrium and increases in association with

    labor onset, suggesting that functional estrogen activation is

    mediated by increased ER expression[95].We also found that

    ERmRNA levels correlate with the PR-A/PR-B mRNA ratio

    [95]indicating a functional link between the nPR and ER sys-

    tems. This association is consistent with studies in a variety

    of species showing that progesterone decreases uterine estrogenresponsiveness by decreasing ER expression [107110]. In the

    pregnant rhesus monkey, treatment with RU486 at mid-gestation

    increased myometrial ER expression indicating that proges-

    terone decreases ER expression through an nPR-mediated

    process[111].Taken together the current data suggest that pro-

    gesterone via its interaction with PR-B inhibits myometrial ER

    expression and causes the myometrium to be refractory to circu-

    lating estrogens. The increase in myometrial PR-A expression

    with advancing gestation decreases PR-B transcriptional activ-

    ity and eventually eliminates the PR-B-mediated inhibition of

    ER expression. According to this model (Fig. 2) functional

    progesterone withdrawal, mediated by increased PR-A, induces

    functional estrogen activation mediated by increased expression

    of ER and therefore coordinates these critical parturition-

    triggering events. Circulating estrogens can then transform the

    myometrium to a contractile state. This paradigm implies that

    a fundamental mechanism by which progesterone maintains

    myometrial relaxation in human pregnancy is by blocking the

    stimulatory actions of estrogens and that functionalprogesterone

    withdrawal induces functional estrogen activation. This phys-

    iologic interaction explains why inhibition of nPR-mediated

    progesterone actions triggers the full parturition cascade, espe-

    ciallyas estrogensare readily available to acton themyometrium

    for most of human pregnancy. It also implies that human par-

    turition is triggered by any event (e.g., local PGs, myometrial

    stretch, inflammation) that increases myometrial PR-A expres-sion and induces functional progesterone withdrawal. Thus,

    multiple parturition trigger pathways may converge on myome-

    trial PR-A expression. A more detailed understanding at the

    molecular level of how the myometrial progesterone-nPR and

    estrogen-ER signaling pathways interact and are regulated

    in human pregnancy may reveal novel targets to therapeuti-

    Fig. 2. Schematic model of the genomic and non-genomic pathwaysby whichsteroid hormonesaffect contractility of the human pregnancy myometrium. For most of

    pregnancy progesterone, via its interaction with PR-B in myometrial cells, inhibits expression of CAP genes and decreases responsiveness to estrogens by inhibiting

    ER expression. Progesterone and/or its metabolite 5-dihydroprogesterone also exert non-genomic effects on the pregnancy myometrium by interacting with a

    variety of mPRs. The effects of non-genomic progesterone actions on myometrial contractility are not well understood but it is generally considered that progesterone

    decreases contractility via this pathway. At parturition PR-A expression increases until the PR-A/PR-B ratio reaches a point whereby the relaxatory actions mediated

    through PR-B are repressed, i.e., functional progesterone withdrawal. As a consequence ERexpression increases and the myometrium becomes more responsive

    to circulating estrogens which increase CAP expression and transform the myometrium to a highly contractile and excitable phenotype leading to the onset of labor.

  • 8/12/2019 1-s2.0-S1084952107000717-main

    7/11

    S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331 327

    cally control human labor and address the problem of preterm

    birth.

    3.3. Genomics of myometrial transformation

    Determination of the human genome sequence has led to the

    development of novel and cutting-edge technologies to charac-

    terize the relationship between global gene transcriptional status

    (i.e., the transcriptome) and phenotype in cells and tissues speci-

    mens. Techniques such as suppression subtractive hybridization

    [112] and multidimensional cDNA and oligonucleotide array

    platforms [113117] have been used to determine changes

    in the myometrial transcriptome, encompassing thousands of

    genes, that may account for its contractile transformation at

    parturition (for review see [118]). The fundamental hypothe-

    sis tested by these powerful techniques is that transformation

    of the pregnancy myometrium from quiescence to contraction

    involves changes in specific gene expression in myometrialcells.

    Although many of these genes have been identified using more

    conventional assay techniques, the global approach incorporatesthe entire transcriptome and therefore provides the opportu-

    nity to reveal the complete spectrum of genes involved. This

    approach generates novel hypotheses, which can then be tested

    using regular approaches.

