PDF (454 KB)

37
Endocrine and Paracrine Regulation of Birth at Term and Preterm* JOHN R.G. CHALLIS, STEPHEN G. MATTHEWS, WILLIAM GIBB, AND STEPHEN J. LYE Departments of Physiology (J.R.G.C., S.G.M., W.G., S.J.L.) and of Obstetrics and Gynaecology (J.R.G.C., S.G.M., S.J.L.), University of Toronto, Toronto, Ontario, Canada M55 1A8; Program in Development and Fetal Health (J.R.G.C., S.J.L.), Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5; MRC Group in Fetal and Neonatal Health and Development (J.R.G.C., S.J.L.); Department of Obstetrics and Gynaecology, and Cellular and Molecular Medicine (W.G.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6 ABSTRACT We have examined factors concerned with the maintenance of uterine quiescence during pregnancy and the onset of uterine ac- tivity at term in an animal model, the sheep, and in primate species. We suggest that in both species the fetus exerts a critical role in the processes leading to birth, and that activation of the fetal hypothalamic-pituitary-adrenal axis is a central mechanism by which the fetal influence on gestation length is exerted. Increased cortisol output from the fetal adrenal gland is a common charac- teristic across animal species. In primates, there is, in addition, increased output of estrogen precursor from the adrenal in late gestation. The end result, however, in primates and in sheep is similar: an increase in estrogen production from the placenta and intrauterine tissues. We have revised the pathway by which en- docrine events associated with parturition in the sheep come about and suggest that fetal cortisol directly affects placental PGHS expression. In human pregnancy we suggest that cortisol increases PGHS expression, activity, and PG output in human fetal mem- branes in a similar manner. Simultaneously, cortisol contributes to decreases in PG metabolism and to a feed-forward loop involving elevation of CRH production from intrauterine tissues. In human pregnancy, there is no systemic withdrawal of progesterone in late gestation. We have argued that high circulating progesterone con- centrations are required to effect regionalization of uterine activ- ity, with predominantly relaxation in the lower uterine segment, allowing contractions in the fundal region to precipitate delivery. This new information, arising from basic and clinical studies, should further the development of new methods of diagnosing the patient at risk of preterm labor, and the use of scientifically based strategies specifically for the management of this condition, which will improve the health of the newborn. (Endocrine Reviews 21: 514 –550, 2000) I. Introduction II. Regulation of Myometrial Contractions III. Pregnancy: Phase 0 of Parturition IV. Myometrial Activation: Phase 1 of Parturition A. Activation: role of fetal hypothalamic-pituitary-ad- renal (HPA) maturation B. Activation mechanism by which cortisol changes placental steroid and PG synthesis C. HPA function in the primate fetus and activation of parturition D. HPA maturation in the primate fetus E. Placental progesterone and human pregnancy: the enigma of the progesterone block V. Myometrial Stimulation: Phase 2 of Parturition A. Stimulation: role of oxytocin B. Stimulation: role of PGs C. Stimulation: role of CRH VI. Application to Clinical Preterm Labor I. Introduction P ARTURITION is the process by which the fetus is ex- pelled from the uterus to the extrauterine environment. Parturition results from a complex interplay of maternal and fetal factors. It requires that the uterus, which has been main- tained in a relative state of quiescence during pregnancy, develops coordinated contractility and that the cervix dilates in a manner that allows passage of the fetus through the birth canal. To be successful, parturition requires also that matu- ration of those fetal organ systems necessary for extrauterine survival has occurred, and that the maternal organism has undergone the changes necessary for lactation in the post- partum period. It is not surprising, therefore, that synchro- nous maturation of the fetus and stimulus to increased uter- ine activity should be desirable, and much evidence suggests that it is the fetus itself that triggers both these series of events. Preterm birth, where there is asynchrony between the labor process and fetal maturation, occurs in 8 –10% of all pregnancies, and its incidence has changed little in the past 40 yr (1). Indeed, factors such as low socioeconomic status of some inner-city populations, the tendency for women to choose to start a family at an older age, and the impact of fertility treatment are contributing to an increase in the in- cidence of preterm delivery (2, 3). Improved neonatal care, however, continues to reduce the mortality rate due to pre- Address reprint requests to: Dr. J. R. G. Challis, Department of Physiology, Medical Sciences Building, 1 King’s College Circle, Uni- versityofToronto,Toronto,OntarioM5S1A8Canada.E-mail:j.challis@ utoronto.ca * Work in the authors’ laboratories has been supported by Medical Research Council (MRC) Group and operating grants from the MRC of Canada. 0163-769X/00/$03.00/0 Endocrine Reviews 21(5): 514 –550 Copyright © 2000 by The Endocrine Society Printed in U.S.A. 514

Transcript of PDF (454 KB)

Page 1: PDF (454 KB)

Endocrine and Paracrine Regulation of Birth at Termand Preterm*

JOHN R.G. CHALLIS, STEPHEN G. MATTHEWS, WILLIAM GIBB, AND

STEPHEN J. LYE

Departments of Physiology (J.R.G.C., S.G.M., W.G., S.J.L.) and of Obstetrics and Gynaecology(J.R.G.C., S.G.M., S.J.L.), University of Toronto, Toronto, Ontario, Canada M55 1A8; Program inDevelopment and Fetal Health (J.R.G.C., S.J.L.), Samuel Lunenfeld Research Institute, Mount SinaiHospital, Toronto, Ontario, Canada M5G 1X5; MRC Group in Fetal and Neonatal Health andDevelopment (J.R.G.C., S.J.L.); Department of Obstetrics and Gynaecology, and Cellular andMolecular Medicine (W.G.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6

ABSTRACTWe have examined factors concerned with the maintenance of

uterine quiescence during pregnancy and the onset of uterine ac-tivity at term in an animal model, the sheep, and in primatespecies. We suggest that in both species the fetus exerts a criticalrole in the processes leading to birth, and that activation of the fetalhypothalamic-pituitary-adrenal axis is a central mechanism bywhich the fetal influence on gestation length is exerted. Increasedcortisol output from the fetal adrenal gland is a common charac-teristic across animal species. In primates, there is, in addition,increased output of estrogen precursor from the adrenal in lategestation. The end result, however, in primates and in sheep issimilar: an increase in estrogen production from the placenta andintrauterine tissues. We have revised the pathway by which en-docrine events associated with parturition in the sheep come aboutand suggest that fetal cortisol directly affects placental PGHS

expression. In human pregnancy we suggest that cortisol increasesPGHS expression, activity, and PG output in human fetal mem-branes in a similar manner. Simultaneously, cortisol contributes todecreases in PG metabolism and to a feed-forward loop involvingelevation of CRH production from intrauterine tissues. In humanpregnancy, there is no systemic withdrawal of progesterone in lategestation. We have argued that high circulating progesterone con-centrations are required to effect regionalization of uterine activ-ity, with predominantly relaxation in the lower uterine segment,allowing contractions in the fundal region to precipitate delivery.This new information, arising from basic and clinical studies,should further the development of new methods of diagnosing thepatient at risk of preterm labor, and the use of scientifically basedstrategies specifically for the management of this condition, whichwill improve the health of the newborn. (Endocrine Reviews 21:514 –550, 2000)

I. Introduction

II. Regulation of Myometrial Contractions

III. Pregnancy: Phase 0 of Parturition

IV. Myometrial Activation: Phase 1 of ParturitionA. Activation: role of fetal hypothalamic-pituitary-ad-

renal (HPA) maturationB. Activation mechanism by which cortisol changes

placental steroid and PG synthesisC. HPA function in the primate fetus and activation of

parturitionD. HPA maturation in the primate fetusE. Placental progesterone and human pregnancy: the

enigma of the progesterone blockV. Myometrial Stimulation: Phase 2 of Parturition

A. Stimulation: role of oxytocinB. Stimulation: role of PGsC. Stimulation: role of CRH

VI. Application to Clinical Preterm Labor

I. Introduction

PARTURITION is the process by which the fetus is ex-pelled from the uterus to the extrauterine environment.

Parturition results from a complex interplay of maternal andfetal factors. It requires that the uterus, which has been main-tained in a relative state of quiescence during pregnancy,develops coordinated contractility and that the cervix dilatesin a manner that allows passage of the fetus through the birthcanal. To be successful, parturition requires also that matu-ration of those fetal organ systems necessary for extrauterinesurvival has occurred, and that the maternal organism hasundergone the changes necessary for lactation in the post-partum period. It is not surprising, therefore, that synchro-nous maturation of the fetus and stimulus to increased uter-ine activity should be desirable, and much evidence suggeststhat it is the fetus itself that triggers both these series ofevents.

Preterm birth, where there is asynchrony between thelabor process and fetal maturation, occurs in 8–10% of allpregnancies, and its incidence has changed little in the past40 yr (1). Indeed, factors such as low socioeconomic status ofsome inner-city populations, the tendency for women tochoose to start a family at an older age, and the impact offertility treatment are contributing to an increase in the in-cidence of preterm delivery (2, 3). Improved neonatal care,however, continues to reduce the mortality rate due to pre-

Address reprint requests to: Dr. J. R. G. Challis, Department ofPhysiology, Medical Sciences Building, 1 King’s College Circle, Uni-versityofToronto,Toronto,OntarioM5S1A8Canada.E-mail: [email protected]

* Work in the authors’ laboratories has been supported by MedicalResearch Council (MRC) Group and operating grants from the MRC ofCanada.

0163-769X/00/$03.00/0Endocrine Reviews 21(5): 514–550Copyright © 2000 by The Endocrine SocietyPrinted in U.S.A.

514

Page 2: PDF (454 KB)

maturity, although preterm birth remains the primary causeof neonatal death. In North America, the cost of caring forinfants in the neonatal intensive care nursery during the firstmonths of life has been estimated at $5–6 billion annually (3).That figure does not take into account the extraordinaryemotional stress to the family of the prematurely deliveredinfant. Nor does it take into account the long-term costsrequired for chronic care of these infants, some of whom havemajor motor and/or mental handicaps and/or long-termneuro-developmental complications. To prevent pretermbirth effectively, we need to understand the fundamentalprocesses that switch the myometrium from its relative qui-escence during pregnancy to the activated and contractilestate at the time of labor. We will develop the thesis thatregulation of myometrial function requires both endocrineand mechanical controls. Furthermore, it is now evident thatthe cause of preterm labor may vary at different times duringpregnancy and will not necessarily reflect acceleration of theprocesses at term gestation. The ability to recognize thesevarious causes of premature delivery, in a clinical setting,and then provide appropriate treatment remains a majorclinical challenge. Furthermore, it is evident that preventionof preterm delivery may not always be desirable, particularlyif the fetus is allowed to develop in a hostile intrauterineenvironment.

Causes of preterm birth in general fall into three catego-ries: iatrogenic, where there is demonstrable complication ofpregnancy such as preeclampsia or fetal distress that requiresobstetrical intervention; premature rupture of the fetal mem-branes with or without infection; and, idiopathic pretermlabor. The relative importance of these causes varies. How-ever, most sources consider that approximately 30–40% ofpreterm birth is associated with an underlying infective pro-cess, and 40–50% of preterm births are idiopathic.

In this review, we will focus attention on experimentalstudies in the sheep, the species of choice for many inves-tigators concerned with understanding the processes of birth(4). We shall then extrapolate from the sheep to an under-standing of parturition in primates, particularly in the hu-man. Our central thesis is that the processes of birth areremarkably similar, at a fundamental level, across species,and in both sheep and human the fetus, through activationof its hypothalamic-pituitary-adrenal (HPA) axis, plays acentral and crucial role. We shall examine how the fetal HPAaxis may be activated in response to a stress circumstanceduring pregnancy, e.g., hypoxemia, such as that perhapsassociated with reduced uteroplacental perfusion in pre-eclampsia. It will be apparent that the fetal signal provokesincreased outputs of stimulatory PGs and other uterotoninsfrom intrauterine tissues. It is evident now that there is aprogression from fetal to maternal control of intrauterine PGproduction. Furthermore, the regulation of PG synthesis andmetabolism in fetal trophoblasts and maternal uterus is ef-fected by different mechanisms.

II. Regulation of Myometrial Contractions

During pregnancy, myometrial activity is characterized bypoorly coordinated contractures, or the Braxton-Hicks con-

tractions of human gestation (5). In late pregnancy, the uterusundergoes preparedness for the stimuli that lead to contrac-tility and labor (6, 7). Those stimuli may be local, maternal,mechanical, or fetal (8). The contracture pattern of uterineactivity has been observed in several species, including thesheep, baboon, and rhesus monkey (9). The development ofcoordinated uterine contractions at term results in a myo-metrium that is excitable, generating high-frequency, high-amplitude contractions. It is spontaneously active and re-sponds to exogenous uterotonins. The transition of themyometrium from a quiescent to an active state has beentermed “activation.” When this has occurred the myome-trium can then undergo “stimulation” in response to endog-enous and/or exogenous agonists (8).

We have found it useful to divide the uterine phenotypeinto different stages of the parturition process (10). Theuterus is relatively quiescent during 95% of pregnancy, cor-responding to phase 0 of parturition. Activation correspondsto phase 1 and is effected predominantly by mechanicalinput, and through regulation by uterotrophins such as es-trogen. Stimulation corresponds to phase 2, when endoge-nous uterotonins, including PGs and oxytocin (OT), act onthe activated myometrium. Postpartum involution corre-sponds to phase 3. In this sequence of events, the “initiation”of parturition corresponds to the transition from phase 0 tophase 1, although clearly one could argue that initiationstarted much earlier in gestation (11).

Contraction of the myometrium at term or preterm de-pends upon conformational changes in the actin and myosinmolecules, which allow actin and myosin filaments to slideover each other, ultimately leading to a shortening of themyocyte (Fig. 1 and Refs. 12 and 13). The confirmationalchanges (involving cross-bridge cycling of the myosin head)require ATP, which is generated by myosin after phosphor-ylation of the 20-kDa light chains of myosin by the enzymemyosin light chain kinase (MLCK). This enzyme is central tosignaling pathways that both stimulate and inhibit myome-trial contractions (14, 15). MLCK is activated through inter-action with the calcium binding protein calmodulin (CAM),which in turn requires 4 Ca21 ions for its own activation.Binding of calcium-activated CAM to MLCK induces a con-formational change in the enzyme, allowing MLCK to phos-phorylate the 20-kDa light chains of myosin. MLCK can alsoundergo phosphorylation by protein kinase A (PKA, cAMP-activated protein kinase), which reduces the affinity of theenzyme for calcium calmodulin (Ca-CAM) and leads to itsinactivation (14, 16). Regulation of MLCK has been reviewedextensively (17, 18). It is evident that activity of this enzymeis altered by intracellular pathways that regulate levels ofcalcium and of cAMP and is critical for the development ofuterine contractility. Uterotonins generally increase intracel-lular calcium levels ([Ca21]i), by increased influx of Ca21

through receptor-operated channels, or release of calciumfrom intracellular stores including sarcoplasmic reticulum(see Ref. 19). Agents that inhibit myometrial activity do so byincreasing intracellular levels of cyclic nucleotides cAMP orcGMP, which in turn inhibit release of calcium from intra-cellular stores or reduce MLCK activity. Binding of agentssuch as b-adrenergic agonists, relaxin and prostacyclin, tomyometrial receptors activates adenylate cyclase activity,

October, 2000 PARTURITION 515

Page 3: PDF (454 KB)

leading to an increase in cAMP generation, while uterineinhibitors such as nitric oxide (NO) activate guanyl cyclase,increasing cGMP. In collaborative studies, Pato et al. (20)characterized MLCK purified from pregnant sheep myome-trium. The enzyme had an apparent molecular mass of 160kDa and high substrate specificity for myosin light chains.Sheep myometrial MLCK has an absolute requirement forCa21 and CAM for activation; in the absence of Ca-CAM,MLCK is inactive. On binding Ca-CAM, MLCK undergoes aconformational change that exposes the catalytic site, whichcan then phosphorylate the 20-kDa myosin light chains toinitiate contraction. Relaxation is achieved either by dephos-phorylation of MLC-20 by the catalytic subunits of type 2Aphosphatase (21) or by reduction in MLCK activity. The latteris achieved, as discussed, by reduction in [Ca21]i, resultingin dissociation of Ca-CAM from MLCK. Sheep myometrialMLCK is also a substrate for PKA, which phosphorylatesserine residues on the sheep myometrial enzyme in the pres-ence or absence of bound Ca-CAM. The ability of PKA toinhibit myometrial MLCK activity, even in the presence ofagonists that increase [Ca21]i, provides a biochemical ratio-nale for the finding that agents that increase intracellularcAMP inhibit uterine contractions even in the presence ofcalcium-activating agents such as OT and stimulatory PG.Ca-CAM can also activate phosphodiesterase to increase thebreakdown of cAMP.

Inhibition of myometrial activity by b-adrenergic agonists,relaxin, and PGI2 is mediated by increases in intracellularcAMP (see Ref. 12). Binding of the inhibitor to its specific cellmembrane receptor causes dissociation of the receptor-linked heterotrimeric GTP-binding protein Gs into b-, g-, anda-subunits. The a-subunit activates adenylate cyclase to ini-tiate cAMP synthesis. cAMP, in turn, activates PKA, whichthen phosphorylates a series of regulatory proteins. Acti-vated PKA either phosphorylates MLCK to reduce its abilityto bind Ca-CAM or phosphorylates a membrane-binding sitefor Ca21 that increases calcium binding and reduces freeintracellular calcium concentrations.

Regulation of myometrial calcium levels has been re-viewed extensively (see Refs. 12 and 22–24). Free resting

Ca21 increases from 150 nm to about 500 nm during con-traction through influx of extracellular Ca21 or by the releaseof Ca21 from intracellular binding sites or intracellular or-ganelles (25, 26). Extracellular Ca21 enters cells through re-ceptor-operated or voltage-gated channels. Release of intra-cellular Ca21 from sarcoplasmic reticulum is activatedthrough the phosphoinositol (PI) pathway. Binding of auterotonin to its plasma membrane receptor activates a Gprotein transducer, coupled to phospholipase C, which freesinositol trisphosphate (IP3) and diacylglycerol (27, 28). FreeIPs, especially IP3, increase cellular calcium from intracel-lular storage sites. Interestingly, IP3 binding in myometriumwas inhibited by calcium, suggesting that this might providea mechanism for regulating the IP3 response by oscillating[Ca21]i. Diacylglycerol formed during IP3 turnover maystimulate PKC to phosphorylate cellular proteins such asMLCK or be rapidly phosphorylated by diacylglycerol ki-nase to phosphatidic acid, a naturally occurring Ca21 iono-phore, or lead to release of arachidonic acid by cellularlipases, resulting in production of eicosanoids (see below).

Function of the myometrium during labor at term or pre-term requires highly developed cell-to-cell coupling, effectedthrough formation of intercellular GAP junctions within ad-jacent cell membranes (14, 29). The proteins forming GAPjunctions are termed connexins and are classified accordingto their apparent molecular weights (30). Connexins are ar-ranged into hexameric hemichannels, which become alignedacross adjacent cells to form an interconnecting pore thatallows low-resistance electrical or ionic coupling between thecells and provides a pathway for metabolite transfer (31).Hundreds of individual channels arrange themselves into anorganized plaque to form a GAP junction. Regulation ofconnexins occurs at the level of transcription and translation(31, 32); mechanisms also operate to control transport ofconnexin protein to the cell membrane and to direct assemblyinto connexons, through apposition, clustering, and forma-tion of functional channels (33, 34). This complex process ispoorly understood, although it is influenced by steroids andby mechanical stretch (35). GAP junction formation requiresthe presence of cell adhesion molecules, and in early studies,

FIG. 1. Cartoon of a myometrial cell in-dicating the intracellular biochemicalpathways involved in regulating con-tractions. MLCK is central to uterinecontractility. It is activated by Ca-CAMafter an increase in intracellular cal-cium levels. This increase is generatedby the action of various uterotonins:PGF acting through PGF receptor (FP),OT acting through OTR. Agents thatincrease cAMP (b-agonists) or cyclicGMP, or NO donors decrease uterinecontractility. AA, Arachidonic acid;Atosiban OTR antagonist.

516 CHALLIS ET AL. Vol. 21, No. 5

Page 4: PDF (454 KB)

Meyer et al. (36) showed that appearance of GAP junctionsin transfected S180 cells was blocked by coincubation withantisera to liver cell adhesion molecule.

Garfield (see Refs. 14 and 16) established clearly that anabsence of GAP junctions in the pregnant myometrium wasresponsible for high-input resistance of these smooth musclecells and poor coordination of uterine contractions. There isa massive increase in numbers of GAP junctions with theonset of labor, which significantly enhances electrical cou-pling and allows the myometrium to develop synchronoushigh amplitude contractions (37). An increase in GAP junc-tions with labor onset has been found in all species studied.In the rat, levels of connexin-43 (CX-43) mRNA and proteinwere low during pregnancy but increased some 48 h beforelabor (38, 39). Highest levels of mRNA and protein werefound during delivery itself. This is critical because the half-life of GAP junctions may be as short as 1–2 h, and hencecontinued synthesis would be required to maintain labor.Increases in CX-43 mRNA have been reported in sheep andhuman myometrium with the onset of labor and correlatedwith increases in CX-43 protein (37, 38). Permeability of GAPjunctions may be facilitated through phosphorylation at con-sensus serine and tyrosine sites within the cytoplasmic do-main of CX-43. Garfield (14) demonstrated that cell-to-cellcommunication in the myometrium is reduced by elevated[Ca21]i and increased levels of cAMP. Importantly, morerecent studies have shown that the pattern of CX-43 in myo-metrium during pregnancy differs from that of CX-26. Con-nexin-26 expression is elevated in midgestation in the rat andappears to be associated more with uterine quiescence (7, 8).

III. Pregnancy: Phase 0 of Parturition

Studies in different species have indicated that a variety ofdifferent inhibitors may play upon the myometrium duringpregnancy. Withdrawal of one or more of these may predictthe onset of delivery; precocious withdrawal may predict theonset of premature parturition. Such an inhibitor, PTH-related peptide (PTHRP), is produced in myometrium, andits rate of transcription is increased by progesterone andtransforming growth factor b (TGFb) (40). PTHRP receptormRNA has also been localized to rat myometrial tissue, sug-gesting that the protein may act in an autocrine/paracrinefashion through specific receptors to activate the Gas sub-units of G proteins and increase intracellular levels of cAMP(40–42).

Relaxin also elevates myometrial cAMP and inhibits OT-induced turnover of phosphoinositide (PI) by the action ofcAMP-dependent protein kinase. Relaxin exerts a dual rolein the inhibition of myometrial contractility and in the reg-ulation of connective tissue changes in the cervix (43, 44).Porter and colleagues (45, 46) were among the first to showthat relaxin suppressed spontaneous uterine contractility inthe rat and guinea pig, but sensitivity to OT was preserved.Thus, the major action of relaxin is one of frequency mod-ulation (47). Hansell et al. (48) and others have demonstratedthat relaxin is expressed in the human fetal membranes,decidua, and placenta, consistent with its exerting para-crine/autocrine effects on intrauterine tissues (49–51). Re-

laxin gene expression is dramatically up-regulated in pa-tients with preterm, premature rupture of membranes(PPROM) (49). Relaxin receptors have been localized to de-cidua and chorionic trophoblast cells, and the protein actsthrough these to up-regulate expression of matrix metallo-proteinases (MMP), especially MMP1, MMP3, and MMP9.Similarly, relaxin increases MMP expression in cervical tis-sue at term. Administration of exogenous relaxin stimulatesseparation of the pubic symphysis in those species in whichit is a prerequisite for delivery (52). In addition, in pigs andrats, relaxin appears necessary for maintaining evolution ofspontaneous uterine contractility in late pregnancy and formaintaining a high frequency of live births (43). In vitrostudies have shown that relaxin blocks the action of stimu-lants such as OT, carbachol, and norepinephrine on the myo-metrium, through mechanisms involving PKA-mediatedphosphorylation of PLC-linked G proteins. This in turn in-hibits IP3 turnover and the increase in [Ca21]i (22). Althoughthe precise role that relaxin plays during pregnancy remainsto be determined, it may be particularly useful in maintain-ing uterine quiescence during the period when progesteroneconcentrations are falling and estrogen levels are beginningto increase before the onset of labor (see Ref. 12). In addition,there are reports that relaxin may act centrally to increasecirculating plasma OT and vasopressin concentrations by anopioid-independent mechanism (53). It is now known thatOT is produced within human intrauterine, choriodecidualtissues. It remains to be established whether a similar rela-tionship exists between relaxin and OT synthesized withinthe intrauterine compartment in women.