    Studies so far have used a variety of platforms to compare the

    transcriptome in myometrial biopsy specimens obtained before

    and after the onset of labor at term [113116] and from the

    fundal and lower uterine segments [117].Most analyses iden-

    tified multiple genes whose expression changed (increased or

    decreased by at least twofold) in association with the onset

    of labor, however, there was little overlap between the cohorts

    of genes identified in different studies. There was also markedvariation between some studies. Notably, Havelock et al. [117]

    foundlittle difference in gene expression profiles between fundal

    and lower uterine segment and in association with labor onset,

    whereas Charpigny et al.[115]found that labor was associated

    with a down-regulation of a large number of developmental-,

    cell adhesion- and proliferation-related genes and a concomitant

    increase in inflammatory- and contraction-associated genes. The

    differences between study outcomes likely stem from variabil-

    ity due to methodological factors related to the array platform,

    the number of samples studied in each of the experimental

    groups and the purity of the biopsy samples with respect to

    myometrial cell content (biopsy specimens contain multiple and

    varied cell types that contribute to total RNA content of theextract). These problems will likely be overcome as the array

    technology improves and price of microarrays decreases, allow-

    ing more replicates of laboring and non-laboring specimens to

    be performed. Thus, at this stage the body of data regarding the

    relationship between the myometrial transcriptome and contrac-

    tile phenotype is inconclusive and no specific labor-associated

    genomic pathways have been identified.

    Interpretation of outcomes from array-based studies largely

    depends on the existing knowledge-base of specific gene

    function and the power of the computational and statistical tech-

    niques to analyze and align gene expression data, which often

    comprises large numbers of genes with unknown function, into

    functional networks. Advances in clustering and hierarchical

    techniques have allowed for the organization of gene expres-

    sion data into functional networks; however, many of the genes

    identified as altered by labor onset have unknown functions and

    their relationship to phenotype is ambiguous at best. Clearly,

    this will be less of a problem as understanding of individual

    genes and gene networks advances.The useof multidimensional

    statistical analyses, such as directed graphs and principal com-

    ponent analysis, have been applied to determine how theoretical

    causal pathways generated by microarray data sets interact to

    impact on phenotype[119,120].This approach utilizes proba-

    bilistic and statistical analyses of the expression data to develop

    hierarchical causal pathways for a particular phenotype or con-

    dition. For example, our analysis of qRT-PCR-based data-sets

    using directed graphs suggested a causal relationship between

    the activation of inflammatory pathways and functional proges-

    terone withdrawal [119] whereby inflammation precedes and

    possibly causes functional progesterone withdrawal. That con-

    clusion is consistent with our earlier study using the PHM1-31

    immortalized human pregnancy myometrial cell line in whichwe found that PGF2 preferentially increases PR-A expres-

    sion[121].Thus, locally produced PGs may initiate myometrial

    transformation and the onset of labor by first inducing func-

    tional progesterone withdrawal via increased myometrial PR-A

    expression. This approach exemplifies the advantage of inte-

    grating outcomes from specific cause-effect studies (e.g., from

    animal models, cell lines, tissue specimens and clinical studies),

    multidimensional cDNA/oligonucleotide array studies and com-

    putational analyses of theoretical causal pathways in an iterative

    and informative paradigm to unravel the physiology of human

    parturition.

    4. Conclusions

    In an evolutionary context, physiological triggers for partu-

    rition would have been subjected to strong selective pressures

    so that the timing for birth favors species survival by optimizing

    neonatal outcome and minimizing risks to themother (and there-

    fore future pregnancies) imposed by the pregnant and parturient

    states. This perspective helps explain the remarkable diversity

    in gestation length/birth timing and parturition control across

    viviparous species. In contrast, the physiologic systems that

    establish and maintain pregnancy exhibit relatively little diver-

    sity. In this case a single hormone, progesterone (and possibly

    its metabolites) maintains pregnancy and promotes myometrialrelaxation through a combination of genomic and non-genomic

    mechanisms. In fact, theprogesterone block appears to be a com-

    mon trait among viviparous species. As pregnancy advances,

    stimulatory influences build up to progressively challenge the

    progesterone block. Although the principal stimulatory drive to

    myometrial contractility is imparted by estrogens (a trait that

    also appears to be conserved), other important factors such as

    uterine stretch, myometrial response to the intrauterine cytokine

    milieu and the activity of a fetal and/or placenta-based physi-

    ologic clock mechanism, are also involved. The combination

    of estrogenic and other physiologic stimulators for parturition

    and the level of co-operativity, functional overlap and redun-

  • 8/12/2019 1-s2.0-S1084952107000717-main

    8/11

    328 S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331

    dancy between them, appear to be species-specific and were

    likely selected based on their overall impact on reproductive

    efficiency. Parturition initiates when the combined effect of all

    stimulatory influences overcomes the progesterone block. Thus,

    progesterone and estrogens play central roles in pregnancy and

    parturition and the mechanisms that control and coordinate their

    actions on the pregnancy myometrium are critical processes in

    the physiology of birth timing.

    Acknowledgements

    Dr. Mesiano is supported by grants from the March of Dimes

    Birth Defects Foundation (#6-FY05-68) and the National Insti-

    tutes of Health (HD051563).

    References

    [1] Csapo A. Progesterone block. Am J Anat 1956;98:27391.

    [2] Chauchereau A, Savouret JF, Milgrom E. Control of biosynthesis

    and post-transcriptional modification of the progesterone receptor. Biol

    Reprod 1992;46:1747.