Lye and Challis (54, 55) first showed, some 20 yr ago, thatprostacyclin infused into nonpregnant sheep inhibited uter-ine contractility in vivo. In parallel studies a similar inhibitoryeffect of prostacyclin was observed on human myometrium(56), and it is clear now that prostacyclin represents the majoreicosanoid present within the pregnant myometrium ofmany species (57), including human. In human term preg-nant myometrial strips maintained in vitro, the initial re-sponse to PGI2 was contraction, but this was followed byrelaxation (58, 59). It is now recognized that PGI2 actsthrough specific IP receptor species to increase adenylatecyclase activity and elevate intracellular cAMP (60). Otheragents such as CRH also stimulate output of cAMP frommyometrial cells and act synergistically with PGI2 in a para-crine/autocrine fashion (61). The role of CRH in pregnancymaintenance and parturition will be discussed later in thisreview.

More recently, interest has arisen over the potential role ofNO as an endogenous inhibitor of myometrial contractility(62). Increases in endogenous synthesis of NO by adminis-tration of the NO precursor l-arginine, or the NO donorsodium nitroprusside, inhibit myometrial contractions in therat and human (62). Nitroprusside has been shown to de-crease force and 20-kDa myosin light chain phosphorylationin human myometrial strips, although the tissue is not assensitive as vascular smooth muscle. Nitric oxide synthase(NOS) isoforms have been detected using RT-PCR in humanfetal membranes and choriodecidua (62). Levels of mRNA-encoding inducible NOS (iNOS) are highest in human myo-metrium at preterm, not in labor patients, and decrease with

October, 2000 PARTURITION 517

Page 5: PDF (454 KB)

a corresponding fall in iNOS protein in myometrium col-lected at term (see Ref. 10). Several authors have suggestedthat NO acts in a paracrine manner, potentially in conjunc-tion with progesterone to effect myometrial quiescence dur-ing pregnancy, although this position has been disputed.There is a decrease in NOS activity of decidua and myome-trium in species such as rat before parturition in a mannerthat would presumably diminish its inhibitory influence onthe uterus. Furthermore, studies by Chwalisz and Garfield(62) have shown that, at term in the rat, there is a corre-sponding increase in NO production by inflammatory cellsof the cervix, indicating a role for NO in cervical effacementand relaxation as its influence on the myometrium is dimin-ished.

Other inhibitors of uterine activity include calcitonin gene-related peptide (CGRP), vasoactive intestinal polypeptide(VIP), and endogenous b-adrenergic agonists (63). Thesecompounds act through increasing intracellular cAMPand/or decreasing intracellular calcium availability (64).

IV. Myometrial Activation: Phase 1 of Parturition

The switch from myometrial quiescence to myometrialactivation is essential to enable the muscle to respond to thestimulation provided by the high levels of uterotonic ago-nists and to generate the synchronous, high-amplitude, high-frequency contractions of labor. We have proposed that myo-metrial activation results from coordinated expression of acassette of proteins, termed contraction-association proteins,or CAPs (12). CAPs include ion channels [which determinethe resting membrane potential and hence excitability ofmyocytes (65)], agonist receptors [e.g., to OT and PG (60)] andGAP junctions [permitting cell-to-cell coupling (16)].

Overall regulation of myometrial activity is geneticallyregulated (Fig. 2). Different species have gestations ofvarying lengths, and studies involving embryo transfersuggest that it is the genotype of the fetus that controls thelength of pregnancy. For example, when sheep embryosfrom short gestation or long gestation breeds were im-planted into random gestation-age recipients, parturitionoccurred at the appropriate time for the fetal rather than

maternal genotype (66, 67). There is a variety of mecha-nisms by which the fetal genotype can influence preg-nancy length, and we have proposed that it includes bothendocrine and mechanical signals. In initial studies, Ouand Lye (68) found, using unilaterally pregnant rats, thatwhile expression of CAP genes, CX-43 and OT receptor(OTR), increased in the gravid uterine horn in labor, therewas no parallel increase in the nongravid horn, eventhough both horns were exposed to the same systemichormonal changes. Next, these workers showed that whena small 3-mm diameter tube was placed into one uterinehorn of bilaterally ovariectomized nonpregnant animals,there was a significant increase in mRNA levels encodingCX-43 in that horn, compared with the contralateral horn.Control experiments showed that this result was not dueto the presence of a foreign body within the uterus. Ad-ministration of progesterone to these animals blockedstretch-induced increases in CX-43 expression.

Subsequent experiments examined whether the endocrineenvironment of pregnancy influenced the ability of stretch toup-regulate CAP gene expression (see Ref. 8). In unilaterallypregnant rats, at day 15 of gestation, the nonpregnant hornreceived either the 3-mm Silastic tube or was left as control.Other animals were operated on at day 18. Five days afterimplanting the tubes, levels of transcripts encoding CX-43,PGF2a receptor (FP receptor), or OTR were measured. Inanimals treated at day 15 and studied at day 20, there wasno effect of the Silastic tube in increasing CX-43 transcripts,but in animals studied at the time of labor there was adramatic increase in the numbers of CX-43 transcripts tovalues similar to those seen in the contralateral pregnanthorn. There was little change in CX-43 transcripts in thenonpregnant control horn. These data suggest that stretch ofthe myometrium appears capable of up-regulating contrac-tion-associated proteins, but the ability to do so is highlydependent on the endocrine environment. If the stretch stim-ulus is applied during pregnancy, it is inadequate to induceCX-43, and presumably its activity is inhibited by circulatingconcentrations of progesterone. However, at term, when ma-ternal systemic progesterone levels have decreased, stretchitself is adequate to produce the same level of CX-43 expres-sion as in the pregnant horn containing the fetus.

The molecular mechanisms by which stretch increasesCX-43 and OTR expression remain to be determined (69).In other systems, such as cardiac myocytes, stretch acti-vates multiple intracellular signaling pathways throughshear stress response elements in the promoter of somestretch-responsive genes (70). The CX-43 gene containssuch an element, suggesting that if wall tension contrib-utes to the regulation of CAP genes in the myometrium,regulation of uterine growth through pregnancy will beimportant in determining the level of shear stress. Lye andcolleagues (8) have argued that, during pregnancy, uterinegrowth follows three distinct phases: an initial phase dur-ing the first trimester where uterine growth is due tohyperplasia and controlled by endocrine factors, a secondphase during the second and third trimester in whichgrowth is closely matched to increased fetal size, and afinal phase in which there is a decline in uterine growthin comparison to fetal growth, and hence an increase in

FIG. 2. The onset of labor is dictated by the fetal genome proceedingthrough either a fetal growth pathway with increases in uterinestretch or fetal endocrine pathway involving activation of the fetalHPA axis. These two arms are not independent because changes inprogesterone and estrogen modulate the ability of uterine stretch toincrease expressions of genes associated with myometrial activation.

518 CHALLIS ET AL. Vol. 21, No. 5

Page 6: PDF (454 KB)

uterine wall stretch and tension. They speculate that pro-gesterone is necessary to support stretch-induced hyper-trophy of the uterus during midgestation in concert withincreasing fetal size. Near term, the fall in progesterone,observed in most animal species (see below), leads to adecline in uterine growth relative to fetal growth andhence increased tension development, which in turn re-sults in increased CAP gene expression and contributes tomyometrial activation. Since the decrease in circulatingprogesterone appears critical for the altered influence ofstretch on myometrial CAP gene expression, we shall con-sider the endocrine pathways that result in progesteronewithdrawal.

A. Activation: role of fetal HPA maturation

More than 35 yr ago, Professors Sir Graham (Mont) Ligginsand Geoffrey Thorburn, working in the sheep and goat,showed conclusively in those species that the fetus, in utero,appeared to provide the trigger mechanism for the onset ofparturition and that it did so through activation of the fetalHPA axis. An endpoint of activation of this axis is proges-terone withdrawal. We shall suggest that the primate fetussimilarly affects gestation lengths through activation of theHPA axis. However, in human gestation there is no systemicprogesterone withdrawal, and we shall argue that, inwomen, sustained circulating concentrations of progesteroneare indeed required at term to effect regionalization of myo-metrial contractility and promote relaxation of the loweruterine segment.

Early studies in the fetal sheep showed that ablation of thefetal pituitary gland, the fetal adrenal gland, pituitary stalksection, or lesioning of the fetal paraventricular nucleus(PVN) resulted in prolongation of gestation (71–73), whereasthe infusion to the fetal lamb in utero of ACTH or of a glu-cocorticoid resulted in premature parturition within 3–5days of beginning the infusion. These studies provided ex-perimental verification of the concept developed from ob-servations of naturally occurring prolonged gestation insheep attributable to ingestion of the teratogen Veratrumcalifornicum at a specific time of gestation. In those animals,gestation length was prolonged by up to 60 or 70 days,although fetal growth continued. Fetuses exhibited grossmalformations, including cyclopean characteristics. At au-topsy, the pituitary and adrenal glands were remarkablyhypoplastic as a result of impaired pituitary development atan early gestational age (see Ref. 81).

Several groups of workers provided clear evidence formaturation of fetal HPA function in the sheep fetus duringlate gestation (74–76). There are progressive increases in fetalplasma ACTH1–39 and cortisol in the plasma of the late-gestation fetal sheep (77–80); the initial increases in ACTHprecede the rise in cortisol (79), but fetal cortisol increases inan exponential fashion over the last 10 days of gestation, withhighest concentrations being established immediately beforeterm (80). This is consistent with the fact that ACTH is im-portant in the development of the adrenal cortex in lategestation. Similar maturation of pituitary adrenocorticalfunction has been demonstrated in several other species,including the guinea pig, which represents a species that

gives birth to mature young. The prepartum surge of cortisolis important for the maturation of several organ systems,particularly the lungs and kidneys (see Ref. 81). It is alsocritical for normal development of programming of the brain.However, the simultaneous increase in fetal plasma ACTHand cortisol has remained somewhat of a paradox because,under normal circumstances, one would expect elevations infetal plasma cortisol concentration to inhibit further ACTHsecretion. Mechanisms have developed to override the in-fluence of negative feedback in the fetus in late gestation, arelationship now described in the guinea pig as well as in thesheep (see below).

Recent studies have explored the molecular mechanismsunderlying changes in fetal pituitary adrenocortical activa-tion in late gestation sheep. Developmental changes in CRHmRNA in the fetal hypothalamic PVN were examined by insitu hybridization (82). By day 60 of gestation, CRH mRNAwas detectable in the fetal PVN. There was an increase inCRH mRNA expression by day 120 of gestation and a furthersubstantial up-regulation of CRH gene expression in the last20 days of pregnancy. This was followed by a decrease inCRH expression in the PVN of the newborn lamb. Through-out development, expression of CRH mRNA appears to beconfined to parvocellular fields of the PVN, with no expres-sion detected in magnocellular neurons (82). Recent studieshave confirmed that the changes in CRH mRNA are trans-lated to CRH peptide in the fetal hypothalamus, indicatinga close association between transcription and translation ofthe CRH gene during development.

In the fetal pituitary, expression of the ACTH precursor,POMC, is detectable in the inferior region of the pars distalisby day 60 of gestation. Levels of POMC mRNA in the su-perior and inferior regions of the pars distalis increased withprogression of gestation until around day 120, when therewas a further increase in expression, peaking at term (83, 84).The increase in POMC expression is combined with a re-markable reorganization of the corticotrophs toward the in-ferior aspect of the fetal pituitary gland. This pattern ofexpression was sustained in the newborn lamb. In the fetalpars intermedia, the developmental profile of POMC mRNAwas quite different. Relatively high levels were present byday 60 of gestation; these increased between days 60 and 100and then remained relatively constant for the remainder ofgestation. Early controversy concerning changes in expres-sion of POMC mRNA in fetal pituitary tissue appears toresult from differences in methodologies. The use of in situhybridization clearly allows separation of different zones ofthe fetal pituitary gland, whereas erroneous results may havebeen obtained through use of Northern blot analysis (85, 86).In a recent carefully conducted study obtaining pituitarytissue from fetuses at specific times in late gestation andduring the labor process itself, the lack of negative feedbackon POMC mRNA, and the sustained increase in POMCmRNA levels, was clearly demonstrated (87). The change inregional distribution of POMC mRNA in the pars distalismay indicate the transition of fetal-like to adult-like cortico-trophs that has been described at this time (see below).Changes in POMC mRNA in the pars distalis are reflected byincreased levels of ir-ACTH and by increased immunostain-ing for ACTH in pituitary corticotrophs (83, 84); at term

October, 2000 PARTURITION 519

Page 7: PDF (454 KB)

ir-ACTH-positive cells represent about 15% of the total cellnumber in the pars distalis.

Arginine vasopressin (AVP) is also an important regulatorof fetal pituitary ACTH secretion and is expressed in the fetalPVN relatively early in gestation (88). AVP mRNA is presentin the supraoptic nucleus, PVN, and the accessory magno-cellular nuclei by day 60 of gestation (82). Differential ex-pression of magnocellular and parvocellular AVP is evidentin the PVN by day 80. In magnocellular neurons, AVP mRNAincreases with gestational age, whereas parvocellular expres-sion of AVP remains relatively unchanged. Levels of AVPmRNA increase dramatically in both regions of the PVN inthe newborn lamb. It is suggested that magnocellular AVP isinvolved primarily in fetal fluid homeostasis, while parvo-cellular AVP is important in stimulation of the pituitarycorticotroph (84). There is a close correlation between AVPmRNA levels and ir-AVP in the anterior hypothalamus, asthere is for CRH. The increase in parvocellular AVP mRNAin the newborn may be associated with the stress of the novelextrauterine environment. Axons containing AVP and OThave been identified in a zone of the pars distalis adjacent tothe pars intermedia in fetal sheep. These axons are probablythose of magnocellular neurons and may represent a mech-anism by which magnocellular AVP and OT directly affectACTH release in vivo.

CRH and AVP induced dose-dependent increases inACTH output from ovine fetal pituitary cells in vitro (89); atequimolar concentrations AVP was more potent than CRH.Simultaneous administration of CRH and AVP showed anadditive interaction between the neuropeptides (90). Treat-ment with CRH significantly increases POMC mRNA levelsin sheep pituitary cells harvested at day 120 and day 138 ofgestation. However, CRH treatment of cells collected fromfetuses at term failed to affect POMC synthesis. AVP in-creased POMC mRNA levels in cells obtained at day 138 ofgestation; in pituitary cells from late-gestation fetuses, AVPand CRH are equally potent in the induction of POMC syn-thesis. Cortisol has little negative feedback effect on basaloutput of ACTH in these cells but inhibits CRH-stimulatedACTH output and POMC gene expression.

Studies by Lu et al. (91) showed that ovine fetal pituitarymembranes expressed CRH receptor activity as early as day100 of gestation. CRH binding increased to its highest levelsat around day 135 (term, 145–150 days) and then decreasedprogressively through late gestation (92). Recent studieshave extended these measurements to show that levels ofmRNA encoding fetal pituitary CRH-receptor type I mayfollow a similar profile (J. C. Rose, personal communication),and this may account for the altered outputs of ACTH inresponse to CRH stimulation in vivo (see below). Factorsregulating CRH receptor expression have been examined invivo and in vitro. In vitro studies indicated that CRH down-regulated expression of its own receptor and cortisol pro-duced a similar attenuation of binding activity (92).

In vivo studies demonstrated that CRH was more potentthan AVP in stimulating ACTH output by pituitary tissuefrom chronically catheterized fetal sheep in late gestation (93,94). The response profiles, however, are quite different. AVPinduced a transient rise in plasma ACTH while CRH stim-ulated a more sustained increase (95). Subsequently, it was

demonstrated that AVP concentrations are about 5 timesthose of CRH in the hypophyseal portal circulation of adultsheep (96), and it remains possible that the relative impor-tance of AVP in fetal corticotroph activation in utero may begreater than that of CRH (97). Fetal pituitary responsivenessto CRH increases between day 110 and 125 and then de-creases toward term (79). This relative insensitivity of thepars distalis to CRH may reflect the increase in negativefeedback influence of rising endogenous cortisol concentra-tions, or the decrease in CRH binding sites indicated above(79). Simultaneous administration of CRH and AVP resultsin an ACTH response that is greater than when either neu-ropeptide is administered independently, and the interactionis synergistic in nature, at least at around day 115 of gestation(95). CRH and AVP affect the corticotrophs through differentsecond messenger systems. CRH exerts this action throughup-regulating a Gas-adenylate cyclase-linked membrane re-ceptor and increasing intracellular levels of cAMP (89). AVPacts through V1b receptors to stimulate PI turnover, stimu-lating phospholipase C and activating protein kinase C.

POMC is processed through different endoproteases, pro-hormone convertase 1 (PC-1) and prohormone convertase 2(PC-2), to yield a spectrum of products. Recent studies havedemonstrated that both PC-1 and PC-2 are present in fetalsheep pituitary tissue in late gestation. However, expressionof these enzymes does not change with labor, and it seemsunlikely that the increase in ACTH output is attributable toaltered prohormone convertase activity (87, 98). However,the pattern of POMC-derived peptides from the fetal pitu-itary does change in the plasma of the fetal lamb in latepregnancy (99). Several groups of investigators have re-ported that large molecular weight POMC-derived ACTHprecursor peptides are present in the circulation (100). Theconcentrations of these larger molecular weight forms de-crease prepartum, whereas those of ACTH1–39 increase. Be-cause the larger molecular weight peptides may act to an-tagonize the action of ACTH1–39 on adrenocortical cells (101–103), a decrease in their concentration prepartum wouldpresumably facilitate ACTH action and an increase in adre-nal glucocorticoid secretion (104). The sources of these pep-tides may be different (105–107). Studies in hypothalmo-pituitary-disconnected fetuses have led to the suggestionthat the pars intermedia may be a potential source of largemolecular weight peptides, whereas the pars distalis is theprimary source of ACTH1–39. In addition, the ovine fetal lungand placenta express POMC mRNA and contain ir-ACTH. Itis not clear whether these potential sources of ACTH con-tribute to circulating ACTH1–39 in a meaningful manner orwhether the peptides have paracrine/autocrine actionswithin the tissues of origin.

Thus, the temporal relationship between hypothalamic-CRH and pituitary POMC expression is consistent with thesimultaneous increase in plasma ACTH and cortisol ob-served in late gestation (84, 108–110). Nevertheless, themechanism by which CRH mRNA and POMC mRNA in-crease in the presence of high plasma glucocorticoid con-centrations is not clear. One possible mechanism is that, inthe fetus, glucocorticoid feedback thresholds within the brainand pituitary become modified. This may occur at severallevels (Fig. 3). We have reported that glucocorticoids up-

520 CHALLIS ET AL. Vol. 21, No. 5

Page 8: PDF (454 KB)

regulate expression of corticosteroid binding globulin (CBG)mRNA in the fetal liver, and of circulating CBG, which is theopposite of the response in adult sheep (111, 112). In the fetus,the pattern of CBG glycosylation varies from that in adultanimals, but the glycoprotein increases in concentration inthe fetal circulation and maintains a relatively constant freecortisol concentration for most of pregnancy (112, 113). Nearterm, however, the increase in adrenal cortisol output ex-ceeds the CBG binding capacity, resulting in a sudden in-crease in free cortisol concentration over the final hours be-fore birth (114). It appears that this increase in free cortisolbefore parturition is a consistent observation across differentanimal species (115). More recently, we have demonstratedexpression of CBG mRNA and the presence of CBG immu-noreactive protein in other fetal tissues including the kidney,pancreas, and pituitary (115). CBG mRNA has been localizedto fetal pituitary cells by in situ hybridization, and its patternof distribution appears to differ from that of POMC, withgreater abundance in superior regions of the gland. As yet,there are no studies demonstrating colocalization of CBGwith ACTH-producing cells in fetal pituitary tissue.

Levels of glucocorticoids may also be modified by inter-conversion of biologically active cortisol and biologicallyinactive cortisone, through the activity of 11b-hydroxyste-roid dehydrogenase (11b-HSD) enzymes (116). We will dis-cuss these later in the context of the placenta as a barrier tothe transfer of maternal cortisol to the fetus. In the pituitaryof fetal sheep, 11b-HSD-1 activity predominates and appearsto operate somewhat unusually in a dehydrogenase direc-tion, i.e., inactivating cortisol to cortisone (116). Presumably,this would effect a local mechanism to inactivate circulating

cortisol and diminish the potential for negative feedback.This pattern of 11b-HSD activity in the pituitary needs sub-stantiating and differs from that in other fetal tissues, e.g., theliver, where 11b-HSD-1 operates predominantly as a reduc-tase, converting cortisone to cortisol, and suggesting a po-tential intrahepatic source of cortisol generation.

A further mechanism by which glucocorticoid feedbackcould be altered locally is through modification of cortico-steroid receptor expression (117). The ovine fetal pituitaryexpresses type II glucocorticoid receptor (GR) from relativelyearly in gestation, and the levels of GR mRNA increasetoward term (118), consistent with glucocorticoid effects inmodulating the switch from fetal to adult corticotroph celltypes in the pituitary (106). During the course of labor, thereis a dramatic decrease in levels of GR mRNA in the fetal parsdistalis, suggesting that the potential for glucocorticoid neg-ative feedback decreases in the pituitary during the course oflabor. More important, perhaps, is the demonstration thatthere are decreases in immunoreactive GR in the hypotha-lamic PVN near term. These changes were specific to CRH-and AVP-positive parvocellular neurons. More recently, weshowed that GR mRNA levels in the PVN of fetal sheep andguinea pigs decrease in late gestation, and in fetal sheeplevels of GR mRNA in the hippocampus also fall prepartum.The hippocampus represents a major site of glucocorticoidfeedback for HPA function, and there are a number of directand indirect connections between the limbic system and thePVN. Together these data suggest that a reduction in thepotential for glucocorticoid feedback occurs in late gestationin brain structures that are central to glucocorticoid negativefeedback action (119).

In addition to classic feedback processes, there are severalother mechanisms by which fetal HPA axis activation mayoccur. Expression of pro-enkephalin mRNA rises to a max-imum in the parvocellular PVN of fetal sheep at day 135 ofgestation and then decreases in older animals (120). A fall inhypothalamic pro-enkephalin mRNA occurs with intrafetalinfusion of cortisol at day 135, suggesting that the prepartumrise in endogenous cortisol may inhibit parvocellular pro-enkephalin synthesis. CRH and met-enkephalin are presentin the same secretory granules in rodents, and met-enkepha-lin inhibits CRH-stimulated ACTH secretion from fetal pi-tuitary cells in vitro. Thus, a decrease in met-enkephalinproduction may facilitate corticotroph function near term(120). OT has been implicated in the control of ACTH se-cretion in adult sheep, and OT stimulates ACTH output fromthe fetal pituitary cells in vitro. OT mRNA is present in bothmagnocellular and parvocellular fields of the PVN and SONand follows a similar developmental profile to AVP mRNA,raising the possibility that it too may influence fetal pituitaryfunction.

In fetal sheep, the kinetically determined production ofcortisol from the adrenal gland increases during the last20–25 days of gestation (77, 121). In part, this results from theincrease in drive to the adrenal from rising levels of ACTH,but, in part, it is attributable to maturation of fetal adrenalfunction (122). Indeed, in hypophysectomized fetuses treatedwith a continuous low-level infusion of ACTH, plasma cor-tisol concentrations increased and parturition occurred at

FIG. 3. Summary of events associated with maturation and devel-opment of the HPA axis in the fetal sheep. Increased expression ofCRH from the PVN of the hypothalamus drives increased expressionof POMC in the anterior pituitary. POMC is processed to ACTH,which drives the adrenal gland. In the fetus normal negative effectsof cortisol on the hypothalamus and pituitary are diminished throughincreases in systemic corticosteroid binding globulin (CBG), pituitary11b HSD, and diminished expression of GR in the pituitary andhypothalamus.

October, 2000 PARTURITION 521

Page 9: PDF (454 KB)

around the normal time, consistent with fetal adrenal mat-uration as the overriding influence (123).

Ovine fetal adrenal responsiveness changes dramaticallyduring the course of pregnancy (124, 124–126). Adrenal cellscollected from animals at days 50–70 of gestation secretecortisol in response to ACTH stimulation in amounts similarto or greater than adrenal tissue from term fetuses (127).However, between approximately days 90–110 of pregnancythe adrenal is relatively insensitive to ACTH stimulation(124). It is now clear that this pattern of response is due, inlarge part, to decreased gene expression of P450C17 andP450SCC steroidogenic enzymes in fetal adrenal cortical cellsat midgestation (128, 129). The abundance of mRNAs forthese enzymes is increased by ACTH administration to thefetus (130, 131). Although 3b-HSD may be rate limiting tocortisol production in the first half of pregnancy (132), im-munoreactive (ir)-3b-HSD-positive cells are presentthroughout the zona fasiculata of the fetal adrenal cortexfrom day 50 until term (133). The midgestational decrease inP450C17 may result by TGFb inhibiting ACTH-induced stim-ulation to P450C17, as demonstrated in vitro in ovine fetal andadult adrenal cells (134). Recent studies have demonstratedthat ACTH receptor mRNA is detectable from around day 60of gestation (135). There is a modest increase through preg-nancy and then a substantial increase between days 126–128and days 140–141 (135). Thus, the low level of basal adrenalresponsiveness to ACTH around day 100 of gestation is notdue to lack of ACTH receptor expression, but may be at-tributable, in part, to very low concentrations of ACTH in thefetal circulation at that time (136). The increase in ACTHreceptor expression in late gestation would appear to con-tribute to increased adrenal responsiveness near term. Thefactors responsible for up-regulating ACTH receptor mRNAabundance are unclear (137). These may include ACTH itself,cortisol, or local intraadrenal interaction with IGF-II and/ordecreased influence of TGFb (138–140).

Both in vivo and in vitro studies have shown that fetaladrenal maturation can be advanced by ACTH1–24 admin-istration (110, 141–143). Exogenous ACTH in vivo enhancescoupling between ACTH receptor and adenylate cyclase andenhanced capacity for cAMP generation (144–146). ACTHtreatment in vivo also increased expression and activity ofP450C17, P450C11, P450C21, and 3b-HSD (130, 147). The adre-nal responds to ACTH early in gestation, although continuedtrophic input is required to maintain increased levels of geneexpression. Interestingly, when ACTH was administered tofetuses in vivo as pulses, rather than as a continuous infusion,it led to a pattern of fetal adrenal steroidogenesis that favoredcortisol over corticosterone output (i.e., directed P450C17 ac-tivity). Thus, the pulse pattern of endogenous ACTH secre-tion in vivo may affect the pattern of adrenal activation (148,149).

These studies suggest that ACTH-induced increases inadrenal steroidogenic enzymes, particularly P450C17, is es-sential to allow C21 steroids to proceed through the 17-hydroxy pathway leading to cortisol biosynthesis (130, 150).An obligatory role for an increase in ACTH drive to the fetaladrenal as a prerequisite for increased responsiveness, how-ever, has been challenged recently. When hypophysecto-mized fetal sheep were infused at a constant, but low level

of ACTH, there was a normal rise in fetal cortisol concen-tration; later, maternal progesterone levels decreased andbirth occurred at about the expected time (123). The molec-ular mechanisms underlying this fascinating result clearlyrequire elucidation.

We have hypothesized that fetal stress, perhaps reflectedin diminished fetal arterial P02, constitutes a stimulus forpreterm birth. Experimental hypoxemia has been used ex-tensively to investigate fetal HPA activation (151, 152). Manystudies have shown that even modest reductions in fetalarterial P02 induce robust increases in fetal plasma ACTHand cortisol concentrations (153, 154). Release of CRH andAVP into the hypophysial portal system is abolished in thehypothalamo-pituitary-disconnected (HPD) fetus (152), andthese animals are incapable of mounting an ACTH responseto stress, implying that increased ACTH output requireshypothalamic input. Studies by Akagi and colleagues (155)demonstrated that changes in fetal P02 of only 4–5 mm Hgwere adequate to elicit increased ACTH concentrations in thecirculation of the fetal lamb. This level of oxygen change issimilar to that seen during spontaneous contractures in lategestation sheep, raising the possibility that uterine activityitself may contribute part of the stimulus to increased fetalHPA maturation. Whether chronic stress is a stimulus tobirth at term (156) or contributes only to some cases of pre-term labor is unclear at the present time.

At 135 days’ gestation, hypoxia (P02 reduction by 8 mmHg) significantly increased CRH mRNA in parvocellularPVN and POMC mRNA in the pars distalis within 6 h. Thisresponse, however, was attenuated by concurrent infusion ofcortisol, indicating effective glucocorticoid feedback mech-anisms in vivo at this time (157). After 48 h of sustainedhypoxemia, levels of POMC in the pars distalis were ele-vated, but expression in the pars intermedia was decreased(158). This suggests differential regulation of these two zonesof the fetal pituitary, consistent with observations that do-pamine, likely from the fetal arcuate nucleus, tonically in-hibits pituitary POMC synthesis, and this inhibition is ex-acerbated in the presence of hypoxemia. Infusion ofbromocriptine, a dopamine D2 receptor agonist at day 130 ofgestation, produced a 50% decrease in pars intermediaPOMC mRNA levels, without affecting POMC mRNA in thepars distalis (159). Thus, the fetal D2 receptor system is func-tional in late pregnancy, but the fetal pars intermedia doesnot appear to secrete ACTH1–39 in amounts that alter fetaladrenal function.

Activation of fetal HPA function in response to hypox-emia, however, is a critical aspect of the story leading topreterm birth (160, 161). A sustained pulsatile hypoxemicstimulus is adequate to up-regulate HPA gene expression,plasma ACTH, and cortisol concentrations. It is reasonable topredict that sustained hypoxemia in conditions of fetal com-promise predisposes to fetal HPA activation and would re-sult in premature birth (162, 163).

B. Activation mechanism by which cortisol changesplacental steroid and PG synthesis (Fig. 4)

Fetal cortisol acts on the sheep placenta to alter the patternof steroidogenesis; as a result, progesterone output falls and

522 CHALLIS ET AL. Vol. 21, No. 5

Page 10: PDF (454 KB)

estrogen concentrations increase (164–167). These changes inplacental steroid output are associated with increased ex-pression and activity of placental P450C17 (168, 169). This isa critical difference between the sheep and the human, wherethis enzyme is not induced in the placenta at term. Ovineplacental tissue contains P450arom activity, and up-regulationof this gene also occurs in late gestation. For many years thegeneral thesis has been that placental estrogen production islimited in ovine pregnancy and occurs in abundance only atterm with the induction of placental P450C17 as a result ofglucocorticoid action (170–172). The fall in progesterone andlater increase in maternal and fetal estrogen concentrationshave been considered as providing the stimulus to increasedPG output by intrauterine tissues, with consequent increasein myometrial contractility (173–180).

For several reasons we have questioned the appropriate-ness of this model. It has been clearly established that thesheep, like the human, has a feto-placental unit of estrogenproduction by which C19 precursors from the fetal adrenalgland can be secreted and aromatized in the placenta to formestrogen (181). Later studies demonstrated output of C19steroids including dehydroepiandrosterone (DHEA) and an-drostenedione by the ovine fetal adrenal gland, stimulationof C19 fetal-adrenal steroid output by ACTH infusion and inresponse to hypoxemia, and conversion of [3H]androstenedi-one infused into the fetus to estrogen measured in maternal

and fetal compartments (182, 183). Although unconjugatedestrogens increase sharply at the time of parturition in sheep(165, 166), there is a progressive increase of conjugated es-trogens in maternal plasma and urine throughout the latterpart of gestation, well before the terminal increase in pla-cental P450C17 activity (184). The ratio of conjugated to un-conjugated estrogen in maternal sheep plasma is high be-cause, in ovine pregnancy, placental sulfotransferase activitypredominates over placental sulfatase activity (184). Thus, itis clear that while increased expression of placental P450C17may contribute to the sharp rise in maternal estrogen con-centrations prepartum, its induction in the placenta is not aprerequisite for ovine placental estrogen output at earlierstages of gestation.

There are other troubling features of the currently ac-cepted model (185). Several groups of investigators haveused either immunohistochemical techniques for localizationof PGHS-1/-2, or PGHS-2, or in situ hybridization forPGHS-2 mRNA, or measurements of PGHS and/or PGHS-2activity in ovine placental cells and microsomal preparations(186–189), to show that PG production by the sheep placentaincreases progressively through the last 20–25 days of ges-tation (190–195). Placental output of PGs is not confined tothe immediate 24–48 h before spontaneous parturition (196,197). The increase in PGHS expression and activity in theplacenta correlates closely with the progressive increase in

FIG. 4. Endocrine pathways leading to the onset of parturition in sheep. A, current model; B, proposed sequence of hormone events. In thecurrent model, activation of the fetal HPA axis leads to increased cortisol thought to up-regulate expression of P450C17 in the placenta. In thenew proposed hypothesis, increased fetal adrenal output of cortisol results in up-regulation of prostaglandin synthase (PGHS)-2 gene expressionin the placenta with increased production of PGE2. PGE2 feeds back to further up-regulate fetal HPA function but is itself responsible forup-regulation of P450C17 gene expression in the placenta. Increased placental estrogen is required for up-regulation of PGHS-2 in maternaltissues but not in fetal tissues. Thus, with the onset of parturition there is progression from fetal trophoblast within the placenta to the maternaluterine tissues.

October, 2000 PARTURITION 523

Page 11: PDF (454 KB)

plasma PGE2 concentrations in the circulation of the chron-ically catheterized fetal lamb (191, 198). The increase in cir-culating PGE2 in the fetus bears a striking temporal relation-ship to the increase in plasma cortisol concentration (198,199). Louis et al. (200) first reported, more than 25 yr ago, thatinfusion of PGE2 into the ovine fetus in late gestation stim-ulated an increase in the plasma cortisol concentration at atime when the fetal adrenal gland was relatively unrespon-sive to ACTH stimulation. Later studies have shown that theeffect of PGE2 infused into the fetus on fetal HPA functioncould be exerted at any one or all of the hypothalamic, pi-tuitary, or adrenal levels (201, 202). Thus, the progressiveincrease in output of PGE2 appears to contribute to the driveto fetal HPA function and augments the stimulus suppliedby ACTH to the fetal adrenal (201, 203). Indeed, fetal PGE2infusion will provoke premature delivery of the ovine fetus(204). Placental PGE2 output would not be subjected to neg-ative feedback regulation by cortisol and may contribute tothe apparent lack of negative feedback relationship betweenACTH and cortisol in the late gestation ovine fetus.

Recent studies have suggested that in addition to PGE2stimulating output of cortisol by the fetal adrenal gland (205,206), fetal cortisol, and not estrogen, may affect placentalPGHS-2 activity and contribute to the rise in fetal plasmaPGE2 concentrations. Evidence in support of this suggestionincluded the observation that infusion of estrogen into fetallambs in late pregnancy was without stimulatory effect onlevels of placental PGHS-2 mRNA (207), although estrogeninfusion into nonpregnant adult sheep did increase PGHS-2expression in the endometrium (see also below). Studies withhuman amnion cell cultures and chorion trophoblast cellshave suggested that glucocorticoids may up-regulatePGHS-2 gene expression in these tissues. Infusion of cortisolto fetal sheep in late gestation also increased levels of PGHS-2mRNA and immunoreactive PGHS-2 protein (by Westernblotting) in placental trophoblast cells. This effect was inde-pendent of changes in estrogen, since a similar stimulationof placental PGHS-2 mRNA levels was observed when cor-tisol was infused in the absence or presence of the aromataseinhibitor, 4-hydroxyandrostenedione.

Using immunohistochemistry we showed that the P450C17enzyme and PGHS-2 both localized to trophoblast epithelialcells, but not binucleate cells in ovine placentomes (208).Moreover, the appearance of ir-PGHS-2 clearly preceded thatof P450C17. Collectively, therefore, these data offer strongreasons to refute the current model of endocrine events oc-curring in the placenta of the sheep in late gestation andsuggest that a different sequence likely pertains. This is sum-marized in Fig. 4. We have argued elsewhere that during lategestation in the fetal sheep, increased output of cortisol fromthe fetal adrenal gland progressively up-regulates PGHS-2gene expression in placental trophoblast cells (208). Themechanism of this action remains unresolved. It may dependon trophoblast-specific transcription factors generated in re-sponse to elevations of cortisol, or it could be a direct actionof cortisol since early studies reported a full GRE consensussequence at approximately 760 bp upstream from thePGHS-2 transcription start site. We suggest that increasedPGHS-2 expression in the sheep placenta contributes to in-creased PGE2 output into the fetal circulation. Fetal PGE2

drives the fetal HPA axis in a positive feed-forward fashion(Fig. 4). PGE2, and not cortisol, is responsible for up-regu-lation of P450C17 in placental trophoblast cells. This occurs ina manner analogous to the effect of PGE2 on P450C17 induc-tion in ovine and bovine adrenal tissue. Ovine placentaltissue expresses PGE receptor subtypes (EP1-EP4), but anychanges in their expression during the course of late gesta-tion remain to be determined (see Ref. 208). We have sug-gested further that increased P450C17 in the placenta allowsthe conversion of C21 D5 steroids directly through to D5 C19steroids, precursors for estrogen biosynthesis, as demon-strated by Flint et al. (209) and Mason and colleagues (210)some years ago. A crucial difference of the current hypothesisis that this change is superimposed on an already substantialbasal output of estrogen by the sheep placenta (measured asconjugated estrogens in maternal plasma and urine), andcontributes principally to the terminal increase in maternalestradiol concentrations. This increase in estrogen is requiredfor expression of CAP genes in the ovine myometrium andfor expression of PGHS-2 in maternal endometrial tissue,predominantly endometrial epithelium. We have found thatwhereas the increase in placental (fetal trophoblast) expres-sion of PGHS-2 after intrafetal cortisol administration wasunaffected by concurrent infusion of 4-hydroxyandro-stenedione, maternal endometrial up-regulation of PGHS-2and output of 13–14 dihydro-15-keto PGF2a (PGFM) into thematernal circulation occurred with cortisol infusion but wasblocked by concurrent administration of the aromatase in-hibitor (211). Thus, in sheep it appears that the fetal placentaand maternal endometrium exist as two separate sites of PGsynthesis in late gestation and that these are differentiallyregulated. In fetal placenta, PGHS-2 is increased by cortisol,independent of changes in estrogen output, whereas in ma-ternal uterine tissue, up-regulation of PGHS-2 and maternalplasma PGFM is dependent upon increased estrogen pro-duction (Fig. 4).

Current studies are directed at examining this hypothesisfurther. Using immunohistochemistry and Western blotanalysis, it is evident that GR is expressed in ovine placentaltissue, predominantly in uninucleate trophoblast cells. Es-trogen receptor (ER) mRNA and activity have been demon-strated in maternal endometrium but is apparently lackingin placental trophoblast (212). Hence, it is difficult to envis-age how estrogen could provide a stimulus to placental PGproduction as previously hypothesized. It remains to beshown whether glucocorticoids affect placental PGHS activ-ity directly or indirectly. However, in early studies we havedemonstrated that glucocorticoids increase output of PGE2by ovine placental trophoblast cells maintained in culture,and this effect is abolished by addition of meloxicam, a spe-cific inhibitor of PGHS-2 activity.

C. HPA function in the primate fetus and activationof parturition

The role of the human and subhuman primate fetus incontrolling gestation length has been, until recently, lessclearly defined than that of the sheep fetus. However, overthe past few years it has become apparent that mechanisms

524 CHALLIS ET AL. Vol. 21, No. 5

Page 12: PDF (454 KB)

leading to activation of fetal HPA function in primates bearconsiderable similarity to processes in sheep, and that fetalcortisol and fetal adrenal C19 steroids appear to play animportant role. In 1933, Malpas (213) in a study of gestationlength in human pregnancies complicated with anencephalyconcluded that “. . . . the fetal pituitary and adrenal glandswas responsible for the trigger to the neuromuscular expul-sive mechanism that led to the onset of labor. ” Early ob-servations indicated that the mean length of gestation inanencephaly, after exclusion of cases with polyhydramnios,was similar to controls, but the proportions of preterm andpostmature births were both higher (see Ref. 12). Similarresults have been obtained after experimental anencephalyin rhesus monkeys (214). In monkeys, fetal hypophysectomypredisposed to prolongation of gestation (215), but fetal ad-renalectomy was without effect on gestational length, al-though five of eight fetuses died in that study (see Ref. 12).Initial studies indicated that removal of the fetus, but leavingthe placenta in utero (fetectomy) had little effect on gestationlength. However, more recent studies have indicated clearlythat placental retention after fetectomy was significantlylonger (195 days) compared with 164 days in controls (216).Fetectomy in baboon pregnancy did not affect gestationlength, although maternal estradiol concentrations fell tobasal values and progesterone concentrations were reducedby 20–45% (217–219). Overall, these experiments are difficultto interpret. The numbers and observations are invariablysmall, no attempt is generally made to sustain uterine vol-ume and the stretch stimulus to the myometrium, and it istechnically very difficult to operate on the primate fetuswithout stimulating uterine contractility.

In intact rhesus monkeys, as in the baboon and human,there is an increase in maternal estrogen concentrations inlate gestation that parallels an increase in the concentrationsof fetal adrenal C19 steroids, particularly DHEA and DHEA-sulfate (DHAS) (220, 221). Maternal estrogen concentrationsincrease progressively and then more rapidly in the laterphases of human gestation; estriol, derived in substantialpart from precursors of fetal adrenal origin, rises rapidly inmaternal plasma and urine in late pregnancy at term, and inpreterm labor (221). When androstenedione was infused intopregnant rhesus monkeys at about three-quarters of the waythrough gestation, there was an increase in maternal plasmaestrogen concentrations and premature birth (222). This ef-fect was blocked by the coinfusion of the aromatase inhibitor4-hydroxyandrostenedione, which prevented maternal en-docrine changes and changes in fibronectin in the fetal mem-branes and inhibited the nocturnal increases in uterine myo-metrial contractility (223). Elevations of maternal systemicestrogen concentrations by infusion increased myometrialactivity, but did not produce premature delivery or fetalmembrane changes. It was suggested that in the primate, asin the sheep, estrogen is important for the normal processesof parturition. The failure of exogenous estrogen to stimulatesustained uterine contractility, even though locally producedestrogen formed after C19 steroid infusion was effective, ledthe authors to suggest that the estrogen had to be generatednear to its site of paracrine/autocrine action (223).

D. HPA maturation in the primate fetus

There is emerging strong evidence that maturation of HPAfunction occurs in the primate fetus in a manner generallyanalogous to that discussed above in the sheep fetus. Excel-lent reviews by Pepe and Albrecht (221, 224) and by Mesianoand Jaffe (225) have provided detailed analyses of pituitary-adrenal function in the primate fetus. In the human, baboon,and monkey fetus the pituitary is necessary for adrenal mat-uration and steroidogenesis, at least during the second halfof gestation. Adrenal development is impaired in anence-phalic human fetuses. In the baboon fetus treated in lategestation with betamethasone, there was suppression of fetalpituitary POMC mRNA and reductions in fetal adrenalweight, and 3b-HSD fetal adrenal ACTH receptor mRNAlevels (221). The authors concluded that increased expressionof fetal adrenal ACTH receptor and mRNA species encodingsteroidogenic enzymes depended upon fetal pituitary ACTHstimulation.

In the human fetus, ACTH activity is present in the pitu-itary by 5 weeks’ gestational age, and CRH- and AVP-likeactivity is present in the fetal hypothalamus by approxi-mately 12 weeks gestation (226). CRH1–41, in addition to alarge molecular weight form of CRH, are contained withinthe human fetal hypothalamic tissue. CRH and AVP syner-gize in promoting ACTH release from the human fetal pi-tuitary tissue in early gestation, and the stimulatory effect ofCRH and ACTH output was reproduced by 8-bromo-cAMP(see Ref. 12).

Levels of POMC mRNA in anterior pituitary tissue fromfetal baboons increased significantly from mid (day 100) andlate (day 165) gestation (term 5 day 184) in nontreated an-imals, and there was a corresponding increase in pituitarycells expressing ACTH peptide (227, 228). In the baboon ithas been suggested that this increase in fetal pituitary POMCmRNA levels might be associated with increased pituitaryCRH receptor activity, rather than increased expression ofCRH peptide in hypothalamic nuclei. However, administra-tion of estrogen to midgestation baboons resulted in an in-crease in levels of POMC mRNA- and ACTH-positive cor-ticotrophs in pituitary tissue to values that approached, butremained significantly different from, those at term (228).Pepe et al. (229) have argued that this increase in POMC issecondary to an effect of estrogen on placental 11b-HSDactivity, particularly 11b-HSD-2. In previous studies, theseinvestigators have shown increased expression of placenta11b-HSD-2 in the baboon during pregnancy and have shownthat activity of this enzyme is increased by treatments thatincrease estrogen and decreased with inhibition of estrogenproduction or action (221, 229). In midgestation, the rela-tively lower levels of placenta 11b-HSD-2 allow passage ofmaternal cortisol into the fetal compartment and relativesuppression of fetal HPA activity (221). With increased 11b-HSD-2 activity at day 160, there would be diminished ma-ternal cortisol reaching the fetus (230), allowing the fetalHPA axis to escape from the presumed negative feedback ofmaternal cortisol. This would allow increases in POMC geneexpression, ACTH output, and fetal adrenal maturation.These results are compatible with observations that produc-tion of cortisol by the primate fetal adrenal gland is relatively

October, 2000 PARTURITION 525

Page 13: PDF (454 KB)

low for much of gestation (231, 232). The bulk of the glandis occupied by the fetal zone with relative deficiency of 3b-HSD, and predominant formation of C19 D5 steroids, par-ticularly DHAS (233–235). In late gestation, there is an in-crease in ACTH receptor mRNA and 3b-HSD activity in thedefinitive zone of the fetal adrenal, and a decrease in ACTHreceptor mRNA and formation of DHAS in the fetal zone(236–238). The expression of fetal adrenal enzymes P450C17

and P450SCC remained relatively unchanged during gesta-tion. Thus, there are subtle differences between fetal adrenaldevelopment in the primate and sheep. In the former, ex-pression of 3b-HSD appears rate limiting toward adrenalcortisol output whereas in the ovine species, expression ofP450C17 appears to regulate fetal adrenal steroidogenesis.