    [3] Bouchard P. Progesterone and the progesterone receptor. J Reprod Med

    1999;44:1537.

    [4] Giangrande PH, McDonnell DP. The A and B isoforms of the human

    progesterone receptor: two functionally different transcription factors

    encoded by a single gene. Rec Prog Horm Res 1999;54:291313.

    [5] Conneely OM, Lydon JP. Progesterone receptors in reproduction: Func-

    tional impact of the A and B isoforms. Steroids 2000;65:5717.

    [6] Hirata S, Shoda T, Kato J, Hoshi K. Isoform/variant mRNAs for

    sex steroid hormone receptors in humans. Trends Endocrinol Metab

    2003;14:1249.

    [7] Giangrande PH, Kimbrel EA, Edwards DP, McDonnell DP. The opposing

    transcriptional activities of the two isoforms of the human proges-

    terone receptor are due to differential cofactor binding. Mol Cell Biol

    2000;20:310215.

    [8] Tung L, Mohamed MK, Hoeffler JP, Takimoto GS, Horwitz KB.

    Antagonist-occupied human progesterone B-receptors activate tran-

    scription without binding to progesterone response elements and are

    dominantly inhibited by A-receptors. Mol Endocrinol 1993;7:125665.

    [9] Vegeto E, Shahbaz MM, Wen DX, Goldman ME, OMalley BW,

    McDonnell DP. Human progesterone receptor A form is a cell- and

    promoter-specific repressor of human progesterone receptor B function.

    Mol Endocrinol 1993;7:124455.

    [10] Brodt-Eppley J, Myatt L. Changes in expression of contractile FP and

    relaxatory EP2 receptors in pregnant rat myometrium during late gesta-

    tion, at labor, and postpartum. Biol Reprod 1998;59:87883.

    [11] Cook JL, Shallow MC, Zaragoza DB, Anderson KI, Olson DM. Mouse

    placental prostaglandins are associated with uterine activation and the

    timing of birth. Biol Reprod 2003;68:57987.

    [12] Dong YL, Yallampalli C. Pregnancy and exogenous steroid treatmentsmodulate the expression of relaxant EP(2) and contractile FP receptors

    in the rat uterus. Biol Reprod 2000;62:5339.

    [13] Fang X, Wong S, Mitchell BF. Effects of RU486 on estrogen, proges-

    terone, oxytocin, and their receptors in the rat uterus during late gestation.

    Endocrinology 1997;138:27638.

    [14] Ou CW, Chen ZQ, Qi S, Lye SJ. Expression and regulation of the mes-

    senger ribonucleic acid encoding the prostaglandin F(2alpha) receptor in

    the rat myometrium during pregnancy and labor. Am J Obstet Gynecol

    2000;182:91925.

    [15] Soloff MS, Fernstrom MA, Periyasamy S, Soloff S, Baldwin S, Wieder

    M. Regulation of oxytocin receptor concentration in rat uterine explants

    by estrogen and progesterone. Can J Biochem Cell Biol 1983;61:62530.

    [16] Adachi S, Oku M. The regulation of oxytocin receptor expres-

    sion in human myometrial monolayer culture. J Smooth Muscle Res

    1995;31:17587.

    [17] Fuchs AR, Periyasamy S, Soloff MS. Systemic and local regulation of

    oxytocin receptors in the rat uterus, and their functionalsignificance. Can

    J Biochem Cell Biol 1983;61:61524.

    [18] OuCW,Chen ZQ,Qi S, Lye SJ.Increasedexpression of therat myometrial

    oxytocin receptor messenger ribonucleic acid during labor requires both

    mechanical and hormonal signals. Biol Reprod 1998;59:105561.

    [19] Swahn ML, Bygdeman M. The effect of the antiprogestin RU 486 on

    uterine contractility and sensitivity to prostaglandin and oxytocin. Br J

    Obstet Gynaecol 1988;95:12634.[20] Selinger M, MacKenzie IZ, Gillmer MD, Phipps SL, Ferguson J. Proges-

    terone inhibitionin mid-trimestertermination of pregnancy: physiological

    and clinical effects. Br J Obstet Gynaecol 1987:94.

    [21] Webster MA, Phipps SL, Gillmer MD. Interruption of first trimester

    human pregnancy following epostane therapy. Effect of prostaglandin

    E2 pessaries. Br J Obstet Gynaecol 1985;92:9638.

    [22] Challis JR, Patel FA, Pomini F. Prostaglandin dehydrogenase and the

    initiation of labor. J Perinat Med 1999;27:2634.

    [23] Patel FA, Challis JR. Cortisol/progesterone antagonism in regulation of

    15-hydroxysteroid dehydrogenase activity and mRNA levels in human

    chorion and placental trophoblast cells at term. J Clin Endocrinol Metab

    2002;87:7008.