In primate pregnancy, estrogen production in the placentadepends extensively on the provision of C19 precursor ste-roids, predominantly from the fetal adrenal gland (239, 240).Fetal adrenal DHAS can be converted to estrone and estra-diol in the placenta, and approximately 50% of circulatingmaternal estrone and estradiol are derived from placentalaromatization of fetal DHAS; the remainder is formed frommaternal adrenal C19 steroids (239, 241). Activation of thepituitary-adrenal axis of the fetus occurs in late gestation.There is a progressive increase in the concentration of DHASin the fetal circulation, which mirrors an increase in maternalplasma estriol concentration (maternal estriol is formed inthe placenta from the precursor 16-hydroxy-DHAS that is90% of fetal origin and formed in the fetal liver from adrenalDHAS). This pattern of fetal adrenal activation, reflected inplasma DHAS concentrations, resembles the time course ofincrease for plasma cortisol in the fetal sheep. Recent studieshave shown that the fetal adrenal in primates is divided intothe outer adult zone that produces predominantly aldoste-rone, the fetal zone that produces DHAS, and the transitionalzone, interposed between the adult and fetal cortex, whichproduces predominantly cortisol (225). Thus, the elegantstudies of Mesiano and Jaffe (225) and Coulter and colleagues(242), have shown that P450scc is expressed throughout theprimate fetal adrenal gland. P450C17 is not expressed in thedefinitive zone but is expressed in the transitional and fetalzones. P450C21 is expressed throughout the gland. 3b-HSD isnot expressed in the fetal adrenal at midgestation but isexpressed in the definitive and transitional zone in late ges-tation fetuses. P450C11 is expressed in the transitional zone inmidgestation and throughout the fetal adrenal cortex in lategestation. ACTH stimulates steroidogenesis in the transi-tional and fetal zone; the major products in late pregnancyare cortisol from the former and DHAS from the latter. Bothin vitro and in vivo studies show dependence on ACTH forfetal adrenal steroidogenesis. More recent studies, however,have indicated that CRH, potentially of placental origin (seebelow), can also stimulate the fetal zone to produce DHAS(243). In addition, this zone of the fetal adrenal appears torespond to trophic inputs from the fetal pituitary other thanACTH. ER-a/b mRNA is also expressed in fetal and defin-itive-transitional zones of the baboon fetal adrenal cortex atmid- and at late gestation (244). The presence of ER in theadrenal cortical cells provides an additional mechanism bywhich estrogen mediates ACTH-dependent functional mat-

uration of the primate fetal adrenal gland. In addition, pre-vious studies had shown that estrogens increase availabilityof LDL-cholesterol as precursor for adrenal steroidogenesis(245, 246).

The difference in fetal adrenal architecture between thesheep and primate fetus has been regarded by many as a clearobstacle to extrapolating from the sheep model of parturitionto the primate. However, it is now apparent that similaritiesbetween these species are greater than the perceived differ-ences (247). In both the sheep and primate fetus the fetaladrenal produces increased amounts of cortisol in late ges-tation (247). It is relatively unprofitable to make detailedcomparison of the minutiae of temporal changes in plasmacortisol because of differences in binding to circulating CBG,transplacental transfer from the mother, and tissue levels of11b-HSD isozymes in the fetus that could locally regulatecortisone-to-cortisol interconversion. In both sheep and pri-mate, the feto-placental unit also produces increasedamounts of estrogen. In the primate, that estrogen resultsprimarily from placental aromatization of precursors gen-erated within the fetal (and to a certain extent maternal)adrenal. There is no induction of placental P450C17 at term,and the primate placenta does not metabolize C21 steroidsthrough to estrogen. In the sheep, a similar fetal-placentalunit of estrogen production exists in pregnancy. The majorfetal adrenal precursors are both D5 and D4 C19 steroidsproduced from the developing zona fasiculata reticularis. Atterm, the prepartum rise in fetal cortisol results directly orindirectly in increased expression of P450C17 in the ovineplacenta, which at that time becomes capable of metabolizingD5 C21 steroids to estrogen. Thus, the apparent difference inthe pattern of estrogen biosynthesis between sheep and pri-mate at term, in its simplest term, reflects the source of C19precursor steroid. The mechanisms of HPA activation mayvary. However, in the primate, the C19 precursor comes fromthe fetal zone of the fetal adrenal gland. In the sheep, thatprecursor comes in part from the fetal adrenal, but there areadditional estrogen precursors produced in the placenta un-der the influence of cortisol from the fetal adrenal gland. Wesuggest that these differences are ones of degree rather thanof absolute distinction.

The role of estriol in the processes leading to the onset ofhuman parturition has remained unresolved over manyyears. Maternal estriol concentrations reflect fetal hepatic16-hydroxylation of DHAS produced from the fetal adrenalgland. It might be anticipated that estriol concentrations inthe maternal circulation would increase in response to fetalstress and might be predictive of impending preterm deliv-ery. Maternal estriol levels increase exponentially towardnormal term. Lachelin and colleagues (248, 249) have shownthat maternal plasma and salivary estriol concentrations areelevated further in a subset of patients with diagnosis ofpreterm labor. Since estriol may affect uterine CAP geneexpression (249), it could contribute to the progressive in-crease in uterine responsiveness in primate pregnancy dur-ing the third trimester of gestation, and its measurementsmay be of predictive value in delineating patients at risk ofpremature delivery (249, 250).

526 CHALLIS ET AL. Vol. 21, No. 5

Page 14: PDF (454 KB)

E. Placental progesterone and human pregnancy: theenigma of the progesterone block

A fall in the plasma progesterone concentration is thesingle most common endocrine event associated with par-turition across species (12, 250, 251). Administration of ex-ogenous progesterone at term not only blocks the expressionof CAP genes, but blocks the onset of labor (252). Even in thehuman, where there is no evidence of a fall in maternalplasma or uterine tissue progesterone, administration of theprogesterone receptor (PR) antagonist RU486 leads to in-creased uterine activity and induction of labor (253). In hu-man pregnancy, the luteoplacental shift in progesterone pro-duction occurs by 5–6 weeks’ gestation (254). Progesteroneis synthesized from pregnenolone by placental syncytiotro-phoblast and by chorionic trophoblasts (Fig. 6 and Ref. 255).However, the levels of 3b-HSD mRNA, protein, and activitydo not change in these tissues with labor at term or preterm(256), although regional changes in 3b-HSD expressionmight still occur (257). For example, expression of 15-hydroxyprostaglandin dehydrogenase (PGDH; the major PGmetabolizing enzyme) in chorion is regulated by progester-one (see below) and levels correlated with 3b-HSD in tissuecollected adjacent to the placenta, but not in the cervicalregion. In this lower segment, it was suggested that the actionof progesterone in maintaining PGDH tonically was over-come near term by the inhibitory influence of proinflamma-tory cytokines (see below). There are reports of an increasein the estrogen-progesterone (E:P) ratio in amniotic fluid ofwomen during labor; however, these changes are not im-pressive (250). We have referred to suggestions that maternalestriol, which increases during term and preterm labor,might promote myometrial activation and labor contrac-tions, but this possibility requires stronger experimental ver-ification (249). Alternatively, another progesterone-like ste-roid, possibly a progesterone metabolite that interacts withthe PR, might serve as the active progestagen in humanpregnancy and decline before labor, or progesterone could beconverted to an inactive metabolite that displaces proges-terone from its receptor (258–260). To date, there are no cleardata to support either of these possibilities. Erb et al. (261)reported recently that levels of allopregnanolone, the 3a,5a-reduced metabolite of progesterone that can bind to g-ami-nobutryic acid-A receptors and inhibits uterine smooth mus-cle, did not decrease with labor. The 5b- metabolite blocks OTbinding to its receptor and inhibits OT-induced contractionsin the human myometrium. However, there is also no evi-dence that levels of this metabolite decrease at term.

Studies of gene expression in the human myometriumhave focused on the lower uterine segment. These studiessuggest that the PR system is functional in this region duringlabor. Increased expression of progesterone-responsivegenes such as CX26 (which would promote relaxation) raisethe possibility that elevated levels of progesterone are re-quired to support establishment of a functional (inhibitory)lower uterine segment during labor. If this were so, it wouldalso require mechanisms within the fundus that would blockthe actions of progesterone, allowing the expression of CAPS,and promote contractility in that region.

Although recent exciting data have shown that proges-

terone can bind directly to the oxytocin receptor (OTR) andinhibit its signaling (262), the majority of the actions of pro-gesterone are mediated through a nuclear ligand-inducibletranscription factor, the PR. It has been suggested that afunctional withdrawal of progesterone may involve antag-onism of its action at the level of the PR or PR interaction withtranscriptional machinery (8). This might include a decreasein PR expression, a switch in PR isoforms, a change in ex-pression of receptor accessory proteins (e.g., heat shock pro-teins and receptor coactivators/repressors), or increased ex-pression of endogenous antagonists of progesterone or PR(such as cortisol, TGFb, or phospholipids). Three isoforms ofthe PR have been described: the full-length PR-B and thetruncated isoforms, PR-A and PR-C. In mammals, PR-B func-tions predominantly as an activator of progesterone-respon-sive genes, while PR-A acts as a modulator or repressor ofPR-B function and of other nuclear receptors including theGR, possibly because it lacks one of the three activationfunction domains (AF3) contained within PR-B (263). Nota-bly, progesterone repression of estrogen-induced gene ex-pression was effected through PR-B and not through PR-A.The expression of PR-A and PR-B isoforms is regulated dif-ferentially during development and by hormone treatment.The PR-C isoform (;60 kDa), which has C-terminal trans-activating domains and lacks the first zinc finger of the DNAbinding domain, can dimerize with and modify (possiblyinhibit) transcriptional activity of both PR-A and PR-B.

Analysis of PR expression is complicated by the multiplemRNA and protein species of the receptor. A decrease in PRimmunostaining in myometrium at term has been reportedbut, given the multiple isoforms of PR, these data are difficultto interpret. There is no change in PR mRNA in myometriumor membranes with labor, and no evidence of change in PR-Bor A 1 B mRNA nor in any immunoreactive PR isoforms insamples of lower segment myometrium during labor that mightindicate a decrease in progesterone signaling (G. Erb, N.McLusky, and S. J. Lye, unpublished results). There wasincreased expression of heat shock proteins (HSP)-90 andHSP-56 as well as the steroid receptor coactivators SRC-1 andTIF-2 (G. Erb and S. J. Lye, unpublished results). These co-activators may interact with several steroid receptors, butany interaction with PR should increase rather than decreaseits transcriptional capability. There are limited data on ERexpression in myometrium with labor. However, in thelower uterine segment at term, ER mRNA, protein, and high-affinity binding all appear to be very low.

There are several candidates for potential endogenous an-tagonists of progesterone action. TGFb has been proposed asan endogenous antiprogestin that reduces progesteronestimulation of genes such as enkephalinase (264). Othershave reported that a phospholipid extract of human fetalmembranes was capable of inhibiting progesterone binding,but not estrogen binding. Cortisol itself may compete withprogesterone in the placenta or membranes to regulate thegene for CRH (263). We have found (see below) that whileprogestagens such as medroxyprogesterone acetate (MPA)increase PGDH activity in human placental and chorion tro-phoblasts, this effect is reversed by cortisol. At the presenttime, it is not clear whether these are separate actions throughGR and PR, or whether cortisol and MPA compete for PR-GR

October, 2000 PARTURITION 527

Page 15: PDF (454 KB)

binding. Although four upstream GREs have been identifiedwithin the PGDH promoter, no putative PRE has been iden-tified. Cytokines [interleukin-1b (IL-1b), tumor necrosis fac-tor-a (TNFa)] also decrease PGDH activity, but their inter-action with progesterone as putative antiprogestins remainsunexplored. In recent studies, Stevens et al. (265) reportedthat CRH receptor type 1 (CRH-R1) was expressed prefer-entially in myometrium and fetal membranes of human ges-tation. Levels of CRH-R1 increased in myometrium collectedfrom patients in term and preterm labor but, importantly,levels of CRH-R1 in lower segment myometrium were con-sistently much higher than levels of CRH-R1 in the fundalregion (265). CRH acts through CRH-R1 to increase levels ofcAMP and promote uterine relaxation (61). We thereforeproposed that the role of CRH-R1 in the lower uterine seg-ment was to promote relaxation of this region during laborand to facilitate descent of the fetus (61, 265). These dataindicated that there might be mechanisms by which CRH-R1expression was regulated differentially in the fundus and thelower segment during labor. In independent studies, Spareyet al. (266) reported that levels of PGHS-1 and PGHS-2 pro-teins were also expressed at greater levels in the lower thanupper uterine segment. Connexin-43 protein, in contrast, wasexpressed at much greater levels in the upper uterine seg-ment. Myometrial GSa protein was uniformly expressed inboth lower and upper segments and down-regulated at thetime of parturition. These authors also concluded that dif-ferential expression of these genes might be important toallow cervical ripening before and dilatation during labor,with orderly propagation of uterine contractions (266).

Our own data suggest considerable differences in the ex-pression of CAP genes in the human myometrium duringlabor compared with other species. In contrast to observa-tions in myometrium of rats, sheep, and cows, Teoh et al. (267,268) did not observe any increase in the expression of CAPgenes, including CX-43, OTR, and the PG receptors that arelinked to stimulation of contractile pathways (FP, EP1, andEP3 receptor subtypes, including four splice variants of theEP3 receptor) in lower segment myometrium at labor. How-ever, Teoh et al. (267) did observe increased expression ofconnexin-26, the EP4 receptor and CRH-R1 receptor thatmight be expected to promote myometrial relaxation after anincreased generation of cAMP. It is known that connexin-26is positively regulated by progesterone.

What is the relevance of these observations to the effect ofprogesterone on the myometrium and the apparent lack ofwithdrawal of the progesterone block to the myometrium inhuman pregnancy? We propose that the biological basis forthe onset of labor in animals and in humans is essentiallysimilar. Both require activation of the myometrium and thegeneration of uterotonins to generate labor contractions. Inhuman fetal membranes and myometrium, however, re-gional differences in gene expression allow functional au-tonomy during labor. We suggest that this functional au-tonomy may be critical for the efficient and effective deliveryof the fetus and speculate that this is a mechanism associatedwith evolution to bipedal life. We have suggested that thisregionalization is established through the action of proges-terone. Early studies, e.g., those of Wiqvist and colleagues(269), support this hypothesis. These authors found that

PGF2a had little effect on the fundal myometrium, but wasstimulatory in lower segment specimens taken before labor.PGE2 induced a biphasic dose-dependent response. How-ever, PGF2a and PGE2 always stimulated fundal myome-trium collected during spontaneous labor. PGE2 inducedinhibition in lower segment samples collected at that timewhile PGF2a had no effect.

We speculate that during pregnancy, progesterone limitsthe generation of stimulatory PG in chorion by inducing highexpression of PGDH (see below), and it also inhibits theexpression of CAP genes within the myometrium, therebymaintaining the muscle in a quiescent state (8). Functionalregionalization of both chorion and myometrium at term isengineered by progesterone. In the cervical, but not fundal,region of chorion, there is a local decrease in PGDH (10),increased production of PGE, and later matrix remodeling.In the myometrium, functional withdrawal of progesteronein the fundus induces CAP gene expression and myometrialactivation. Enhanced progesterone signaling in the loweruterine segment, however, promotes the expression of genesthat induce relaxation, facilitating descent of the fetus (8). Themechanisms inducing functional withdrawal of progester-one in fundal myometrium and cervical chorion need notnecessarily be the same (270). Cortisol and/or cytokines mayantagonize progesterone- induced PGDH activity in chorion(see below). In myometrium, potential mechanisms includechanges in PR isoforms, steroid receptor co-activator/repres-sors, or other putative antagonists of progesterone action. Wespeculate that this concept of human labor provides an ex-planation as to why progesterone levels remain high in thisspecies. Rather than being an impediment to labor onset, wesuggest that progesterone is required to induce lower seg-ment relaxation and the safe and efficient delivery of theprimate fetus.

Recent exciting studies have pointed to a role for proges-terone in maintaining cervical function during pregnancy,and to metabolism of progesterone within the cervix as beinga critical step in cervical dilatation and parturition. Mahen-droo and colleagues (271, 272) showed that parturition wasdelayed in mice lacking steroid 5a-reductase type 1 enzyme.They showed subsequently that basal and stimulated levelsof uterine contractility were similar in these animals and inwild-type controls. However, cervical distention did not oc-cur in 5a-reductase-deficient animals, and cervical compli-ance was less on day 20 of gestation than earlier in preg-nancy. As expected, relaxin, which is known to promotecervical ripening, induced delivery in both wild-type and5a-reductase knockout animals. Subsequent studies demon-strated that while serum progesterone concentrations de-clined in knockout animals in a manner generally similar tothat of controls, the concentration of progesterone in cervicaltissue and in whole uterus remained elevated. As expected,cervical ripening and parturition occurred after ovariectomy.Thus, these studies point to the role of progesterone metab-olism in facilitating normal cervical dilatation that must ac-company uterine contractility to allow birth (273). In theuterus of pregnant mice, progesterone can be metabolized atterm through either 5a-reductase or 20a -HSD pathways. Inthe cervix, however, there is limited 20a-HSD activity, andnormally 5a-reductase provides the pathway for progester-

528 CHALLIS ET AL. Vol. 21, No. 5

Page 16: PDF (454 KB)

one metabolism, progesterone withdrawal, and cervical rip-ening and dilatation (272). Further studies of other genesassociated with cervical ripening are clearly warranted inthis fascinating model, as are measurements of 5a-reductaseactivity in human cervix from patients at term and pretermlabor.

V. Myometrial Stimulation: Phase 2 of Parturition

Activation prepares the myometrium to respond opti-mally to the production of those myometrial stimulants thatprovoke myometrial contractility during labor. Althoughmany agonists have been described to stimulate myometrialcontractions, convincing information is available only for OTand stimulatory PGs (274). The physiological role of otherputative agonists such as CRH is uncertain and equivocal.The actions of these three groups of compounds are dis-cussed below.

A. Stimulation: role of OT

OT is a nonapeptide synthesized by hypothalamic mag-nocellular neurons located in the supraoptic and paraven-tricular nuclei (275–277). Hypothalamic OT is released intothe circulation from the posterior pituitary. Its classical ef-fects include promoting myometrial contractility during latepregnancy and parturition and stimulating milk release fromthe mammary gland in lactation (275, 278, 279). The dilemmasurrounding the role of OT in the process of labor arose whenit was unclear whether levels of OT in the maternal circu-lation actually increased before the onset of labor (279, 280).The recent report that mice bearing a null mutation in the OTgene have normal pregnancies and labors may reflect a com-pensatory effect of AVP (281, 282). Studies showing the rel-ative ineffectiveness of OTR antagonists in preventing pre-term labor, however, suggest that while this hormonecontributes to labor, it may not be an essential element (283).

One aspect of the solution to the apparent discrepancybetween circulating OT levels and parturition was the dra-matic increase in myometrial sensitivity to OT before andduring labor, associated with a several-fold increase in myo-metrial OTR gene expression, which coincides with peakuterine responsiveness (276, 284–286). Thus, changes in cir-culating OT levels would not be necessary for the peptide tohave a physiological role in labor (280). A parallel conclusionis drawn from the 24-h pattern of OT secretion, and myo-metrial sensitivity (287). Recent studies also suggest that OTmay act as a local mediator of parturition. OT gene expres-sion has been demonstrated in the human and rat uterus andfetal membranes (288–290). In the rat, fetal membranes, pla-centa, and uterus synthesize OT mRNA transcripts withextended poly-A tails (289). Levels of OT mRNA in rat fetalmembranes declined from gestational day 14 to term, bututerine OT transcripts increased during gestation 150-foldand exceeded levels of OT mRNA in the hypothalamus atterm (289). Human fetal membranes, amnion, chorion, anddecidua synthesize OT mRNA, and levels of OT mRNAtranscripts increased in these tissues at the time of parturition(290). In vitro studies with rat and human chorio-decidualtissue have indicated that estrogen, generated locally, could

up-regulate OT gene expression (291–293), consistent alsowith the presence of an ERE in the OT promoter region (293).Other, fascinating studies have indicated that OT may pro-mote uterine activity by antagonizing the relaxant effect ofCRH through receptors coupled to adenylate cyclase (seebelow). The general consensus is that OT appears to have arole to play in the stimulus to uterine contractility at term andin uterine involution (294). Whether that role is indispensableremains in dispute.

B. Stimulation: role of PGs

There is a substantial body of evidence to support a rolefor PGs in the labor process, at term and preterm (207, 295).PGs contribute to the transition from phase 1 to phase 2rather than initiating the labor process. Mice carrying nullmutations for genes encoding the PGF2a receptor (296), cy-tosolic phospholipase A2, and prostaglandin synthase type1 (PGHS-1) (297) have delayed labor onset although neonatalviability is diminished. Mice lacking the PGHS-2 gene (298)have not been studied in relation to gestation length andpregnancy outcome because fertility is impaired, and ovu-lation and implantation are blocked. Lack of PGF2a (FP)receptor prevents effective luteolysis at the end of gestation,so plasma progesterone concentrations are maintained. Inthese animals OTR expression in the uterus is suppressed,presumably in response to the elevation in progesterone,since ovariectomy allowed OTR up-regulation and delivery.The extent to which information from these murine modelsis applicable to human gestation may be questioned, sincethe primary site of PG action is at the level of the corpusluteum, which is not required for pregnancy maintenance inwomen after the first 5–6 weeks of pregnancy. Perhaps thebest indicator for a role of PG in parturition in primates aswell as sheep and other species is the measurement of in-creased PG output before the appearance of labor-like myo-metrial contractions (299–301) and the effectiveness withwhich drugs that block PG synthesis suppress myometrialcontractility and prolong gestation length.

PGs are formed from membrane phospholipids throughthe initial activity of phospholipase A2 or C isozymes form-ing unesterified arachidonic acid (302–304). PLA2 isozymes,localized by immunostaining to fetal membranes and myo-metrium (305), may include the larger molecular mass (85–110 kDa) cytosolic form (cPLA2), as well as secretory typesI, II, and III, extracellular 14-kDa forms. Activation of secre-tory PLA2 (sPLA2) requires millimolar concentrations of cal-cium, whereas cPLA2 is activated at micromolar calciumconcentrations (see Ref. 8).

Cytosolic PLA2 translocates to the cell membrane in re-sponse to agonist stimulation and liberates arachidonic acidfrom the sn-2 position of phospholipid (306). Activity ofcPLA2 is reportedly greater in amnion from patients not inlabor at term or preterm than from patients in labor, ex-plained as depletion of cPLA2 at this time (304). Previousstudies had shown that cPLA2 expression was up-regulatedin WISH cells, a transformed amnion epithelial cell line, inresponse to cytokine stimulation, and that this occurs inparallel with increased expression of PGHS-2 by these cells(307, 308). The general consensus, however, is that in human

October, 2000 PARTURITION 529

Page 17: PDF (454 KB)

pregnancy, expression of PLA2 increases gradually in fetalmembranes during gestation but does not increase appre-ciably at the time of labor (309).