    [24] Patel FA, Funder JW, Challis JR. Mechanism of cortisol/progesterone

    antagonism in the regulation of 15-hydroxyprostaglandin dehydroge-

    nase activity and messenger ribonucleic acid levels in human chorionand placental trophoblast cells at term. J Clin Endocrinol Metab

    2003;88:292233.

    [25] Patel FA, Sun K, Challis JR. Local modulation by 11beta-hydroxysteroid

    dehydrogenase of glucocorticoid effects on the activity of 15-

    hydroxyprostaglandin dehydrogenase in human chorion and placental

    trophoblast cells. J Clin Endocrinol Metab 1999;84:395400.

    [26] Garfield RE, Hayashi RH. Appearance of gap junctions in the

    myometrium of women during labor. Am J Obstet Gynecol 1981;140:

    25460.

    [27] Di WL, Lachelin GC, McGarrigle HH, Thomas NS, Becker DL. Oestriol

    and oestradiol increase cell to cell communication and connexin43

    protein expression in human myometrium. Mol Hum Reprod 2001;7:

    6719.

    [28] Zhao K, Kuperman L, Geimonen E, Andersen J. Progestin represses

    human connexin43 gene expression similarly in primary cultures of

    myometrial and uterine leiomyoma cells. Biol Reprod 1996;54:60715.

    [29] Hendrix EM,Myatt L, Sellers S, Russell PT, LarsenWJ. Steroid hormone

    regulation of rat myometrial gapjunction formation: effects oncx43 levels

    and trafficking. Biol Reprod 1995;52:54760.

    [30] Sanborn BM, Ku CY, Shlykov S, Babich L. Molecular signaling through

    G-protein-coupled receptors and the control of intracellular calcium in

    myometrium. J Soc Gynecol Investig 2005;12:47987.

    [31] Colledge M, Scott JD. AKAPs: from structure to function. Trends Cell

    Biol 1999;9:21621.

    [32] Edwards AS, Scott JD. A-kinase anchoring proteins: protein kinase A

    and beyond. Curr Opin Cell Biol 2000;12:21721.

    [33] Feliciello A, Gottesman ME, Avvedimento EV. The biological functions

    of A-kinase anchor proteins. J Mol Biol 2001;308:99114.

    [34] Ku CY, Word RA, Sanborn BM. Differential expression of protein kinase

    A, AKAP79, and PP2B in pregnant human myometrial membranes prior

    to and during labor. J Soc Gynecol Investig 2005;12:4217.

    [35] KuCY, SanbornBM. Progesteronepreventsthe pregnancy-relateddecline

    in proteinkinase A association withrat myometrial plasma membraneand

    A-kinase anchoring protein. Biol Reprod 2002;67:6059.

    [36] Condon JC, Hardy DB, Kovaric K, Mendelson CR. Up-regulation of

    the progesterone receptor (PR)-C isoform in laboring myometrium

    by activation of nuclear factor-kappaB may contribute to the onset

    of labor through inhibition of PR function. Mol Endocrinol 2006;20:

    76475.

    [37] Merlino AA, Welsh TN, Tan H, Yi LJ, Cannon V, Mercer BM, et al.

    Nuclear progesterone receptors in the human pregnancy myometrium:

    evidence that parturition involves functional progesterone withdrawal

    mediated by increased expression of progesterone receptor-A. J Clin

    Endocrinol Metab 2007;92:192733.

  • 8/12/2019 1-s2.0-S1084952107000717-main

    9/11

    S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331 329

    [38] Pieber D, Allport VC, Hills F, Johnson M, Bennett PR. Interaction

    between progesterone receptor isoforms in myometrial cells in human

    labour. Mol Hum Reprod 2001;7:8759.

    [39] Mulac-Jericevic B, Mullinax RA, DeMayo FJ, Lydon JP, Conneely OM.

    Subgroup of reproductive functions of progesterone mediated by proges-

    terone receptor-B isoform. Science 2000;289:17514.

    [40] Shyamala G, Yang X, Silberstein G, Barcellos-Hoff MH, Dale E. Trans-

    genic mice carrying an imbalance in the native ratio of A to B forms of

    progesterone receptor exhibit developmental abnormalities in mammaryglands. Proc Natl Acad Sci USA 1998;95:696701.

    [41] Perusquia M. Nongenomic action of steroids in myometrial contractility.

    Endocrine 2001;15:6372.

    [42] Chanrachakul B, Pipkin FB, Warren AY, Arulkumaran S, Khan RN. Pro-

    gesterone enhances the tocolytic effect of ritodrine in isolated pregnant

    human myometrium. Am J Obstet Gynecol 2005;192:45863.

    [43] Lofgren M, HolstJ, BackstromT. Effects in vitro of progesteroneand two

    5 alpha-reduced progestins, 5 alpha-pregnane-3,20-dione and 5 alpha-

    pregnane-3 alpha-ol-20-one, on contracting human myometrium at term.

    Acta Obstet Gynecol Scand 1992;71:2833.

    [44] Perusquia M, Garcia-Yanez E, Ibanez R, Kubli-Garfias C. Non-genomic

    mechanism of action of delta-4 and 5-reduced androgens and pro-

    gestins on the contractility of the isolated rat myometrium. Life Sci

    1990;47:154753.

    [45] PerusquiaM, Jasso-Kamel J. Influence of 5alpha- and 5beta-reduced pro-gestins on the contractility of isolated human myometrium at term. Life

    Sci 2001;68:293344.

    [46] Pinto RM, Lerner U, Pontelli H. The effect of progesterone on oxytocin-

    induced contraction of the three separate layers of human gestational

    myometrium in theuterine body andlower segment.Am J Obstet Gynecol

    1967;98:54754.

    [47] Fu X, Rezapour M, Lofgren M, Ulmsten U, Backstrom T. Unexpected

    stimulatory effect of progesteroneon human myometrial contractile activ-

    ity in vitro. Obstet Gynecol 1993;82:238.

    [48] Fu X, Rezapour M, Lofgren M, Ulmsten U, Backstrom T. Antitachyphy-

    lactic effects of progesterone and oxytocin on term human myometrial

    contractile activity in vitro. Obstet Gynecol 1993;82:5328.

    [49] Rezapour M, Hongpaisan J, Fu X, Backstrom T, Roomans GM, Ulm-

    sten U. Effects of progesterone and oxytocin on intracellular elemental

    composition of term human myometrium in vitro. Eur J Obstet Gynecol

    Reprod Biol 1996;68:1917.

    [50] Hendricks CH, Brenner WE, Gabel RA, Kerenyi T. The effect of pro-

    gesterone administered intra-amniotically in late human pregnancy. In:

    Brook Lodge Symposium. 1961. p. 5364.

    [51] Pinto RM, Montuori E, Lerner U, Baleiron H, Glauberman M,

    Nemirovsky H. Effect of progesterone on the oxytocicaction of estradiol-

    17-beta. Am J Obstet Gynecol 1965;91:10849.

    [52] da Fonseca EB, Bittar RE, Carvalho MH, Zugaib M. Prophylactic

    administration of progesterone by vaginal suppository to reduce the

    incidence of spontaneous preterm birth in women at increased risk: a ran-

    domized placebo-controlled double-blind study. Am J Obstet Gynecol

    2003;188:41924.

    [53] Meis PJ, Klebanoff M, Thom E, Dombrowski MP, Sibai B, Moawad

    AH, et al. Prevention of recurrent preterm delivery by 17 alpha-

    hydroxyprogesterone caproate. N Engl J Med 2003;348:237985.

    [54] Sexton DJ, OReilly MW, Friel AM, Morrison JJ. Functional effects

    of 17alpha-hydroxyprogesterone caproate (17P) on human myometrial

    contractility in vitro. Reprod Biol Endocrinol 2004;2:80.

    [55] Bogacki M, Silvia WJ, Rekawiecki R, Kotwica J. Direct inhibitory effect

    of progesteroneon oxytocin-inducedsecretion of prostaglandin F(2alpha)

    from bovine endometrial tissue. Biol Reprod 2002;67:1848.

    [56] Dunlap KA, Stormshak F. Nongenomic inhibition of oxytocin binding by

    progesterone in the ovine uterus. Biol Reprod 2004;70:659.

    [57] Grazzini E, Guillon G, Mouillac B, Zingg HH. Inhibition of oxy-

    tocin receptor function by direct binding of progesterone. Nature

    1998;392:50912.

    [58] Kubli-Garfias C, Hoyo-Vadillo C, Lopez-Nieto E, Ponce-Monter H.

    Inhibition of spontaneous contractions of the rat pregnant uterus by pro-

    gesterone metabolites. Proc West Pharmacol Soc 1983;26:1158.

    [59] Kubli-Garfias C, Medrano-Conde L, Beyer C, Bondani A. In vitro inhibi-

    tion of rat uterine contractility induced by 5 alpha and 5 beta progestins.

    Steroids 1979;34:60917.

    [60] Thornton S, Terzidou V, Clark A, Blanks A. Progesterone metabolite

    and spontaneous myometrial contractions in vitro. Lancet 1999;353:

    13279.

    [61] Sheehan PM, Rice GE, Moses EK, Brennecke SP. 5 Beta-dihy-

    droprogesterone and steroid 5 beta-reductasedecrease in association with

    human parturition at term. Mol Hum Reprod 2005;11:495501.[62] Mitchell BF, Mitchell JM, Chowdhury J, Tougas M, Engelen SM, Senff

    N, et al. Metabolites of progesterone and the pregnane X receptor: a

    novel pathway regulating uterine contractility in pregnancy? Am J Obstet

    Gynecol 2005;192:130413 [discussion 13131305].