Arachidonic acid is further metabolized to the intermedi-ate PGH2 by PGHS enzymes, which have both cyclooxygen-ase and peroxidase activities (310, 311). There are two formsof PGHS; both are heme proteins composed of two approx-imately 70-kDa subunits. The constitutive form (PGHS-1)and the inducible form (PGHS-2) are distinct gene productsalthough they have considerable sequence homology, andtheir cDNAs are 60–65% homologous (312). PGHS-1 hassimilar properties to other housekeeping genes. PGHS-2 ischaracteristically up-regulated by growth factors and cyto-kines. The activity of PGHS-1 and PGHS-2 is inhibited by awide spectrum of nonsteroidal antiinflammatory drugs.These differ in their Ki values for the two PGHS isoforms,suggesting the potential to develop specific inhibitors of ei-ther isoform for therapeutic management (313–315).

Arachidonic acid may also be metabolized through dif-ferent lipoxygenase pathways including 5-lipoxygenase,platelet-type-12-lipoxygenase, leukocyte-type-12-lipoxygen-ase, and 15-lipoxygenase (316). Arachidonic acid metabolismthrough 5-lipoxygenase forms 5 H(P)ETE, which can be con-verted to leukotriene A4 (LTA4), which is subsequently hy-drolyzed to LTB4 or LTC4. 12-Lipoxygenase or 15-lipoxy-genase activity results in the formation of 12-H(P)ETE and15H(P)ETE. There are some suggestions that these productscan weakly stimulate contractility of smooth muscle. It hasalso been suggested that arachidonic acid metabolism inhuman fetal membranes during pregnancy is directed pref-erentially toward lipoxygenase products, but there is a pro-gressive switch toward the more potent PGHS (also cyclo-oxygenase, COX) activity at term (317). Primary PGs areformed from PGH2 through the activity of specific isomer-ases and synthases. There is unfortunately very little infor-mation concerning the expression, localization, and changein activity of these enzymes in intrauterine tissues at term orpreterm labor, and this will be an obvious area of furtherinvestigation.

The major pathway in the metabolism of PGE2 and PGF2a

involves the action of a type 1 NAD1- dependent PGDH thatcatalyzes oxidation of 15-hydroxy groups resulting in for-mation of 15-keto and 13,14 dihydro-15-keto metaboliteswith reduced biological activity (318, 319). We have reportedthat PGDH expression and activity are decreased in chorio-decidual tissue of women at spontaneous and preterm labor(see below), raising the possibility that failure to inactivatePGs produced within intrauterine tissues during pregnancymay be one cause of preterm labor (320).

The action of PGs is exerted through specific receptorsincluding the four main subtypes for PGE2, EP1, EP2, EP3,and EP4, and FP for PGF2a (60, 321). EP1 and EP3 receptorsmediate contractions of smooth muscle through intracellularsignaling pathways that elevate free calcium and decreaseintracellular cAMP (27). EP2 and EP4 receptors are coupledthrough adenylate cyclase and increase cAMP formation,leading to relaxation of smooth muscle. Consistent with this,various groups have reported that EP2 expression in myo-metrium is higher preterm than at term. In the rat, parturitionis associated with down-regulation of EP receptor subtypes

and with up-regulation of myometrial FP receptors, effectinga switch from inhibition to stimulation.

1. PG synthesis. Regulation of PGHS-2 and PGHS-1 genes areclearly multifactorial (322–324). There are two nuclear factor(NF)-kB binding elements within the proximal promoter re-gion of PGHS-2 (325, 326). p50 And p65, key members of theNF-kB Rel family of proteins are present in trophoblasts andlikely serve as mediators of cytokine-induced up-regulationof PGHS-2 expression (327). The PGHS-2 promoter also in-cludes response elements resembling NF-IL6, GRE, CRE, andAP2 sites (323, 325). Levels of PGHS-2 are increased up to80-fold in response to various cytokines and growth factors,whereas levels of PGHS-1 are usually increased only 2- to3-fold in response to these stimulators (328, 329). Studies inseveral species, including the human, have indicated that thePGHS-2 isoform is the principal form of the enzyme involvedin the increased PG production seen at the time of parturi-tion. Effects of CRH in up-regulating PG output, at leastwithin fetal membranes (see below), is likely mediatedthrough proximal CRE sequences (326). Although glucocor-ticoids inhibit PGHS-2 expression in WISH cells and in mostother cell types, apparently by interference with the NF-kBsignaling system (330), they stimulate PGHS expression andactivity in trophoblast-derived cells including amnion, andchorionic trophoblast (58, 331–334). Kniss (327) reported asimilar effect of dexamethasone in stimulating PGHS-2mRNA expression in human breast adenocarcinoma cells.The stimulatory effect of glucocorticoids on PGHS gene ex-pression in human fetal membranes is central to our currenthypotheses of human parturition and will be discussed inmore detail below.

In human pregnancy, the PG synthesizing and metabo-lizing enzymes are compartmentalized discretely betweenthe amnion and chorion, decidua, and myometrium (Fig. 5;Refs. 335 and 336). PGHS activity predominates in amnion,PGE2 is the principal PG formed (337), and there is an in-crease in PG synthesis and levels of PGHS-2, but not PGHS-1mRNA at preterm and term labor (338–343). Immunohisto-chemical and in situ hybridization studies have localized thePGHS-2 enzyme and mRNA to the amnion epithelium (344–346), the subepithelial cells in the mesenchyme and in thechorion laeve trophoblasts with lower expression found indecidua (347–349). Decidua has been reported to produceincreased amounts of PGs at the time of labor, but this is nota consistent observation (348). Human decidua is made upof decidualized stromal cells, bone marrow-derived macro-phages, and other cell types including trophoblasts that in-terface with chorion (350). Variability in cell populationsused for in vitro studies may contribute to the variability ofresponses that have been obtained. In chorion, interposedbetween amnion and decidua, PGDH activity predominates,although PGHS is also expressed (347, 351). Output of PGsand PGHS activity is greater in chorion from patients atspontaneous labor than at elective term cesarean section; inpreterm labor chorion both PGHS-1 and PGHS-2 mRNAlevels are increased (352, 353).

It is generally considered that activity of PGDH predom-inates in chorion (354), forming a relative metabolic barrierthat prevents passage of PGs generated within amnion or

530 CHALLIS ET AL. Vol. 21, No. 5

Page 18: PDF (454 KB)

chorion from reaching underlying decidua or myometriumthrough most of pregnancy (320, 355). The presence of highPGDH activity in chorion trophoblasts (356) implies that atfull term those PGs acting on the myometrium would morelikely be derived from decidua, or from the myometriumitself (41, 357). There are variable reports, however, ofchanges in PGHS activity in human myometrium at the timeof labor (59). Some workers have reported increased PGHS-2expression and activity, while others have reported no

change, or even decreased activity. In myometrium throughpregnancy, PGHS-1 or PGHS-2 must be present to generatethe predominant PGI2 which, as discussed above, contributesto maintenance of uterine quiescence (59). It has been sug-gested that PGI2 formation in myometrium may be decreasedby glucocorticoids. Unfortunately, it is difficult, experimen-tally, to obtain consistent specimens of human myometriumfor biochemical analysis. Generally, tissue is obtained fromlower segment uterus, but at term with ensuing cervical

FIG. 5. Diagrammatic representation of sites of PG synthesis and metabolism at term labor (panel A) and preterm labor (panel B). PGHS-2,prostaglandin H synthase 2; PGDH, 15-OH prostaglandin dehydrogenase.

October, 2000 PARTURITION 531

Page 19: PDF (454 KB)

dilatation, the proportion of myocytes in the tissue is likelyto have changed. Further, recent studies indicating that thereare regional differences in CAP genes between the fundusand lower segment of the human uterus in late pregnancy(see above) may suggest the need for further reexaminationof these issues, ideally combined with experimental manip-ulation in subhuman primates.

It remains crucial to understand regulation of PGHS-1 andPGHS-2 expression in human fetal membranes and to de-lineate the major site of PG production at term and pretermlabor (Fig. 5). These may not necessarily be the same. Forexample, instances of preterm labor may be associated withelevated PG production in amnion or chorion, whereas termlabor may require increased PGHS-2 expression in deciduaand myometrium (344). Given that PGs act generally as para-crine or autocrine regulators, it will be exceedingly difficultto obtain in vivo evidence for altered PG production specif-ically at these sites. Amniotic fluid concentrations of PGsincrease at labor, and the initial changes precede the onset ofmyometrial contractility. Levels of PGF2a in amniotic fluidpresumably reflect, in part, production from decidua, sincePGE2 and not PGF2a is the major eicosanoid formed fromamnion and chorion (Figs. 5 and 6). However, these mea-surements probably provide no more than a crude estimateof the pattern of PG change at a local cellular level and giveno information concerning receptor subtypes and distribu-tion (358).

Primary cultures of mixed and purified cells from humanamnion or chorion have been used extensively as models to

study the regulation of PG formation in response to cyto-kines, growth factors, CRH, and lipopolysaccharides. In ad-dition, the amnion-derived epithelial cell line (WISH cells)has also been used extensively (359–361). A crucial reserva-tion with all of these in vitro studies is that, in general, singlecompounds have been studied in isolation of the in vivoenvironment; the extent to which results can be extrapolatedfrom in vitro to in vivo will remain, unfortunately, a matterof conjecture.

Many cytokines have been shown to act on amnion, cho-rion leave, and decidua to increase PG output (360, 362–364).IL-1b stimulates PG output by cultured amnion, chorionleave, and decidua (195, 317, 365) while IL-6 stimulates PGoutput by decidua and amnion (366, 367). IL-8 did not alterPG production by chorion or decidua, but augmented thestimulatory action of other cytokines (368). The effect of IL-1bis certainly associated with increased expression of PLA-2and PGHS-2 (329). The action of IL-1b can be reduced by thenaturally occurring receptor antagonist, which has beenshown to prevent IL-1b-induced labor in mice (369). IL-1bstimulation of PGHS in amnion and chorion may be medi-ated through the NF-kB system (370–372). In WISH cellsstimulated with interleukin-1b, I-kBa was degraded by morethan 90% within 15 min of stimulation, and this was asso-ciated temporally with nuclear translocation and binding ofNF-kB (373). PGHS-2 mRNA was increased within 30 minand reached steady state by 4 h. PGHS-2 protein then in-creased more than 80-fold, and this was associated with acorresponding time-dependent increase in PG production.

FIG. 6. Summary to indicate factors leading to up-regulation (1) or down-regulation (2) of prostaglandin H2 synthase in intrauterine tissues.The role of progesterone remains equivocal.

532 CHALLIS ET AL. Vol. 21, No. 5

Page 20: PDF (454 KB)

Inhibition of I-kBa degradation by calpain-I inhibitionblocked NF-kB translocation, and increases in PGHS-2mRNA and protein, and PG synthesis (373). Wang and Tai(374) provided similar information and showed that in WISHcells, dexamethasone blocked IL-1b-mediated stimulation ofPGE2 output consistent with the general model of mutualtranscriptional antagonism from GR/NF-kB interaction(330).

Human amnion cells can be maintained as mixed popu-lations in culture or can be separated into primary epithelialcells and cells of the subepithelial mesenchymal layer (375).We have reported recently that the output of PG by mesen-chymal cells exceeds that of epithelial cells in the basal state.Epithelial cell production of PGs was stimulated by glu-cocorticoids, whereas there was no significant change in thealready elevated output of PGs from mesenchymal cells(375). Previously, in mixed cultures, glucocorticoids andIL-1b were shown to increase PGHS-2 mRNA, protein, andPGE2 output predominantly from the subepithelial mesen-chymal cells (331, 376). It remains possible that this apparentdifference can be explained by epithelial-mesenchymal cellinteraction, and current studies are directed at resolving thisissue.

The effect of glucocorticoids on primary cultures of am-nion cells and on chorion trophoblast cells is surprising (377)and striking (323, 378, 379). Although dexamethasone inhib-ited PGE2 output by freshly dispersed amnion cells, it stim-ulated PGE2 output by amnion cells after 4–5 days in culture(376). The effect was dose dependent and associated withincreased expression of PGHS-2 mRNA and protein. Theactivity of glucocorticoids is also receptor mediated and canbe inhibited by addition of GR antagonist (380). In previousstudies, we had localized GR to amnion epithelial cells, sub-epithelial fibroblasts, and chorion laeve trophoblasts in hu-man pregnancy (381). GR exists as both a-form and b-form(330). GRa is retained in the cytoplasm in an inactive state byits association with the regulatory heat shock proteins suchas HSP-56 and HSP-90. GRb, formed from alternate splicingof the same mRNA transcript as GRa, is localized in the cellnucleus independent of binding to ligand. It appears thatGRb functions as a dominant negative regulator of GRatransactivation. Thus, earlier studies of GR localization to celltypes within human fetal membranes require repeating withspecific identification of GRa and GRb forms.

Peptides such as CRH could be released from amnionepithelial cells to act in a local paracrine manner and up-regulate PGHS-2 expression in mesenchymal cells (see Ref.207). Full thickness fetal membranes treated in culture withCRH were stimulated to increase output of PGE2 and in-creased levels of PGHS-2 mRNA within 4 h in culture. Thus,the stimulatory effect of glucocorticoids on PG production byamnion, known to involve an intermediary protein syntheticstep, could be the result of synergistic epithelial-mesenchy-mal interaction, in addition to, or instead of, any direct effecton amnion cell types. Similar interactions may contribute tothe response to cytokines such as IL-1b in vitro (382). Inter-estingly, recent studies have shown that in amnion explants,in contrast to chorion and decidua, the antiinflammatorycytokine IL-10 stimulates rather than inhibits PG production,and the normally antiinflammatory cytokine IL-4 stimulates

PGE2 output in amnion cultures (329). The authors havesuggested that amnion may therefore be refractory to inhib-itory cytokines as part of an evolutionary mechanism de-signed to expedite the parturition processes.

Over the past 10 yr, in vitro studies have generated animpressive list of substances capable of increasing PG outputby human fetal membranes in culture (383–386). Clearly,availability of free calcium is a critical requirement. Epider-mal growth factor (EGF), platelet activating factor (PAF), andagents that activate protein kinase C stimulate PG output(387, 388). Importantly, b-sympathomimetic drugs andagents that increase intracellular cAMP levels also increasedPG output by cultured chorion and decidual cells (389). Cat-echolamines are present in increasing concentrations in hu-man amniotic fluid in late gestation (390), and both amnionand decidua express components of the adenylate cyclasesystem, which undergoes stimulation with b-agonists suchas isoproteronol (391). Effects of these activators of adenylatecyclase can be mimicked by (Bu)2cAMP or phosphodiester-ase inhibitors such as methylxanthine (389). Studies such asthese may help explain the disappointing lack of efficacy ofb2-sympathomimetic drugs in sustaining uterine quiescencewhen used in the treatment of preterm labor (392). Althoughthese compounds are effective in the short term by elevatingcAMP and decreasing activity of MLCK, in the longer termelevations of cAMP may up-regulate PGHS-2 through aproximal CRE, resulting in increased output of stimulatoryPGs, uterotonins whose action the administration of b2-mimetic was intended to antagonize.

2. PG metabolism. The major metabolizing enzyme for PGs(393), PGDH, is exquisitely localized in fetal membranes totrophoblast cells of chorion (Fig. 5). Thus, it could act as ametabolic barrier to the passage of unmetabolized PGs, gen-erated in amnion or chorion, and prevent their reaching theunderlying decidua or myometrium in a biologically activeform (354, 394, 395). Some years ago, we identified a groupof patients presenting in idiopathic preterm labor with de-ficiency of PGDH in chorion trophoblast cells (396). Therewas a further reduction of ir-PGDH, PGDH mRNA, andPGDH activity in chorion trophoblast cells, but not placentaltrophoblast, in patients in preterm labor with an underlyinginfective process (397). Thus, with preterm labor in the pres-ence of an inflammatory response, loss of chorion tropho-blast cells leads to loss of PGDH activity. PGs generated, forexample in response to elevations of cytokines, will not bemetabolized and will be available to stimulate underlyingmyometrium.

In idiopathic preterm delivery, in the absence of infection,it is clear that PGDH activity is specifically regulated inchorion trophoblast (Fig. 7). During in vitro studies withchorion trophoblast cells maintained in culture, we foundthat the glucocorticoids, cortisol and dexamethasone, inhib-ited PGDH activity and decreased levels of PGDH mRNA(398). Cortisone was as effective as cortisol, since choriontrophoblasts contain 11b-HSD Type 1 (11b-HSD-1) capableof reducing cortisone to biologically active cortisol (399). Thisactivity could be inhibited by carbenoxolone, an active in-gredient of licorice. Chorion trophoblast cells also expressed3b-HSD and converted pregnenolone to progesterone (400,

October, 2000 PARTURITION 533

Page 21: PDF (454 KB)

401). Inhibition of 3b-HSD activity with trilostane led todecreased PGDH activity and reduced levels of PGDHmRNA in the cells. These could be restored by concurrentaddition of progesterone, or of the synthetic progestagens,MPA or R5020 (398). Effects of these compounds, in turn,were antagonized by onapristone and RU486, inhibitors ofprogesterone action (398, 402). Furthermore, inhibition ofPGDH mRNA and activity by cortisol could be reversed byaddition of progesterone (320).

These data could be explained by glucocorticoids and pro-gesterone acting through independent receptors, or by theirinteraction at the same binding sites on GRa (403). Previ-ously, Karalis and Majzoub (404) provided evidence thatsimilar interaction between progesterone and cortisol forbinding to GR explains the interactive effect of these com-pounds on the output of CRH by placenta trophoblast cells.

In recent studies we found that CRH also decreased PGDHactivity in chorion trophoblast cells in a dose-dependentfashion (F. Patel and J. R. G. Challis, unpublished observa-tions). We believe this activity is mediated through cAMPgeneration, since CRH binds to CRH-R1 species in fetal mem-branes where it may increase cAMP, and cAMP decreasesPGDH activity (405), presumably acting through a consensusCRE in its promoter region. Thus a pattern is emerging thatseveral agents which up-regulate PGHS-2 in human fetalmembranes (CRH, cortisol, IL-1b, TNF) down-regulatePGDH in chorion (Fig. 8). Effects of cortisol in the membranesmay be enhanced by local conversion of cortisone to cortisol,through the reductase activity of chorionic 11b-HSD-1 (406).The activity of this enzyme is increased by PGE2 and PGF2a

in a dose-dependent fashion that is associated with, anddependent upon, a transient increase in intracellular Ca21 (N.Alfaidy and J. R. G. Challis, unpublished results). Therefore,a further feed-forward paracrine/autocrine loop exists inwhich increased output of PG should stimulate 11b-HSD-1,resulting in increased production of cortisol, which leads tofurther increases in PGHS-2 and decreases in PGDH (Fig. 8).

We have referred previously to the finding of regionalvariation in PGDH activity. We suggest that this might reflectprogesterone stimulation of the enzyme (407, 408) in a re-gional pattern. Chorion collected from patients at electivecesarean section at term in the absence of labor had higherPGDH activity in the region of the membranes overlying theinternal os than chorion collected from a region adjacent tothe placenta or between the placenta and cervix (41). How-ever, at cesarean section in labor, there was a dramatic re-duction in PGDH activity in chorion from the lower uterinesegment. We suggested that this altered response could re-flect an antagonism of the effect of progesterone on the en-zyme by elevations of cytokines derived from vaginaland/or cervical fluids. We and others have shown thatwhereas IL-1b and TNFa increase PG synthesis, these cyto-kines decrease PGDH activity and PGDH gene expression(409, 410). Importantly, IL-10, the antiinflammatory cytokinethat attenuates IL-1b-induced up-regulation of PGHS, alsoreverses IL-1b down-regulation of PGDH (409). The impor-tance of this observation is that PGs generated within amnionand chorion in the lower segment may escape metabolism inchorion specifically in that region at the time of labor to reachthe cervix and effect effacement and dilatation.

FIG. 7. Diagrammatic summary of factors regulating expression of the acitivity of PGDH in human chorion.

534 CHALLIS ET AL. Vol. 21, No. 5

Page 22: PDF (454 KB)

3. PGs and infection. Approximately 30–40% of preterm la-bors are associated with an underlying infective process.Romero, Mitchell, and collaborators (411–413) have demon-strated elegantly the role for infection in preterm labor. Bac-terial organisms themselves secrete phospholipases, result-ing in increased release of arachidonic acid from intrauterinetissues and increased PG production. Alternatively, bacterialendotoxin, such as lipopolysaccharide, acts on amniotic ormembrane macrophages, causing either PG release or furtherrelease of cytokines (414–417). Cytokines in turn elevate PGproduction within amnion, chorion, and decidua as dis-cussed previously (418). Administration of cytokines or bac-terial endotoxins to pregnant mice provokes premature de-livery and allows examination of the precise temporalsequence of events in infection-driven preterm labor (419,420). A number of cytokines including IL-1b, TNFa, IL-6, andIL-8 (neutrophil-activating protein-1) are increased in amni-otic fluid of patients undergoing preterm labor associatedwith infection (421–424). Cytokines are produced not only bymacrophages, but are synthesized and secreted by humanfetal membranes in decidua, and these tissues may be thesources of the cytokines found in amniotic fluid. IL-1b, IL-6,and IL-8 mRNA were expressed in amnion, chorion leave,and decidua, particularly in tissues obtained after labor. Inaddition, cultured decidual and chorion cells produce IL-6and IL-8 when stimulated with IL-1b and TNFa, and amnionproduces IL-8 in response to IL-1b (425). Thus, these studieshave led to the suggestion that there is a complex cytokinenetwork at the chorio-decidual interface, as has been pro-posed to exist in other tissues (269). It is also possible thatcytokines cause release of other uterotonins, including OTand CRH in decidua (426, 427), myometrium, and/or pla-centa. These compounds may affect the myometrium di-rectly or indirectly. Lipopolysaccharide also inhibits repli-cation of amnion cells, and it has been suggested that thismight be a mechanism by which lipopolysaccharide contrib-utes to premature ruptured membranes.

The paradigm of infection-driven preterm labor has been

proposed as a means of understanding regulation of PGproduction in labor at term (348). However, preterm labor inthe absence of infection can occur without demonstrablechanges in amniotic fluid PGE concentrations and appar-ently without enhanced PG biosynthetic activity in fetalmembranes. It has been argued that changes in PG andcytokine concentrations in the amniotic fluid of women inpreterm labor with infection are not reproducible, and thatthese compounds accumulate there as a result of pretermlabor, rather than as a cause (428). It has also been argued thatinvasion of the amniotic sac by microorganisms occurs whenlabor has been initiated, when tissues of the forebag areexposed. Furthermore, since parturition is an inflammatoryprocess, the presence of mediators of inflammation in am-niotic fluid could be a natural event of parturition withoutarguing for a role of infection as a cause of preterm labor. Thebody of evidence currently available has tended to counterthis latter view. However, as in all human studies of this type,it is extremely difficult to delineate precisely the cause-and-effect sequence of relationships. Furthermore, a low-gradeinflammatory response, where accumulation of cytokinesoccurs without an infective process, may be present normallyat term and contribute to the stimulus of labor or remain asa parallel, but unrelated, event.

C. Stimulation: role of CRH

Over the past 10 yr there has been considerable interest inthe possible role that CRH, produced from intrauterine tis-sues, plays in the regulation of human pregnancy and par-turition (429, 430). Pro-CRH mRNA is present in placentaltissue (431) and decidua in increasing amounts during preg-nancy. These levels correlate with increased concentrationsof ir-CRH peptides in the placenta and with the exponentialincrease in CRH1–41 concentrations in maternal peripheralplasma (432–435). CRH also increases in cord plasma, al-though the concentrations are generally lower than those inthe maternal compartment (432, 436, 437). Several groups of

FIG. 8. Interrelationship between cortisol, PGHS-2, PGDH, and CRH. In chorion, cortisone can be converted to cortisol through the activityof 11b HSD-1, and the activity of this enzyme is increased locally by PGs.

October, 2000 PARTURITION 535

Page 23: PDF (454 KB)

investigators have reported that maternal plasma CRH con-centrations are elevated significantly in the plasma of pa-tients presenting in preterm labor (433, 438–440) and may beused to discriminate patients presenting in preterm laborwho will deliver within 24–48 h from those patients with asimilar diagnosis, but in whom labor is not imminent (441).

The biological activity of CRH in maternal plasma is at-tenuated by the presence of a circulating CRH binding pro-tein (CRH-BP), produced in the liver and placenta (429, 442).CRH-BP blocks the ability of circulating CRH to promoteACTH release from pituitary corticotrophs, and it inhibits thestimulatory effect of CRH on uterine PG production. Con-centrations of CRH-BP decrease during the last 5–6 weeks ofnormal pregnancy and before preterm labor, coincident withthe increase in maternal CRH concentrations (443), and ap-parently in response to increased CRH secretion. In the pla-centa, CRH is produced by syncytiotrophoblast and inter-mediate trophoblasts (444), and immunoreactive CRHlocalizes to these cell layers (429, 445). In culture, CRH outputfrom placental and chorion trophoblast cells is inhibited bynitric oxide and progesterone and increased by cat-echolamines, OT, cytokines, and glucocorticoids (Fig. 9; Refs.427 and 444). Majzoub and colleagues (446, 447) demon-strated that dexamethasone increases levels of CRH mRNAin placental trophoblast cells maintained in culture in a time-and dose-dependent fashion, although later suggested thatthis “apparent” stimulation resulted in fact from reversal ofprogesterone-induced inhibition of CRH expression (263,404). Glucocorticoids compete with and displace progester-one from GRa binding, and diminished inhibition is mea-sured as an apparent increase in secretion of CRH.

We demonstrated in vivo that patients receiving prenatalglucocorticoids to promote pulmonary maturation inamounts that decreased maternal ACTH and cortisol con-centrations by more than 80% provoked stimulation of ma-ternal CRH concentration by almost 50% over pretreatmentvalues (448). Administration of glucocorticoids to pregnantwomen with singleton or multiple fetuses at risk of pretermlabor actually stimulates uterine contractility, although theeffect may be transient (449, 450). From the foregoing dis-cussion it is evident that this could be the result of up-regulation of PGHS, down-regulation of PGDH, and/orstimulation of placental CRH which, in turn, provokes afurther increase in PGHS-2 expression (451). Administrationof glucocorticoids eventually suppresses fetal HPA function,decreases estrogen output from the placenta, and might beexpected to diminish uterotrophic activation of the myome-trium, perhaps accounting in part for the (fortunately) tran-sient nature of this response.

Based upon these results, and the demonstration of acti-vation of fetal HPA function in response to hypoxemia inanimal fetuses, we proposed that the human fetus would alsorespond to an adverse intrauterine environment such asacute hypoxemia with activation of the fetal HPA axis (10).With time, increased pituitary drive to the adrenal increasessteroidogenic enzyme potential and cortisol output. Fetalcortisol, then acting through placental and/or membraneGRa, up-regulates placental CRH gene expression, leading tothe increased CRH concentrations in the plasma of patientspresenting in preterm labor. Accordingly, cord CRH con-centrations are elevated in the presence of intrauterinegrowth restriction (IUGR), or decreased values of cord PO2

FIG. 9. Summary of regulation of expression and output of CRH in human intrauterine tissues and placenta.

536 CHALLIS ET AL. Vol. 21, No. 5

Page 24: PDF (454 KB)

(440, 452). CRH is a vasodilator in the placental vascular bedand reverses the vasoconstrictor influence of PGF2a (453). Inthe placenta, the vasodilator action of CRH is associated withup-regulation of the NO-cyclic GMP pathway. Hence, ele-vations of CRH within the placenta should signal increasedblood flow and correction of a hypoxemic insult to the fetus.However, if the hypoxemia persists, placental CRH outputpresumably remains elevated (Fig. 10). CRH, secreted intothe fetal circulation, drives further pituitary ACTH secretionand also drives DHAS output from the fetal zone of the fetaladrenal gland (243); hence, maternal estrogen output shouldrise as a secondary response to fetal distress. Increased es-trogen leads to uterine activation. CRH contributes to in-creased expression of PGHS (451) by up-regulating adenyl-ate cyclase activity in placental and membrane cells (61). Itwill be recalled that the PGHS promoter contains a CRE.Thus, we speculate that activation of a feed-forward loop inresponse to a hostile intrauterine environment is a mecha-nism by which a compromised fetus may signal pretermlabor and induce premature delivery (Fig. 10). In addition,maternal stress with elevations of maternal glucocorticoidconcentrations may also contribute to elevations of placentalCRH output and preterm birth. Hobel and colleagues (454)reported increases in maternal CRH concentrations inwomen with elevated scores for perceived stress and anxiety.These values predicted preterm labor, even as early as 20–24weeks of gestation.

It is extremely difficult to prove or disprove this hypoth-esis with in vivo studies in normal human pregnancy. Studiescannot be performed in nonprimates, since these species donot appear to produce placental CRH. The pattern of pla-cental CRH output during pregnancy in the baboon andrhesus monkey has been described but differs from the ex-ponential increase of plasma CRH concentration observed inhuman gestation (429). Women receiving betamethasone de-liver at variable times after treatment. Current obstetric prac-tice in North America, in fact, makes it difficult to obtain“control” placental tissue from patients in preterm labor who

have not received exogenous corticosteroid; such patientsmay have increased endogenous corticosteroids before tissuecollection in any case.

A further reservation is related to CRH receptor specificity.CRH exerts its effects through activating specific G protein-coupled receptors, which exist in two subtypes: CRH-R1 andCRH-R2. These arise from different genes with multiplesplice variants (455). The two receptors share approximately70% homology at the amino acid level. CRH-R1 exists in atleast three variant forms (R1a, R1b, and R1C). Recently, anadditional form, CRH-R1D, has been isolated, which is iden-tical to CRH-R1a except that it contains an exon deletionresulting in loss of 14 amino acids in the seventh transmem-brane domain (456). CRH-R2 exists in at least three splicevariant forms (R2a, R2b, and R2g). CRH-R1 predominates inhuman myometrium (455, 457). CRH-R2 is expressed in fetalmembranes, but at lower levels than CRH-R1. Parentheti-cally, this pattern is reversed in rats in which CRH-R2 pre-dominates in myometrium (Y. Stevens and J. R. G. Challis,unpublished observations). CRH-R2 has higher specificityfor urocortin than CRH, raising the possibility that in rodentgestation, placental output of urocortin rather than CRH,may determine activity of this pathway.

Because CRH-R1 is linked to the adenylate cyclase systemthrough GSa regulatory proteins, it is not surprising that CRHstimulates cAMP output by human myometrial cells main-tained in vitro (61). Herein lies the paradox. CRH-inducedincreases in cAMP should inhibit myometrial activity,through mechanisms described above, yet elevations in ma-ternal peripheral plasma CRH concentration are suggested topredict women at risk of increased uterine activity and pre-term labor (61). This may be resolved if CRH action onmyometrium is independent of effects on PG synthesis inother tissues (458). Affinity of CRH binding in myometriumincreases with pregnancy, and then decreases in late gesta-tion (459). Hence, we (8) and others (61) have speculated thatduring gestation CRH acts as a myometrial relaxant, ratherthan as a uterotonin. At term, OT up-regulates protein kinase

FIG. 10. Diagram to indicate interrela-tionships between mother, placenta,and fetus concerned with up-regulationof placental CRH output in human ges-tation in response to stress. It is pro-posed that cortisol from either maternalor fetal adrenal can up-regulate placen-tal CRH expression. Placental CRH, inturn, affects fetal adrenal function in-directly through stimulation of fetal pi-tuitary ACTH release, and directly bystimulating secretion of DHAS from thefetal zone of the fetal adrenal gland.

October, 2000 PARTURITION 537

Page 25: PDF (454 KB)

C, which phosphorylates CRH receptor protein resulting inits desensitization and loss of inhibitory influence (61, 460).Stevens et al. (265) showed that levels of mRNA for CRH-R1heptohelical glycoprotein increase in lower segment myo-metrium from patients in labor whether at term or preterm.Hence, CRH may contribute to regionalization of uterineactivity responses at this time, producing inhibition of ac-tivity, or relaxation in the lower segment, but stimulation ofactivity through up-regulation of PG synthesis in the fundalregion of the uterus.

In our view, the putative role of CRH in pregnancy main-tenance and parturition remains unclear. The concept of pla-cental CRH as “a placental clock controlling the length ofhuman pregnancy” implied a stimulatory effect on the myo-metrium (461, 462), which is difficult to reconcile with theknown biochemical effects of CRH (61). Certainly, CRH aug-ments OT- and PGF2a-induced contractility of myometrialstrips in vitro (289, 463). However, it decreases output of PGI2by myometrial cells and has no direct stimulatory action onits own. Perhaps increased levels of CRH are required tosustain relaxation, rather than stimulation, of the uterusthrough late gestation. However, lowered concentrations ofCRH in maternal plasma are associated with postterm de-livery in which, presumably, relative myometrial quiescencehas been maintained. Resolution of this interesting dilemmain which a single ligand may have different actions depend-ing upon differential expression of its receptor subtypes andcoupling through second messenger systems is required asa scientific basis to understanding CRH action in pregnancy(61).

VI. Application to Clinical Preterm Labor

Rates of preterm labor in North America have remainedrelatively unchanged over the last 30–40 yr, despite sub-stantial advances in our understanding of this process (1–3).It is apparent, however, that new knowledge has not yet beenextrapolated to clinical diagnosis and management (464,465), and that there may be reluctance to develop new drugsfor administration to women in pregnancy without guaran-tees of safety for mother and fetus. There is a clear need torecognize first those preterm labors in which prevention isundesirable because it constitutes a greater compromise tofetal health. There is a need to develop diagnostic indicators,likely specific for particular windows of gestation, to deter-mine the patient in whom the diagnosis of preterm labor iscorrect. Ideally, only these patients should be subjected totocolytics and to prenatal glucocorticoids. There is a need todevelop effective methods of tocolysis ideally related on apatient-specific basis to the cause of preterm labor in thatindividual. Hence, diagnosis of preterm labor should en-compass a multiple-test approach. The new generation ofspecific PGHS-2 inhibitors offers great promise, since in-creased expression of PGHS-2 appears to represent a com-mon final pathway of birth and preterm labor mechanismsamong species (466). The ability to regulate CRH or PGeffects through specific and appropriate agonists and/or an-tagonists is a potential alternative approach.

Both these approaches, however, act on agents of phase 2

parturition, in which uterine activation has already takenplace. Inhibition of uterotonin action or secretion does notnecessarily affect myometrial activation, although recentstudies in sheep treated with nimesulide, a PGHS-2 inhibitor,have shown reversal of some CAP gene expression. Ideally,a future strategy for preterm labor diagnosis and manage-ment should address uterine activation. Those studies willrequire careful animal studies before the introduction of newdrugs into clinical practice. A satisfactory outcome may be todelay rather than actually to prevent preterm birth, provid-ing that there is improvement in mortality and morbidity ofthe newborn.

We remain concerned about the capricious use of glu-cocorticoids in preterm labor patients (467). There is no ques-tion of the beneficial effect of these compounds in promotingpulmonary maturation in infants of women who give birthprematurely within an appropriate time for treatment. How-ever, a central thesis of this review is that glucocorticoidsprovide a stimulus to the labor process and that evidence isaccumulating to suggest that the model derived from animalexperiments may have substantial applicability to the hu-man. We recognize from animal studies that repeated ad-ministration of glucocorticoids to pregnant animals pro-duces, in a dose-dependent fashion, inhibition of fetalgrowth (468). Prenatal corticosteroids alter postnatal HPAfunction and the setting of negative feedback. Prenatal cor-ticosteroids, in animals, may result in the development ofhypertension postnatally, and in a pattern of pancreatic re-sponse to a glucose load that resembles insulin resistance(469). Prenatal and postnatal administration of corticoste-roids affect levels of type 1 and type 2 GRs in critical brainregions, particularly the hippocampus, associated withmemory and, in later life, with memory loss and neurode-generative disease. Future research into the control of pre-term labor, and to the tocolytic management of the patient atrisk of preterm labor, will need to define the relative risks andbenefits of different management paradigms that may beproposed (469).

Acknowledgments

We are indebted to Jenny Katsoulakos, Linda Vranic, and Fal Patel fortheir help in the preparation of this manuscript and to Maggie Haworthfor her patience with us.

References

1. Creasy RK 1991 Preventing preterm birth. N Engl J Med 325:727–729

2. Meis PJ, Goldenberg RL, Mercer JE, Iams JD, Moawad AH, Mi-odovnik M, Menard MK, Caritis SN, Thurnau GR, Bottoms SF,Das A, Roberts JM, McNellis D 1998 The preterm predictionstudy: risk factors for indicated preterm births. Maternal-FetalMedicine Units Network of the National Institute of Child Healthand Human Development. Am J Obstet Gynecol 178:562–567

3. Hannah ME, Amankwah KS, Barrett JFR, Bonin B, Burrows R,Cheng MM, et al 1995 The Canadian consensus on the use oftocolytics for preterm labour. J Soc Obstet Gynecol Can 17:1089–1115

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

5. Harding R, Poore ER, Bailey A, Thorburn GD, Jansen CAM,

538 CHALLIS ET AL. Vol. 21, No. 5

Page 26: PDF (454 KB)

Nathanielsz PW 1982 Electromyographic activity on the nonpreg-nant and pregnant sheep uterus. Am J Obstet Gynecol 142:448–457

6. Lye SJ, Freitag CL 1990 Local and systemic control of myometrialcontractile activity during labour in the sheep. J Reprod Fertil90:483–492

7. Lye SJ 1994 The initiation and inhibition of labour: towards amolecular understanding. Semin Reprod Endocrinol 12:284–294

8. Lye SJ, Ou C-W, Teoh T-G, Erb G, Stevens Y, Casper R, Patel FA,Challis JRG 1998 The molecular basis of labour and tocolysis. FetalMatern Med Rev 10:121–136

9. Nathanielsz PW, Binienda Z, Wimsatt J, Figueroa JP, MassamanA 1988 Patterns of myometrial activity and their regulation in thepregnant monkey. In: McNellis D, Challis JRG, MacDonald PC,Nathanielsz PW, Roberts JM (eds) The Onset of Labour: Cellularand Integrative Mechanisms. Perinatology Press, Ithaca, NY, pp359–373

10. Challis JRG 1998 Characteristics of parturition. In: Creasy RK,Resnik R (eds) Maternal-Fetal Medicine: Principles and Practice.W.B. Saunders Co., Philadelphia, pp 484–497

11. Norwitz ER, Robinson JN, Challis JRG 1999 The control of labor.N Engl J Med 341:660–666

12. Challis JRG, Lye SJ 1994 Parturition. In: Knobil E, Neil JD (eds) ThePhysiology of Reproduction. Raven Press, New York, pp 985–1031

13. Schoenber CF 1977 The contractile mechanism and ultrastructureof the myometrium. In: Wynn RM (ed) Biology of the Uterus.Plenum, New York, pp 497–554

14. Garfield RE 1988 Structural and functional studies of the controlof myometrial contractility and labour. In: McNellis D, Challis JRG,MacDonald PC, Nathanielsz PW, Roberts JM (eds) The Onset ofLabour: Cellular and Integrative Mechanisms. Perinatology Press,Ithaca, NY, pp 55–80

15. Izumi H, Ichihara J, Uchiumi Y, Shirakawa K 1990 Gestationalchanges in mechanical properties of skinned muscle tissues ofhuman myometrium. Am J Obstet Gynecol 163:638–647

16. Garfield RE 1990 Intercellular coupling and modulation of uterinecontractility. In: Garfield RE (ed) Uterine Contractility. SeronoSymposia USA, Norwell MA, pp 21–40

17. Hsu CJ, Sanborn BM 1986 Relaxin treatment alters the kineticproperties of myosin light chain kinase activity in rat myometrialcells in culture. Endocrinology 118:499–505

18. MacKenzie LW, Word RA, Casey ML, Stull JT 1990 Myosin lightchain phosphorylation in human myometrial smooth muscle cells.Am J Physiol 258:C92–C98

19. Ohya Y, Sperelakis N 1989 Fast Na1 and slow Ca21 channels insingle uterine smooth muscle cells from pregnant rats. Am J Physiol257:C408–C412

20. Pato MD, Lye SJ, Kerc E 1991 Purification and characterization ofpregnant sheep myometrium myosin light chain kinase. Arch Bio-chem Biophys 287:24–32

21. Hartshorne DJ, Ito M, Erdodi F 1998 Myosin light chain phos-phatase: subunit composition, interactions and regulation. J MuscleRes Cell Motil 19:325–341

22. Sanborn BM, Anwer K 1990 Hormonal regulation of myometrialintracellular calcium. In: Garfield RE (ed) Uterine Contractility.Serono Symposia USA, Norwell, MA, pp 69–82

23. Spencer GG, Khan I, Grover AK 1990 Ca21 regulation in smoothmuscle. In: Garfield RE (ed) Uterine Contractility. Serono SymposiaUSA, Norwell, MA, pp 53–68

24. Toro L, Stefani E, Erulkar S 1990 Hormonal regulation of potas-sium currents in single myometrial cells. Proc Natl Acad Sci USA87:2892–2895

25. Word RA, Stull JT, Kamm K, Casey ML 1990 Regulation ofsmooth-muscle contractility: Ca21 and myosin phosphorylation.In: Garfield RE (ed) Uterine Contractiity. Serono Symposia USA,Norwell, MA, pp 43–53

26. Word RA, Casey ML, Kamm K, Stull JT 1991 Effects of cGMP on[Ca21]i myosin light chain phosphorylation, and contraction inhuman myometrium. Am J Physiol 260:C861–C867

27. Asboth G, Phaneuf S, Europe-Finner GN, Toth M, Lopez-BernalA 1996 Prostaglandin E2 activates phospholipase C and elevatesintracellular calcium in cultured myometrial cells: involvement ofEP1 and EP3 receptor subtypes. Endocrinology 137:2572–2579

28. Walsh MP 1991 Calcium-dependent mechanism of regulation ofsmooth muscle contraction. Biochem Cell Biol 69:771–800

29. Lye SJ, Freitag CL 1988 An in vivo model to examine the electro-myographic activity of isolated myometrial tissue from pregnantsheep. J Reprod Fertil 82:51–61

30. Beyer EC, Kistler J, Paul DL, Goodenough DA 1989 Antiseradirected against connexin-43 peptides react with a 43 kD proteinlocalized to gap junctions in myocardium and other tissues. J CellBiol 108:595–605

31. Risek B, Guthrie S, Kumar N, Gilula NB 1990 Modulation of gapjunction transcript and protein expression during pregnancy in therat. J Cell Biol 110:269–282

32. Yu W, Dahl G, Werner R 1994 The connexin-43 gene is responsiveto oestrogen. Proc R Soc Lond B Biol Sci 255:125–132

33. Laird DW, Puranam KL, Revel J-P 1991 Turnover and phosphor-ylation dynamics of connexin-43 gap junction protein in culturedcardiac myocytes. Biochem J 273:67–72

34. Piersanti M, Lye SJ 1995 Increase in messenger ribonucleic acidencoding the myometrial gap junction protein, connexin-43, re-quires protein synthesis and is associated with increased expres-sion of the activator protein-1, c-fos. Endocrinology 136:3571–3578

35. Wathes DC, Porter DG 1982 Effect of uterine distension and oes-trogen treatment on gap junction formation in the myometrium ofthe rat. J Reprod Fertil 65:497–505

36. Meyer RA, Laird DW, Revel J-P, Johnson RG 1992 Inhibition ofgap junction and adherens junction assembly by connexin andA-CAM antibodies. J Cell Biol 119:179–189

37. Sakai N, Tabb T, Garfield RE 1992 Modulation of cell-to-cellcoupling between myometrial cells of the human uterus duringpregnancy. Am J Obstet Gynecol 167:472–480

38. Chow L, Lye SJ 1994 Expression of the gap junction protein, con-nexin-43, is increased in the human myometrium towards term andwith the onset of labour. Am J Obstet Gynecol 170:788–795

39. Winterhager E, Stutenkemper R, Traub O, Beyer EC, Willecke K1991 Expression of different connexin genes in rat uterus duringdecidualization and at term. Eur J Cell Biol 55:133–142

40. Fergusen II JE, Gorman JV, Bruns DE, Weir EC, Burtis WJ, MartinTJ, Bruns ME 1992 Abundant expression of parathyroid hormone-related protein in human amnion and its association with labor.Proc Natl Acad Sci USA 89:8384

41. van Meir CA, Matthews SG, Keirse MJNC, Ramirez MM, Bock-ing AD, Challis JRG 1997 15-Hydroxyprostaglandin dehydroge-nase (PGDH): implications in preterm labor with and withoutascending infection. J Clin Endocrinol Metab 82:969–976

42. Thiede MA, Daifotis AG, Weir EC, Brines ML, Burtis WJ, IkedaK, Dreyer BE, Garfield RE, Broadus AE 1990 Intrauterine occu-pancy controls expression of the parathyroid hormone-related pep-tide gene in preterm rat myometrium. Proc Natl Acad Sci USA87:6969–6973

43. Downing SJ, Sherwood OD 1985 The physiological role of relaxinin the pregnant rat. II. The influence of relaxin on uterine contractileactivitiy. Endocrinology 116:1206–1214

44. Porter DG, Downing SJ, Bradshaw JMC 1979 Relaxin inhibitsspontaneous and prostaglandin driven myometrial activity in an-aesthetized rats. J Endocrinol 83:183–192

45. Porter DG 1982 Unsolved problems of relaxin’s physiological role.Ann NY Acad Sci 380:151–162

46. Porter DG, Lye SJ, Bradshaw JMC, Kendall JZ 1981 Relaxin in-hibits myometrial activity in the ovariectomized non-pregnantewe. J Reprod Fertil 61:409–414

47. Porter DG, Watts AD 1986 Relaxin and progesterone are myome-trial inhibitors in the ovariectomized non-pregnant mini-pig. J Re-prod Fertil 76:205–213

48. Hansell DJ, Bryant-Greenwood GD, Greenwood FC 1991 Expres-sion of the human relaxin H1 gene in the decidua, trophoblast, andprostate. J Clin Endocrinol Metab 72:899–904

49. Bryant-Greenwood GD 1991 The human relaxins: consensus anddissent. Mol Cell Endocrinol 79:C125–132

50. Castracane VD, Lessing J, Brenner S, Weiss G 1985 Relaxin in thepregnant baboon: evidence for local production in reproductivetissues. J Clin Endocrinol Metab 60:133–136

51. Sakbun V, Ali SM, Greenwood FC, Bryant-Greenwood GD 1990Human relaxin in the amnion, chorion, decidua parietalis, basal

October, 2000 PARTURITION 539

Page 27: PDF (454 KB)

plate, and placental trophoblast by immunocytochemistry andnorthern analysis. J Clin Endocrinol Metab 70:508–514

52. MacLennan AH, Grant P, Borthwick AC 1991 Relaxin and relaxinc-peptide levels in human reproductive tissues. Reprod Fertil Dev3:577–583