    [63] Astle S, Khan RN, Thornton S. The effects of a progesterone metabo-

    lite, 5 beta-dihydroprogesterone, on oxytocin receptor binding in human

    myometrial membranes. BJOG 2003;110:58992.

    [64] Burger K, Fahrenholz F, Gimpl G. Non-genomic effects of progesterone

    on the signaling function of G protein-coupled receptors. FEBS Lett

    1999;464:259.

    [65] Mellon SH, Griffin LD. Neurosteroids: biochemistry and clinical signif-

    icance. Trends Endocrinol Metab 2002;13:3543.

    [66] Putnam CD, Brann DW, Kolbeck RC, Mahesh VB. Inhibition of uterine

    contractility by progesterone and progesterone metabolites: mediation

    by progesterone and gamma amino butyric acidA receptor systems. BiolReprod 1991;45:26672.

    [67] Sergeev PV, Sizov PI, Dukhanin AS, Mineeva EN. Study of the GABA-

    benzodiazepine receptor system of the human myometrium. Biull Eksp

    Biol Med 1990;110:3824.

    [68] Edwards DP. Regulation of signal transduction pathways by estrogen and

    progesterone. Annu Rev Physiol 2005;67:33576.

    [69] LeonhardtSA, Boonyaratanakornkit V, Edwards DP. Progesterone recep-

    tor transcription and non-transcription signaling mechanisms. Steroids

    2003;68:76170.

    [70] Falkenstein E, Heck M, Gerdes D, Grube D, Christ M, Weigel M, et

    al. Specific progesterone binding to a membrane protein and related

    nongenomic effects on Ca2+-fluxes in sperm. Endocrinology 1999;140:

    59996002.

    [71] Falkenstein E, Schmieding K, Lange A, Meyer C, Gerdes D, Welsch U,

    et al. Localization of a putative progesterone membrane binding pro-

    tein in porcine hepatocytes. Cell Mol Biol (Noisy-le-grand) 1998;44:

    5718.

    [72] Gerdes D, Wehling M, Leube B, Falkenstein E. Cloning and tissue

    expression of two putative steroid membrane receptors. Biol Chem

    1998;379:90711.

    [73] Mifsud W, Bateman A. Membrane-bound progesterone receptors con-

    tain a cytochrome b5-like ligand-binding domain. Genome Biol 2002;3.

    RESEARCH0068.

    [74] Mourot B, Nguyen T, Fostier A, Bobe J. Two unrelated putative

    membrane-bound progestin receptors, progesterone membrane receptor

    component 1 (PGMRC1) and membrane progestin receptor (mPR) beta,

    are expressed in the rainbow trout oocyte and exhibit similar ovarian

    expression patterns. Reprod Biol Endocrinol 2006;4:6.

    [75] Peluso JJ, Pappalardo A, Fernandez G, Wu CA. Involvement of an

    unnamed protein, RDA288, in the mechanism through which proges-

    terone mediates its antiapoptotic action in spontaneously immortalized

    granulosa cells. Endocrinology 2004;145:301422.

    [76] Peluso JJ, Pappalardo A, Losel R, Wehling M. Expression and func-

    tion of PAIRBP1 within gonadotropin-primed immature rat ovaries:

    PAIRBP1 regulation of granulosa and luteal cell viability. Biol Reprod

    2005;73:26170.

    [77] Peluso JJ. Multiplicity of progesterones actions and receptors in the

    mammalian ovary. Biol Reprod 2006;75:28.

    [78] Zhu Y, Bond J, Thomas P. Identification, classification, and partial

    characterization of genes in humans and other vertebrates homolo-

    gous to a fish membrane progestin receptor. Proc Natl Acad Sci USA

    2003;100:223742.

    [79] Zhu Y, Rice CD, Pang Y, Pace M, Thomas P. Cloning, expression, and

    characterization of a membrane progestin receptor and evidence it is an

  • 8/12/2019 1-s2.0-S1084952107000717-main

    10/11

    330 S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331

    intermediary in meiotic maturation of fish oocytes. Proc Natl Acad Sci

    USA 2003;100:22316.

    [80] Karteris E, Zervou S, Pang Y, Dong J, Hillhouse EW, Randeva HS, et

    al. Progesterone signaling in human myometrium through two novel

    membrane G protein coupled receptors: potential role in functional pro-

    gesterone withdrawal at term. Mol Endocrinol 2006.

    [81] Chapman NR, Kennelly MM, Harper KA, Europe-Finner GN, Rob-

    son SC. Examining the spatio-temporal expression of mRNA encoding

    the membrane-bound progesterone receptor-alpha isoform in humancervix and myometrium during pregnancy and labour. Mol Hum Reprod

    2006;12:1924.

    [82] Fernandes MS, Pierron V, Michalovich D, Astle S, Thornton S, Peltoketo

    H, et al. Regulated expression of putative membrane progestin receptor

    homologues in human endometrium and gestational tissues. J Endocrinol

    2005;187:89101.