53. Way SA, Leng G 1992 Relaxin increases the firing rate of supraopticneurones and increases oxytocin secretion in the rat. J Endocrinol132:149–158

54. Lye SJ, Challis JRG 1982 Inhibition by PGI2 of myometrial activityin vivo in non-pregnant ovariectomized sheep. J Reprod Fertil 66:311–315

55. Challis JRG, Lye SJ 1986 Parturition. In: Clarke MR (ed) OxfordReviews of Reproductive Biology. Oxford University Press, Ox-ford, UK, vol 8:61–129

56. Omini C, Folco GC, Pasargiklian R, Fano M, Berti F 1979 Pros-tacyclin (PG12) in pregnant human uterus. Prostaglandins 17:113–120

57. Williams KI, El Tahir KEH, Marcinkiewicz E 1979 Dual actions ofprostacyclin (PGI2) on the rat pregnant uterus. Prostaglandins 17:667–672

58. Mitchell MD, Lytton FD, Varticovski L 1988 Paradoxical stimu-lation of both lipocortin and prostaglandin production in humanamnion cells by dexamethasone. Biochem Biophy Res Commun151:137–141

59. Zuo J, Lei ZM, Rao CV, Pietrantoni M, Cook VD 1994 Differentialcyclooxygenase-1 and -2 gene expression in human myometriafrom preterm and term deliveries. J Clin Endocrinol Metab 79:894–899

60. Negishi M, Sugimoto Y, Ichikawa A 1995 Molecular mechanismsof diverse actions of prostanoid receptors. Biochim Biophys Acta1259:109–120

61. Grammatopoulos D, Hillhouse EW 1999 Role of corticotropin-releasing hormone in onset of labour. Lancet 354:1546–1549

62. Chwalisz K, Garfield RE 1997 Regulation of the uterus and cervixduring pregnancy and labor. Role of progesterone and nitric oxide.In: Bulleti C, De Ziegler D, Guller S, Levitz M (eds) The Uterus:Endometrium and Myometrium. New York Academy of Sciences,New York, p 238

63. Dayes BA 1990 Characterization of myometrial desensitization tob-adrenergic agonists. Can J Physiol Pharmacol 68:1377–1384

64. Riemer RK, Goldfien A, Roberts JM 1987 Rabbit myometrial ad-renergic sensitivity is increased by estrogen but is independent ofchanges in a adrenoceptor concentration. J Pharmacol Exp Ther240:44–50

65. Boyle MB, MacLusky NJ, Naftolin F, Kaczmarek LK 1987 Hor-monal regulation of K1-channel messenger RNA in rat myome-trium during oestrus cycle and in pregnancy. Nature 330:373–375

66. Kitts DD, Anderson GB, Bon Durant RG, Stabenfeldt GH 1984Temporal withdrawal patterns of D4C-21 steroids in coexisting,genetically dissimilar twin lamb fetuses throughout late gestation.Endocrinology 114:703–711

67. Kitts DD, Anderson GB, Bon Durant RG, Kindahl H, StabenfeldtGH 1985 Studies on the endocrinology of parturition: relative ste-roidogenesis in coexisting genetically dissimilar ovine fetuses, con-comitant with the temporal patterns of maternal C18 and C19 ste-roids and prostaglandin F2a release. Biol Reprod 33:67–78

68. Ou CO, Lye SJ 1997 Expression of connexin-43 and connexin-26 inthe rat myometrium during pregnancy and labour is regulated bymechanical and hormonal signals. Endocrinology 138:5398–5407

69. Chen Z-Q, Lefebvre DL, Bai X-H, Reaume A, Rossant J, Lye SJ1995 Identification of two regulatory elements within the promoterregion of the mouse connexin-43 gene. J Biol Chem 270:3863–3868

70. Sadoshima J, Izumo S 1993 Mechanical stretch rapidly activatesmultiple signal transduction pathways in cardiac myocytes: po-tential involvement of an autocrine/paracrine mechanism. EMBOJ 12:1681–1692

71. McDonald TJ, Nathanielsz PW 1991 Bilateral destruction of thefetal paraventricular nuclei prolongs gestation in sheep. Am J Ob-stet Gynecol 165:764–770

72. Gluckman PD, Mallard C, Boshier DP 1991 The effect of hypo-thalamic lesions on the length of gestation in fetal sheep. Am JObstet Gynecol 165:1464–1468

73. McDonald TJ, Hoffmann GE, Nathanielsz PW 1992 Hypotha-

lamic paraventricular nuclear lesions delay corticotroph matura-tion in the fetal sheep anterior pituitary. Endocrinology 131:1101–1106

74. Challis JRG, Lye SJ, Welsh J 1986 Ovine fetal adrenal maturationat term and during fetal ACTH administration: evidence that themodulating effect of cortisol may involve cAMP. Can J PhysiolPharmacol 64:1085–1090

75. Challis JRG, Brooks AN 1989 Maturation and activation of hy-pothalamic-pituitary adrenal function in fetal sheep. Endocr Rev10:182–204

76. Rose JC, Meis PJ, Morris M 1981 Ontogeny of endocrine (ACTH,vasopressin, cortisol) responses to hypotension in lamb fetuses.Am J Physiol 240:E656–E661

77. Bassett JM, Thorburn GD 1969 Foetal plasma corticosteroids andthe initiation of parturition in the sheep. J Endocrinol 44:285–286

78. Magyar DM, Fridshal D, Elsner CW, Glatz T, Eliot J, Klein AH,Lowe KC, Buster JE, Nathanielsz PW 1980 Time-trend analysis ofplasma cortisol concentrations in the fetal sheep in relation toparturition. Endocrinology 107:155–159

79. Norman LJ, Lye SJ, Wlodek ME, Challis JRG 1985 Changes inpituitary responses to synthetic ovine corticotrophin releasing fac-tor in fetal sheep. Can J Physiol Pharmacol 63:1398–1403

80. MacIsaac RJ, Bell RJ, McDougall JG, Tregear GW, Wang X, Win-tour EM 1985 Development of the hypothalamic-pituitary axis inthe ovine fetus: ontogeny of action of ovine corticotropin-releasingfactor. J Dev Physiol 7:329–338

81. Liggins GC 1994 The role of cortisol in preparing the fetus for birth.Reprod Fertil Dev 6:141–150

82. Matthews SG, Challis JRG 1995 Regulation of CRH and AVPmRNA in the developing ovine hypothalamus: effects of stress andglucocorticoids. Am J Physiol 268:E1096–E1107

83. Matthews SG, Han X, Lu F, Challis JRG 1994 Developmentalchanges in the distribution of pro-opiomelanocortin and prolactinmRNA in the pituitary of the ovine fetus and lamb. J Mol Endo-crinol 13:175–185

84. Matthews SG, Challis JRG 1996 Regulation of the hypothalamo-pituitary-adrenocortical axis in fetal sheep. Trends EndocrinolMetab 7:239–246

85. Merei JJ, Rao A, Clarke IJ, McMillen IC 1993 Proopiomelanocor-tin, prolactin and growth hormone messenger ribonucleic acidlevels in the fetal sheep pituitary during late gestation. Acta En-docrinol (Copenh) 129:263–267

86. McMillen IC, Mercer JE, Thorburn GD 1988 Pro-opiomelanocor-tin mRNA levels fall in the fetal sheep pituitary before birth. J MolEndocrinol 1:141–145

87. Holloway AC, Gyomorey S, Challis JRG 2000 Effects of labor onpituitary expression of proopiomelanocortin prohormone conver-tase (PC)-1, PC-2 and glucocorticoid receptor mRNA in fetal sheep.Endocrine, in press

88. Brieu V, Tonon MC, Lutz Bucher B, Durand P 1989 Corticotropin-releasing factor-like immunoreactivity, arginine vasopressin-likeimmunoreactivity and ACTH-releasing bioactivity in hypotha-lamic tissue from fetal and neonatal sheep. Neuroendocrinology49:164–168

89. Durand P, Cathiard AM, Dacheux F, Naaman E, Saez JM 1986 Invitro stimulation and inhibition of adrenocorticotropin release bypituitary cells from ovine fetuses and lambs. Endocrinology 118:1387–1394

90. Matthews SG, Challis JRG 1995 Corticotropin-releasing hormoneand vasopressin induced changes in pro-opiomelanocortin syn-thesis and adrenocorticotropin output from ovine fetal cortico-trophs, in vitro (abstract P353). J Soc Gynecol Invest 2 [Suppl]:393

91. Lu F, Yang K, Challis JRG 1991 Characteristics and developmentalchanges of corticotrophin-releasing hormone binding sites in thefoetal sheep anterior pituitary. J Endocrinol 130:223–229

92. Lu F, Yang K, Challis JRG 1994 Regulation of ovine fetal pituitaryfunction by corticotrophin-releasing hormone, arginine vasopres-sin and cortisol in vitro. J Endocrinol 143:199–208

93. Hargrave BY, Rose JC 1986 By 95 days of gestationCRF increasesplasma ACTH and cortisol in ovine fetuses. Am J Physiol 250:E422–E427

94. Norman LJ, Brooks AN, Challis JRG 1986 Pituitary and adrenal

540 CHALLIS ET AL. Vol. 21, No. 5

Page 28: PDF (454 KB)

responses to pulsatile ovine corticotrophin releasing factor (oCRF)administered to fetal sheep. Endocrinology 120:2383–2388

95. Norman LJ, Challis JRG 1987 Synergism between systemic corti-cotropin-releasing factor and arginine vasopressin on adrenocor-ticotrophin release in vivo varies as a function of gestational age inthe ovine fetus. Endocrinology 120:1052–1058

96. Liu J-P, Clarke IJ, Funder JW, Engler D 1994 Studies of the se-cretion of corticotropin-releasing factor and arginine vasopressininto the hypophysial-portal circulation of the conscious sheep. II.The central noradrenergic and neuropeptide Y pathways causeimmediate and prolonged hypothalamic-pituitary-adrenal activa-tion. Potential involvement in the pseudo-Cushing’s syndrome ofendogenous depression and anorexia nervosa. J Clin Invest 93:1439–1450

97. Levidiotis ML, Wintour EM, McKinley MJ, Oldfield BJ 1989Hypothalamic-hypophyseal vascular connections in the fetalsheep. Neuroendocrinology 49:47–50

98. Bell ME, Myers TR, Myers DA 1998 Expression of proopiomela-nocortin and prohormone convertase-1 and -2 in the late gestationfetal sheep pituitary. Endocrinology 139:5135–5143

99. Carr GA, Jacobs RA, Young IR, Schwartz J, White A, Crosby J,Thorburn GD 1995 Development of adrenocorticotropin-(1–39)and precursor peptide secretory responses in the fetal sheep duringthe last third of gestation. Endocrinology 136:5020–5027

100. Roebuck MM, Jones C, Holland D, Silman R 1980 In vitro effectsof high molecular weight forms of ACTH on the fetal sheep adrenal.Nature 284:616

101. Jones C, Roebuck MM 1980 ACTH peptides and the developmentof the fetal adrenal. J Steroid Biochem 12:77–82

102. Schwartz J, Ash P, Ford V, Raff H, Crosby S, Shite A 1994 Se-cretion of adrenocorticotrophin (ACTH) and ACTH precursors inovine anterior pituitary cells: actions of corticotrophin-releasinghormone, arginine vasopressin and glucocorticoids. J Endocrinol140:189–195

103. Schwartz J, Kleftogiannis F, Jacobs R, Thorburn GD, Crosby S,White A 1995 Biological activity of adrenocorticotropic hormoneprecursors on ovine adrenal cells. Am J Physiol 268:E623–E629

104. Saphier PW, Glynn BP, Woods RJ, Shepherd DA, Jeacock MK,Lowry PJ 1993 Elevated levels of N-terminal pro-opiomelanocortinpeptides in fetal sheep plasma may contribute to fetal adrenal glanddevelopment and the pre-parturient cortisol surge. Endocrinology133:1459–1461

105. Mulvogue HM, McMillen IC, Robinson PM, Perry RA 1986 Im-munocytochemical localization of progMSH, gMSH, ACTH andbendorphin/blipotrophin in the fetal sheep pituitary: an ontoge-netic study. J Dev Physiol 8:355–368

106. Antolovich GC, McMillen IC, Perry RA, Robinson PM, Silver M,Young IR 1988 The development of corticotrophs in the fetal sheeppars distalis. The effect of cortisol infusion or adrenalectomy orhypothalamo-pituitary disconnection (HPD). In: Jozak S (ed) Re-search in Perinatal Medicine. Perinatology Press, Ithaca, NY, pp243–246

107. Antolovich GC, McMillen IC, Robinson PM, Silver M, Young IR,Perry RA 1991 The effect of hypothalamo-pituitary disconnectionon the functional and morphological development of the pituitary-adrenal axis in the fetal sheep in the last third of gestation. Neu-roendocrinology 54:254–261

108. Myers DA, Myers TR, Grober MS, Nathanielsz PW 1993 Levelsof corticotrophin-releasing hormone messenger ribonucleic acid(mRNA) in the hypothalamic paraventricular nucleus and pro-opiomelanocortin mRNA in the pars distalis during late gestationin fetal sheep. Endocrinology 132:2109–2116

109. Wintour EM, Bell RJ, Fei DT, Southwell C, Tregear GW, WangX 1984 Synthetic ovine corticotropin-releasing factor stimulatesadrenocorticotropin release in the ovine fetus over the last fifth ofgestation. Neuroendocrinology 38:86–87

110. Wintour EM 1984 Developmental aspects of hypothalamic-pitu-itary-adrenal axis. J Dev Physiol 6:291–299

111. Challis JRG, Nancekievill EA, Lye SJ 1985 Possible role of cortisolin the stimulation of cortisol binding capacity in the plasma of fetalsheep. Endocrinology 116:1139–1144

112. Berdusco ET, Hammond GL, Jacobs R, Grolla A, Akagi K,Langlois D, Challis JRG 1993 Glucocorticoid-induced increase in

plasma corticosteroid binding globulin levels in fetal sheep is as-sociated with increased biosynthesis and alterations in glycosyla-tion. Endocrinology 132:2001–2008

113. Ballard PL, Kitterman JA, Bland RD, Clyman RI, Gluckman PD,Platzker ACG, Kaplan SL, Grumbach MM 1982 Ontogeny andregulation of corticosteroid binding globulin capacity in plasma offetal and newborn lambs. Endocrinology 110:359–366

114. Fairclough RJ, Liggins GC 1975 Protein binding of plasma cortisolin the foetal lamb near term. J Endocrinol 67:333–341

115. Challis JRG, Berdusco ET, Jeffray TM, Yang K, Hammond GL1995 Corticosteroid-binding globulin (CBG) in fetal development.J Steroid Biochem Mol Biol 53:523–527

116. Yang K, Matthews SG, Challis JRG 1995 Developmental andglucocorticoid regulation of pituitary 11-hydroxysteroid dehydro-genase 1 gene expression in the ovine fetus and lamb. J Mol En-docrinol 14:109–116

117. Yang K, Hammond GL, Challis JRG 1992 Characterization of anovine glucocorticoid receptor cDNA and developmental changes inits mRNA levels in the fetal sheep hypothalamus, pituitary andadrenal gland. J Mol Endocrinol 8:173–180

118. Matthews SG, Yang K, Challis JRG 1995 Changes in glucocorti-coid receptor mRNA in the developing ovine pituitary and theeffects of exogenous cortisol. J Endocrinol 144:483–490

119. McDonald TJ, Hoffmann GE, Myers DA, Nathanielsz PW 1990Hypothalamic glucocorticoid implants prevent fetal ovine adre-nocorticotropin secretion in response to stress. Endocrinology 127:2862–2868

120. Matthews SG, Challis JRG 1995 Developmental regulation of pre-proenkephalin mRNA in the ovine paraventricular nucleus: effectsof stress and glucocorticoids. Dev Brain Res 86:259–267

121. Hennessy DP, Coghlan JP, Hardy KJ, Scoggins BA, Wintour EM1982 The origin of cortisol in the blood of fetal sheep. J Endocrinol95:71

122. Challis JRG, Manchester EL, Mitchell BF, Patrick JE 1982 Acti-vation of adrenal function in fetal sheep by the infusion of adre-nocorticotropin (ACTH) to the fetus in utero. Biol Reprod 27:1026–1032

123. Jacobs R, Young IR, Hollingworth SA, Thorburn GD 1994Chronic administration of low doses of adrenocorticotropin to hy-pophysectomized fetal sheep leads to normal term labor. Endo-crinology 134:1389–1394

124. Glickman JA, Challis JRG 1980 The changing response pattern ofsheep fetal adrenal cells throughout the course of gestation. En-docrinology 106:1371–1376

125. Challis JRG, Lye SJ, Mitchell BF, Olson DM, Sprague C, NormanL, Power SGA, Siddigi J, Wlodek ME 1985 Fetal signals for birth.In: Jones C, Nathanielsz PW (eds) Physiological Development ofthe Fetus and Newborn. Academic Press, London, pp 363–370

126. Rose JC, Meis PJ, Urban RB, Greiss Jr FC 1982 In vivo evidence forincreased adrenal sensitivity to adrenocorticotrophin-(1–24) in thelamb fetus in late gestation. Endocrinology 111:80–85

127. Wintour EM, Brown EH, Denton DA, Hardy KJ, McDougall JG,Oddie CJ, Whipp GT 1975 The ontogeny and regulation of cor-ticosteroid secretion by the ovine foetal adrenal. Acta Endocrinol(Copenh) 79:301–316

128. Manchester EL, Challis JRG 1982 The effects of adrenocortico-tropin, guanylylimidodiphosphate, dibutyryl adenosine 39,59-monophosphate and exogenous substrates on corticosteroid out-put by ovine fetal adrenal cells at different times in pregnancy.Endocrinology 111:889–895

129. Tangalakis K, Coghlan JP, Connell J, Crawford R, Darling P,Hammond VE, Haralambidis J, Penschow J, Wintour EM 1989Tissue distribution and levels of gene expression of three steroidhydroxylases in ovine fetal adrenal glands. Acta Endocrinol(Copenh) 120:225–232

130. Conley AJ, Bird IM 1997 The role of cytochrome P450 17a-hydroxylase and 3b-hydroxysteroid dehydrogenase in the integra-tion of gonadal and adrenal steroidogenesis via the D5 and D4pathways of steroidogenesis in mammals. Biol Reprod 56:789–799

131. Tangalakis K, Coghlan JP, Crawford R, Hammond RE 1990 Ste-roid hydroxylase gene expression in the ovine fetal adrenal glandfollowing ACTH infusion. Acta Endocrinol (Copenh) 123:371–377

132. Riley SC, Boshier DP, Labrie F, Challis JRG 1992 Immunohisto-

October, 2000 PARTURITION 541

Page 29: PDF (454 KB)

chemical localization of 3b-hydroxysteroid dehydrogenase/D5-D4isomerase, tyrosine hydroxylase and phenylethanolamine N-methyltransferase in the adrenal glands of sheep fetuses throughout gestationand in neonates. J Reprod Fertil 96:127–134

133. Han VK, Lu F, Bassett N, Yang KP, Delhanty P, Challis JRG 1992Insulin-like growth factor-II (IGF-II) mRNA is expressed in steroi-dogenic cells of the developing ovine adrenal: evidence for anautocrine/paracrine role of IGF-II. Endocrinology 131:3100–3109

134. Holta M, Baird A 1986 Differential effects of transforming growthfactor type b on the growth and function of adrenocortical cells invitro. Proc Natl Acad Sci USA 83:7795–7799

135. Fraser M, Jeffray TM, Challis JRG Developmental regulation ofcorticotrophin receptor (ACTH-R) gene expression in the adrenalgland of the ovine fetus and newborn lamb: effects of cortisolinfusion during late pregnancy. Proceedings of the InternationalSociety of Fetal Physiology, Aspen, CO, 1999 (Abstract)

136. Wintour EM, Coghlan JP, Hardy KJ, Hennessy DP, Lingwood BE,Scoggins BA 1980 Adrenal corticosteroids and immunoreactiveACTH in chronically cannulated ovine fetuses with bilateral ad-renalectomy. Acta Endocrinol (Copenh) 95:546–552

137. Cone RD, Mountjoy KG 1993 Molecular genetics of the ACTH andmelanocyte-stimulating hormone receptors. Trends EndocrinolMetab 4:242–247

138. Durand P, Cathiard AM, Saez JM 1985 Involvement of the regu-latory protein Ns in the maturation of ACTH-sensitive adenylatecyclase of ovine fetal adrenal during late gestation. Mol Cell En-docrinol 39:145–150

139. Naaman E, Chatelain P, Saez JM, Durand P 1989 In vitro effect ofinsulin and insulin-like growth factor-I on cell multiplication andadrenocorticotropin responsiveness of fetal adrenal cells. Biol Re-prod 40:570–577

140. Penhoat A, Jaillard C, Saez JM 1989 Synergistic effects of corti-cotropin and insulin-like growth factor I on corticotropin receptorsand corticotropin responsiveness in cultured bovine adrenocorticalcells. Biochem Biophys Res Commun 165:355–359

141. Boshier DP, Holloway H, Liggins GC 1981 Effects of cortisol andACTH on adrenocortical growth and cytodifferentiation in thehypophysectomized fatal sheep. J Dev Physiol 3:355–373

142. Durand P, Cathiard AM, Saez JM 1982 In vitro maturation of ovinefetal adrenal cells adenylate cyclase: corticotropin-dependent andindependent development of the response to corticotropin. Bio-chem Biophys Res Commun 106:8–15

143. Durand P, Cathiard AM, Saez JM 1984 In vitro maturation ofsteroidogenic capacity of ovine fetal and neonatal adrenal cells.Endocrinology 114:1128–1134

144. Durand P, Cathiard AM, Morera A-M, Dazord A, Saez JM 1981Maturation of adrenocorticotropin-sensitive adenylate cyclase ofovine fetal adrenal during late pregnancy. Endocrinology108:2114–2119

145. Challis JRG, Huhtanen D, Sprague CL, Mitchell BF, Lye SJ 1985Modulation by cortisol of adrenocorticotropin-induced activationof adrenal function in fetal sheep. Endocrinology 116:2267–2272

146. Durand P, Locatelli A, Cathiard AM, Dazord A, Saez JM 1981ACTH induction of the maturation of ACTH-sensitive adenylatecyclase system in the ovine fetal adrenal. J Steroid Biochem 15:445–448

147. Durand P, Cathiard AM, Locatelli A, Saez JM 1982 Modificationsof the steroidogenic pathway during spontaneous and adrenocor-ticotropin-induced maturation of ovine fetal adrenal. Endocrinol-ogy 110:500–505

148. Lye SJ, Sprague CL, Mitchell BF, Challis JRG 1983 Activation ofovine fetal adrenal function by pulsatile or continuous adminis-tration of ACTH1–24. I. Effects on fetal plasma corticosteroids. En-docrinology 113:770–782

149. Manchester EL, Lye SJ, Challis JRG 1983 Activation of ovine fetaladrenal function by pulsatile or continuous administration of ad-renocorticotropin-(1–24). II. Effects on adrenal cell responses invitro. Endocrinology 113:777–782

150. Rainey WE, Oka K, Magness RR, Mason JI 1991 Ovine fetaladrenal synthesis of cortisol: regulation by adrenocorticotropin,angiotensin II and transforming growth factor-b. Endocrinology129:1784–1790

151. Jones C, Boddy K, Robinson JS, Ratcliffe JG 1977 Developmental

changes in the responses of the adrenal glands of the foetal sheepto endogenous adrenocorticotrophin as indicated by the hormoneresonses to hypoxaemia. J Endocrinol 72:279–292