    [83] Fu X, Moberg C, Backstrom T, Ulmsten U, Gylfe E. Anisomycin and

    verapamil influence the action of progesterone on regulation of term

    human myometrial contractile activity. Clin Endocrinol (Oxf) 1997;47:

    34955.

    [84] Krietsch T, Fernandes MS, Kero J, Losel R, Heyens M, Lam EW,

    et al. Human homologs of the putative G protein-coupled membrane

    progestin receptors (mPRalpha, beta, and gamma) localize to the endo-

    plasmic reticulum and are not activated by progesterone. Mol Endocrinol

    2006;20:314664.[85] Challis JRG, Lye SJ. Parturition. In: Knobil E, Neil JD, editors. The

    physiology of reproduction. New York: Raven Press; 1994. p. 9851031.

    [86] Liggins GC. The physiological role of prostaglandins in parturition. J

    Reprod Fertil 1973;18(Suppl):14350.

    [87] Liggins GC, Fairclough RJ, Grieves SA, Kendall JZ, Knox BS. The

    mechanism of initiation of parturition in the ewe. Recent Prog Horm

    Res 1973;29:11159.

    [88] Young IR. The comparative physiology of parturition in mammals. Front

    Horm Res 2001;27:1030.

    [89] Boroditsky RS, Reyes FI, Winter JS, Faiman C. Maternal serum estrogen

    and progesterone concentrations preceding normal labor. Obstet Gynecol

    1978;51:68691.

    [90] TulchinskyD, Hobel CJ,Yeager E, MarshallJR. Plasma estrone,estradiol,

    estriol, progesterone, and 17-hydroxyprogesterone in human pregnancy.

    I. Normal pregnancy. Am J Obstet Gynecol 1972;112:1095100.

    [91] Walsh SW, Stanczyk FZ,NovyMJ. Daily hormonalchangesin themater-

    nal, fetal,and amniotic fluidcompartments before parturition in a primate

    species. J Clin Endocrinol Metab 1984;58:62939.

    [92] Mesiano S. Myometrial progesterone responsiveness and the control of

    human parturition. J Soc Gynecol Investig 2004;11:193202.

    [93] MesianoS. Myometrial progesterone responsiveness.Semin Reprod Med

    2007;25:513.

    [94] Haluska GJ, Wells TR, Hirst JJ, Brenner RM, Sadowsky DW, Novy

    MJ. Progesterone receptor localization and isoforms in myometrium,

    decidua, and fetal membranes from rhesus macaques: evidence for func-

    tional progesterone withdrawal at parturition. J Soc Gynecol Investig

    2002;9:12536.

    [95] Mesiano S, Chan EC, Fitter JT, Kwek K, Yeo G, Smith R. Progesterone

    withdrawal and estrogen activation in human parturition are coordi-

    nated by progesterone receptor A expression in the myometrium. J Clin

    Endocrinol Metab 2002;87:292430.

    [96] Henderson D, Wilson T. Reduced binding of progesterone receptor to its

    nuclear response element after human labor onset. Am J Obstet Gynecol

    2001;185:57985.

    [97] Condon JC, Jeyasuria P, Faust JM, Wilson JW, Mendelson CR. A decline

    in the levels of progesterone receptor coactivators in the pregnant uterus

    at term may antagonize progesterone receptor function and contribute

    to the initiation of parturition. Proc Natl Acad Sci USA 2003;100:

    951823.

    [98] Dong X, Shylnova O, Challis JR, Lye SJ. Identification and characteri-

    zation of the protein-associated splicing factor as a negative co-regulator

    of the progesterone receptor. J Biol Chem 2005;280:1332940.

    [99] Shukla S, Del Gatto-Konczak F, Breathnach R, Fisher SA. Competition

    of PTB with TIA proteins for binding to a U-rich cis-element determines

    tissue-specific splicing of the myosin phosphatase targeting subunit 1.

    RNA 2005;11:172536.

    [100] Csapo A. In: Wolstenholme GEW, Knight J, editors. Progesterone: Its

    regulatory effect on the myometrium. London: Ciba Foundation, J & A

    Churchill; 1969. p. 412.

    [101] Dawe ST, Husband AJ, Langford CM. Effects of induction of partu-

    rition in ewes with dexamethasone or oestrogen on concentrations of

    immunoglobulins in colostrum, and absorption of immunoglobulins by

    lambs. Aust J Biol Sci 1982;35:2239.[102] Wu WX, Ma XH, Coksaygan T, Chakrabarty K, Collins V, Rose J,

    et al. Prostaglandin mediates premature delivery in pregnant sheep

    induced by estradiol at 121 days of gestational age. Endocrinology

    2004;145:144452.

    [103] Mecenas CA, Giussani DA, Owiny JR, Jenkins SL, Wu WX, Honnebier

    BO, et al. Production of premature delivery in pregnant rhesus monkeys

    by androstenedione infusion. Nat Med 1996;2:4438.

    [104] Nathanielsz PW, Jenkins SL, Tame JD, Winter JA, Guller S, GiussaniDA.

    Local paracrine effects of estradiol are central to parturition in the rhesus

    monkey. Nat Med 1998;4:4569.

    [105] Giussani DA, Jenkins SL, Mecenas CA, Winter JA, Honnebier BO,

    Wu W, et al. Daily and hourly temporal association between delta4-

    androstenedione-induced preterm myometrial contractions and maternal

    plasma estradiol and oxytocin concentrations in the 0.8 gestation rhesus

    monkey. Am J Obstet Gynecol 1996;174:10505.[106] Novy MJ, Walsh SW. Dexamethasone and estradiol treatment in preg-

    nant rhesus macaques: effects on gestational length, maternal plasma

    hormones, and fetal growth. Am J Obstet Gynecol 1983;145:92031.

    [107] Katzenellenbogen BS. Mechanisms of action and cross-talk between

    estrogen receptor and progesterone receptor pathways. J Soc Gynecol

    Investig 2000;7:S337.

    [108] Katzenellenbogen BS, Nardulli AM, Read LD. Estrogen regulation of

    proliferation and hormonal modulation of estrogen and progesterone

    receptor biosynthesis and degradation in target cells. Prog Clin Biol Res

    1990;322:20111.

    [109] TsaiM-J, ClarkJH, SchraderWT,OMalleyBW. Mechanismsof action of

    hormones that act as transcription-regulatory factors. In: Wilson J, Foster

    D, Kronenberg H, Larsen P, editors. Williams textbook of endocrinology.

    Philadelphia: W.B. Saunders Company; 1998. p. 5594.

    [110] Leavitt WW, Cobb AD, Takeda A. Progesterone-modulation of estrogen

    action: rapid down regulation of nuclear acceptor sites for the estrogen

    receptor. Adv Exp Med Biol 1987;230:4978.

    [111] Haluska GJ, West NB, Novy MJ, Brenner RM. Uterine estrogen receptors

    are increased by RU486 in late pregnant rhesus macaques but not after

    spontaneous labor. J Clin Endocrinol Metab 1990;70:1816.

    [112] Chan EC, Fraser S, Yin S, Yeo G, Kwek K, Fairclough RJ, et al.

    Human myometrial genes are differentiallyexpressed in labor: a suppres-

    sion subtractive hybridization study. J Clin Endocrinol Metab 2002;87:

    243541.

    [113] Aguan K, Carvajal JA, Thompson LP, Weiner CP. Application of a func-

    tional genomics approach to identify differentially expressed genes in

    human myometrium during pregnancy and labour. Mol Hum Reprod

    2000;6:11415.

    [114] Bethin KE, Nagai Y, Sladek R, Asada M, Sadovsky Y, Hudson TJ, et

    al. Microarray analysis of uterine gene expression in mouse and human

    pregnancy. Mol Endocrinol 2003;17:145469.

    [115] Charpigny G, Leroy MJ, Breuiller-Fouche M, Tanfin Z, Mhaouty-Kodja

    S, Robin P, et al. A functional genomic study to identify differential gene

    expression in the preterm and term human myometrium. Biol Reprod

    2003;68:228996.

    [116] Esplin MS, Fausett MB, Peltier MR, Hamblin S, Silver RM, Branch DW,

    et al. The use of cDNA microarray to identify differentially expressed

    labor-associated genes within the human myometrium during labor. Am

    J Obstet Gynecol 2005;193:40413.

    [117] Havelock JC, Keller P, Muleba N, Mayhew BA, Casey BM, Rainey WE,

    et al. Human myometrial gene expression before and during parturition.

    Biol Reprod 2005;72:70719.

    [118] Breuiller-Fouche M, Germain G. Gene and protein expression in the

    myometrium in pregnancy and labor. Reproduction 2006;131:83750.

  • 8/12/2019 1-s2.0-S1084952107000717-main

    11/11

    S. Mesiano, T.N. Welsh / Seminars in Cell & Developmental Biology 18 (2007) 321331 331

    [119] Bisits AM, Smith R, Mesiano S, Yeo G, Kwek K, Macintyre DA, et

    al. Inflammatory aetiology of human myometrial activation tested using

    directed graphs. PLoS Comput Biol 2005;1:1326.

    [120] Salomonis N, Cotte N, Zambon AC, Pollard KS, Vranizan K, Doniger

    SW, et al. Identifying genetic networks underlying myometrial transition

    to labor. Genome Biol 2005;6:R12.

    [121] Madsen G, Zakar T, Ku CY, Sanborn BM, Smith R, Mesiano S.

    Prostaglandins differentially modulate progesterone receptor-A and -B

    expression in human myometrial cells: evidence for prostaglandin-

    induced functional progesterone withdrawal. J Clin Endocrinol Metab

    2004;89:10103.