152. Ozolins IZ, Young IR, McMillen IC 1992 Surgical disconnectionof the hypothalamus from the fetal pituitary abolishes the corti-cotrophic response to intrauterine hypoglycemia or hypoxemia inthe sheep during late gestation. Endocrinology 130:2438–2445

153. Stark RI, Daniel SS, Husain MK, Zubrow AB, James LS 1984Effects of hypoxia on vasopressin concentrations in cerebrospinalfluid and plasma of sheep. Neuroendocrinology 38:453–460

154. Stark RI, Daniel SS, Husain MK, Tropper PJ, James LS 1985Cerebrospinal fluid and plasma vasopressin in the fetal lamb: basalconcentration and the effect ofhypoxia. Endocrinology 116:65–72

155. Akagi K, Berdusco ET, Challis JRG 1990 Cortisol inhibits ACTHbut not the AVP response to hypoxaemia in fetal lambs at days123–128 of gestation. J Dev Physiol 14:319–324

156. McMillen IC, Phillips ID, Ross JT, Robinson JS, Owens JA 1995Chronic stress — the key to parturition? Reprod Fertil Dev 7:499–507

157. Matthews SG, Challis JRG 1995 Levels of pro-opiomelanocortinand prolactin mRNA in the fetal sheep pituitary following hypox-emia and glucocorticoid treatment in late gestation. J Endocrinol147:139–146

158. Braems GA, Matthews SG, Challis JRG 1996 Differential regula-tion of pro-opiomelanocortin mRNA in the ovine fetal pituitarypars distalis and pars intermedia following 48 hours of hypoxemiain late gestation. Endocrinology 137:2731–2738

159. Matthews SG, Fraser M, Challis JRG 1996 Dopaminergic regu-lation of pituitary function in the late gestation fetal sheep. J En-docrinol 150:187–194

160. Lackman F, Capewell V, Gagnon R, Richardson B, Fetal cord PO2,O2 saturation and fractional extraction (FE) values in relation tosize at birth. Program of the 49th Annual Meeting of the Society forGynecologic Investigation, Chicago, IL, 2000 (Abstract 233)

161. Richardson B, Nodwell A, Webster R, Alshimmiri M, Gagnon R,Natale R 1998 Fetal oxygen saturation and fractional extraction atbirth and the relationship measures of acidosis. Am J Obstet Gy-necol 178:572–579

162. Fraser M, Oliver MH, Harding JE, Gluckman PD, Challis JRG1999 Maternal undernutrition in late ovine pregnancy: effects onfetal adrenal corticotrophin receptor and steroidogenic enzymemRNA expression. Program of the 46th Annual Meeting of theSociety for Gynecologic Investigation, Atlanta, GA, 1999 (Abstract271)

163. Murotsuki J, Challis JRG, Gagnon R 1995 Increased fetal plasmaprostaglandin E2 concentrations during fetal embolization in preg-nant sheep. Am J Obstet Gynecol 173:30–35

164. Power SGA, Patrick JE, Carson GD, Challis JRG 1982 The fetalmembranes as a possible source of progesterone in the amnioticand allantoic fluids of pregnant sheep. Endocrinology 110:481–486

165. Challis JRG 1971 Sharp increase in free circulating oestrogen im-mediately before parturition in sheep. Nature 229:208

166. Challis JRG, Patrick JE 1981 Fetal and maternal estrogen concen-trations throughout pregnancy in the sheep. Can J Physiol Phar-macol 59:970–978

167. Jenkin G, Thorburn GD 1985 Inhibition of progesterone secretionby a 3b-hydroxysteroid dehydrogenase inhibitor in late pregnantsheep. Can J Physiol Pharmacol 63:136–142

168. Steele PA, Flint APP, Turnbull AC 1976 Activity of steroid C-17,20-lyase in the ovine placenta: effect of exposure of foetal glucocor-ticoid. J Endocrinol 69:239–246

169. Steele PA, Flint APF, Turnbull AC 1976 Increased utero-ovarianadrostenedione production before parturition in sheep. J ReprodFertil 46:443–445

170. Ma XH, Wu WX, Nathanielsz PW 1999 Differential effects of nat-ural and synthetic glucocorticoids on cytochrome 17a-hydroxylase(P45017a) and cytochrome P450 side-chain cleavage (P450scc) mes-senger ribonucleic acid in the sheep placenta. Am J Obstet Gynecol180:1215–1221

171. Wu WX, Owiny J, Zhang Q, Ma XH, Nathanielsz PW 1996 Reg-ulation of the estrogen receptor and its messenger ribonucleic acidin the ovariectomized sheep myometrium and endometrium: Therole of estradiol and progesterone. Biol Reprod 55:762–768

542 CHALLIS ET AL. Vol. 21, No. 5

Page 30: PDF (454 KB)

172. Wu WX, Ma XH, Nathanielsz PW 1999 Tissue-specific ontogenicexpression of prostaglandin H synthase 2 in the ovine myome-trium, endometrium, and placenta during late gestation and atspontaneous term labor. Am J Obstet Gynecol 181:1512–1519

173. Liggins GC, Fairclough RJ Grieves SA, Kendall JZ, Knox BS 1973The mechanism of initiation of parturition in the ewe. Recent ProgHorm Res 29:111–159

174. Evans CA, Kennedy TG, Patrick JE, Challis JRG 1982 The effectsof indomethacin on uterine activity and prostaglandin (PG) con-centrations during labor induced by administering ACTH to fetalsheep. Can J Physiol Pharmacol 60:1200–1209

175. Evans CA, Kennedy TG, Challis JRG 1982 Gestational changes inprostanoid concentration in intrauterine tissues and fetal fluidsfrom pregnant sheep, and the relation to prostanoid output in vitro.Biol Reprod 127:1–11

176. Wimsatt J, Nathanielsz PW, Sirois J 1993 Induction of prostaglan-din endoperoxide synthase isoform-2 in ovine cotyledonary tissuesduring late gestation. Endocrinology 133:1068–1073

177. Liggins GC, Grieves SA 1971 Possible role for prostaglandin F2a

in parturition in sheep. Nature 232:629–631178. Wu WX, Unno N, Ma XH, Nathanielsz PW 1998 Inhibition of

prostaglandin production by nimesulide accompanied by changesin expression of the cassette of uterine labor-related genes in preg-nant sheep. Endocrinology 139:3096–3103

179. Wu W, Ma XH, Zhang Q, Buchwalder L, Nathanielsz PW 1997Regulation of prostaglandin endoperoxide H synthase 1 and 2 byestradiol and progesterone in nonpregnant ovine myometrium andendometrium in vivo. Endocrinology 138:4005–4012

180. Wu WX, Ma XH, Nathanielsz PW 1999 Changes in prostacyclinsynthase in pregnant sheep myometrium, endometrium, and pla-centa at spontaneous term labor and regulation by estradiol andprogesterone. Am J Obstet Gynecol 180:744–749

181. Davies IJ, Ryan KJ, Petro Z 1970 Estrogen synthesis by adrenal-placental tissues of the sheep and the Iris monkey in vitro. Endo-crinology 86:1457–1459

182. Yu HK, Cabalum T, Jansen CAM, Buster JE, Nathanielsz PW 1983Androstenedione, testosterone, and estradiol concentrations in fe-tal and maternal plasma in late pregnancy in the sheep. Endocri-nology 113:2216–2220

183. Mitchell BF, Lye SJ, Lukash L, Challis JRG 1986 Androstenedionemetabolism in the late gestation sheep fetus. Endocrinology 118:63–68

184. Dwyer RJ, Robertson HA 1980 Oestrogen sulphate and sulpho-transferase activitites in the endometrium of the sow and eweduring pregnancy. J Reprod Fertil 60:187–191

185. Liggins GC, Thorburn GD 1994 Initiation of parturition. In: Lam-ming GE (ed) Marshall’s Physiology of Reproduction. Chapmanand Hall, London, pp 863–1002

186. Olson DM, Lye SJ, Skinner K, Challis JRG 1984 Early changes inprostaglandin concentrations in ovine maternal and fetal plasmaamniotic fluid and from dispersed cells of intrauterine tissues be-fore the onset of ACTH-induced preterm labor. J Reprod Fertil71:45–55

187. Olson DM, Lye SJ, Skinner K, Challis JRG 1985 Prostanoid con-centrations in maternal/fetal plasma and amniotic fluid and in-trauterine tissue prostanoid ouptut in relation to myometrial con-tractility during the onset of adrenocorticotropin-induced pretermlabor in sheep. Endocrinology 116:389–397

188. Wimsatt J, Myers DA, Myers TR, Nathanielsz PW 1995 Prosta-glandin synthase activity of fetal sheep cotyledons at 122 days ofgestation and term: expression of prostaglandin synthetic capacityin fetal cotyledonary tissue near labor is location-dependent. BiolReprod 52:737–744

189. Wimsatt J, Nathanielsz PW 1995 Prostaglandin H synthase activityin the sheep placenta during cortisol-induced labor at 128–131 daysof gestation and during spontaneous delivery at term. Prostaglan-dins Leukot Essent Fatty Acids 53:53–58

190. Boshier DP, Jacobs RA, Han VK, Smith W, Riley SC, Challis JRG1991 Immunohistochemical localization of prostaglandin H syn-thase in the sheep placenta from early pregnancy to term. BiolReprod 45:322–327

191. Gibb W, Matthews SG, Challis JRG 1996 Localization and de-velopmental changes in prostaglandin H synthase (PGHS) and

PGHS messenger ribonucleic acid in ovine placenta throughoutgestation. Biol Reprod 54:654–659

192. Langlois DA, Fraher LJ, Khalil MW, Fraser M, Challis JRG 1993Preferential increase in cyclooxygenase compared to lipoxygenaseactivity in sheep placenta and amnion at term pregnancy and afterintrafetal glucocortical administration. J Endocrinol 139:195–204

193. Rice GE, Wong MH, Thorburn GD 1988 Gestational changes inprostaglandin synthase activity in ovine cotyledonary microsomes.J Endocrinol 118:265–270

194. Rice GE, Payne MJ, Wong MH, Thorburn GD 1992 Immunore-active prostaglandin G/H synthase content increases in ovine cot-yledons during late gestation. Placenta 13:429–437

195. Rice GE, Freed KA, Aitken MA Jacobs RA 1995 Gestational- andlabour-associated changes in the relative abundance of prostaglan-din G/H synthase-1 and -2 mRNA in ovine placenta. J Mol En-docrinol 14:237–245

196. Risbridger GP, Leach Harper CM, Wong MH, Thorburn GD 1985Gestational changes in prostaglandin production by ovine fetaltrophoblast cells. Placenta 6:117–126

197. Zhang Q, Wu W, Brenna T, Nathanielsz PW 1996 The expressionof cytosolic phospholipase A2 and prostaglandin endoperoxidesynthase in ovine maternal uterine and fetal tissues during lategestation and labor. Endocrinology 137:4010–4017

198. Challis JRG, Dilley SR, Robinson JS, Thorburn GD 1976 Pros-taglandins in the circulation of the fetal lamb. Prostaglandins 11:1041–1052

199. Fowden AL, Harding R, Ralph MM, Thorburn GD 1987 Thenutritional regulation of plasma prostaglandin E concentrations inthe fetus and pregnant ewe during late gestation. J Physiol 394:1–12

200. Louis TM, Challis JRG, Robinson JS, Thorburn GD 1976 Rapidincrease of foetal corticosteroids after prostaglandin E2. Nature264:797–798

201. Hollingworth SA, Deayton JM, Young IR, Thorburn GD 1995Prostaglandin E2 administered to fetal sheep increases the plasmaconcentration of adrenocorticotropin (ACTH) and the proportionof ACTH in low molecular weight forms. Endocrinology 136:1233–1240

202. Liggins GC, Scroop GC, Haughey KG 1982 Comparison of theeffects of prostaglandin E2, prostacyclin and 1–24 adrenocortico-trophin on plasma cortisol levels of fetal sheep. J Endocrinol 95:153–162

203. Brooks AN, Gibson F 1992 Prostaglandin E2 enhances AVP-stim-ulated but not CRF-stimulated ACTH secretion from cultured fetalsheep pituitary cells. J Endocrinol 132:33–38

204. Young IR, Deayton JM, Hollingworth SA, Thorburn GD 1996Continuous intrafetal infusion of prostaglandin E2 prematurelyactivates the hypothalamo-pituitary-adrenal axis and induces par-turition in sheep. Endocrinology 137:2424–2431

205. Thorburn GD, Rice GE 1990 Placental PGE2 and the initiation ofparturition in the sheep. In: Mitchell MD (ed) Eicosanoids in Re-production. CRC Press, Boca Raton, FL, pp 73–86

206. Unno N, Wu WX, Wong CH, Bennett PR, Shinozuka N,Nathanielsz PW 1998 Prostaglandin regulation of fetal plasmaadrenocorticotropin and cortisol concentrations in late-gestationsheep. Biol Reprod 58:514–519

207. Challis JRG, Lye SJ, Gibb W 1997 Prostaglandins and parturition.Ann NY Acad Sci 828:254–267

208. Gyomorey S, Lye SJ, Gibb W, Challis JRG 2000 Fetal to maternalprogression of prostaglandin H(2) synthase-2 expression in ovineintrauterine tissues during the course of labor. Biol Reprod 62:797–805

209. Flint APP, Anderson ABM, Steele PA, Turnbull AC 1975 Themechanism by which foetal cortisol controls the onset of parturitionin the sheep. Biochem Soc Trans 3:1189–1194

210. Mason JI, France JT, Magness RR, Murry BA, Rosenfeld CR 1989Ovine placental steroid 17a-hydroxylase/C-17,20-lyase, aromataseand sulphatase in dexamethasone-induced and natural parturition.J Endocrinol 122:351–359

211. Whittle WL, Holloway AC, Lye SJ, Gibb W, Challis JRG 2000Prostaglandin production at the onset of ovine parturition is reg-ulated by both estrogen-independent and estrogen dependentpathways. Endocrinology, in press

212. Leung ST, Wathes DC, Young IR, Jenkin G 1999 Effect of labor

October, 2000 PARTURITION 543

Page 31: PDF (454 KB)

induction on the expression of oxytocin receptor, cytochrome P450aromatase, and estradiol receptor in the reproductive tract of thelate-pregnant ewe. Biol Reprod 60:814–820

213. Malpas P 1933 Postmaturity and malformations of the foetus. JObstet Gynaecol Br Emp 40:1046

214. Novy MJ, Walsh SW, Kittinger GW 1977 Experimental fetal anen-cephaly in the rhesus monkey: effect on gestational length and fetaland maternal plasma steroids. J Clin Endocrinol Metab 45:1031–1038

215. Chez RA, Hutchinson DL, Salazar H, Mintz DH 1970 Some effectsof fetal and maternal hypophysectomy in pregnancy. Am J ObstetGynecol 108:643–650

216. Nathanielsz PW, Figueroa JP, Honnebier MBOM 1992 In therhesus monkey placental retention after fetectomy at 121 to 130days’ gestation outlasts the normal duration of pregnancy. Am JObstet Gynecol 166:1529–1535

217. Albrecht ED, Haskins AL, Pepe GJ 1980 The influence of fetectomyat mid gestation upon the peripheral serum concentrations of pro-gesterone, estrone and estradiol in baboons. Endocrinology 107:766–770

218. Albrecht ED, Pepe GJ 1985 The placenta remains functional fol-lowing fetectomy in baboons. Endocrinology 116:843–845

219. Lanman JT 1977 Parturition in non-human primates. Biol Reprod16:28–38

220. Challis JRG, Davies IJ, Benirschke K, Hendrickx AG, Ryan KJ1974 The concentrations of progesterone, estrone and estradiol-17bin the peripheral plasma of the rhesus monkey during the finalthird of gestation and after the induction of abortion with PGF2a.Endocrinology 95:547–553

221. Pepe GJ, Albrecht ED 1995 Actions of placental and fetal adrenalsteroid hormones in primate pregnancy. Endocr Rev 16:608–648

222. Figueroa JP, Honnebier MBOM, Binienda Z, Wimsatt J,Nathanielsz PW 1989 Effect of 48 hours intravenous 4A andro-stenedione infusion on the pregnant rhesus monkey during thelast third of gestation: changes in maternal plasma estradiolconcentrations and myometrial contractility. Am J Obstet Gy-necol 161:481– 486

223. Mecenas CA, Giussani DA, Owiny J, Jenkins SL, Wu WX, Hon-nebier MBOM, Lockwood CJ, Kong L, Guller S, Nathanielsz PW1996 Production of premature delivery in pregnant rhesus mon-keys by androstenedione infusion. Nat Med 2:443–448

224. Pepe GJ, Albrecht ED 1990 Regulation of the primate fetal adrenalcortex. Endocr Rev 11:151–176

225. Mesiano S, Jaffe RB 1997 Developmental and functional biologyof the primate fetal adrenal cortex. Endocr Rev 18:378–403

226. Ackland JF, Ratter SJ, Bourne GL, Rees LH 1986 Corticotrophin-releasing factor-like immunoreactivity and bioactivity of humanfetal and adult hypothalami. J Endocrinol 108:171–180

227. Berghorn KA, Albrecht ED, Pepe GJ 1991 Responsivity of thebaboon fetal pituitary to corticotropin-releasing hormone in uteroat mid-gestation. Endocrinology 129:1424–1428

228. Pepe GJ, Davies WA, Albrecht ED 1994 Activation of the baboonfetal pituitary-adrenocortical axis at midgestation by estrogen: en-hancement of fetal pituitary proopiomelanocortin messenger ribo-nucleic acid expression. Endocrinology 135:2581–2587

229. Pepe GJ, Babischkin JS, Burch MG, Leavitt MG, Albrecht ED1996 Developmental increase in expression of the messenger ribo-nucleic acid and protein levels of 11b-hydroxysteroid dehydroge-nase types 1 and 2 in the baboon placenta. Endocrinology 137:5678–5684

230. Stewart PM, Rogerson FM, Mason JI 1995 Type 2 11b-hydroxy-steroid dehydrogenase messenger ribonucleic acid and activity inhumanplacenta and fetal membranes: its relationship to birthweight and putative role in fetal adrenal steroidogenesis. J ClinEndocrinol Metab 80:885–890

231. Jaffe RB, Seron-Ferre M, Mitchell BF 1979 Perinatal regulation ofcortisol in the primate. J Steroid Biochem 11:549–555

232. Voutilainen R, Miller WL 1987 Coordinate trophic hormone reg-ulation of mRNAs for insulin-like growth factor II and the cho-lesterol side-chain cleavage enzyme, P450scc in human steroido-genic tissues. Proc Natl Acad Sci USA 84:1590–1594

233. Albrecht ED, Henson MC, Walker ML, Pepe GJ 1990 Modulationof adrenocorticotropin-stimulated baboon fetal adrenal dehydro-

epiandrosterone formation in vitro by estrogen at mid- and late-gestation. Endocrinology 126:3083–3088

234. Pepe GJ, Albrecht ED 1985 Regulation of baboon fetal adrenalandrogen production by adrenocorticotropin hormone, prolactin,and growth hormone. Biol Reprod 33:545–550

235. Pepe GJ, Albrecht ED 1985 Prolactin stimulates adrenal androgensecretion in infant baboons. Endocrinology 117:1968–1973

236. Aberdeen GW, Babischkin JS, Davies WA, Pepe GJ, Albrecht ED1997 Decline in adrenocorticotropin receptor messenger ribonu-cleic acid expression in the baboon fetal adrenocortical zone in thesecond half of pregnancy. Endocrinology 138:1634–1641

237. Albrecht ED, Aberdeen GW, Babischkin JS, Tilly JL, Pepe GJ1996 Biphasic developmental expression of adrenocorticotropinreceptor messenger ribonucleic acid levels in the baboon fetal ad-renal gland. Endocrinology 137:1292–1298

238. Pepe GJ, Waddell BJ, Albrecht ED 1989 Effect of estrogen onpituitary peptide-induced dehydroepiandrosterone secretion inthe baboon fetus at mid gestation. Endocrinology 125:1519–1524

239. Ryan KJ 1969 Theoretical basis for endocrine control of gestation-acomparative approach. In: Pecile A, Finzi C (eds) Feto-PlacentalUnit. Exerpta Medica Foundation, Amsterdam, pp 120–132

240. Novy MJ 1977 Endocrine and pharmacological factors which in-fluence the onset of labour in rhesus monkeys. Ciba Found Symp47:259–295

241. Tulchinsky D, Hobel CJ, Yeager E, Marshall JR 1972 Plasmaestrone, estradiol, progesterone and 17-hydroxy-progesterone inhuman pregnancy. I. Normal pregnancy. Am J Obstet Gynecol112:1095–1100

242. Coulter CL, Goldsmith PC, Mesiano S, Voytek CC, Martin MC,Mason JI, Jaffe RB 1996 Functional maturation of the primate fetaladrenal in vivo. II. Ontogeny of corticosteroid synthesis is depen-dent upon specific zonal expression of 3 b-hydroxysteroid dehy-drogenase/isomerase. Endocrinology 137:4953–4959

243. Smith R, Mesiano Chan EC, Brown S, Jaffe RB 1998 Corticotropin-releasing hormone directly and preferentially stimulates dehydro-epiandrosterone sulfate secretion by human fetal adrenal corticalcells. J Clin Endocrinol Metab 83:2916–2920

244. Albrecht ED, Babischkin JS, Davies WA, Leavitt MG, Pepe GJ1999 Identification and developmental expression of the estrogenreceptor a and b in the baboon fetal adrenal gland. Endocrinology140:5953–5961

245. Simpson ER, Carr BR, John ME, Parker CR, Zuber MX, OkamuraT, Waterman MR, Mason JI 1985 Cholesterol metabolism in theadrenals of normal and anencephalic fetuses. In: Albrecht E, PepeGJ (eds) Research in Perinatal Medicine: Perinatal Endocrinology.Perinatology Press, Ithaca, NY, pp 161–173

246. Winkel CA, Snyder JM, MacDonald PC, Simpson ER 1980 Reg-ulation of cholesterol and progesterone synthesis in human pla-cental cells in culture by serum lipoproteins. Endocrinology 106:1054–1060

247. Yoon BH, Romero R, Jun JK, Maymon E, Gomez R, Mazor M,Park JS 1998 An increase in fetal plasma cortisol but not dehy-droepiandrosterone sulfate is followed by the onset of pretermlabor in patients with preterm premature rupture of the mem-branes. Am J Obstet Gynecol 179:1107–1114

248. Darne J, McGarrigle HHG, Lachelin GCL 1987 Saliva oestriol,oestradiol, oestrone and progesterone levels in pregnancy: spon-taneous labour at term is preceded by a rise in the salive oestriol:progesterone ratio. Br J Obstet Gynaecol 94:227–235

249. Darne J, McGarrigle HHG, Lachelin GCL 1987 Increased salivaoestriol to progesterone ratio before idiopathic preterm delivery: apossible predictor for preterm labor? Br Med J 294:270–272

250. Romero R, Scoccia B, Mazor M, Wu YK, Benveniste R 1988 Ev-idence for a local change in the progesterone/estrogen ratio inhuman parturition. Am J Obstet Gynecol 159:657–660

251. Csapo A 1977 The “see-saw” theory of parturition. In: Knight J,O’Connor M (eds) The Fetus and Birth (Ciba Foundation Sympo-sium). Elsevier, Amsterdam, pp 159–172

252. Lye SJ, Porter DG 1978 Demonstration that progesterone “blocks”uterine activity in the ewe in vivo by a direct action on the myo-metrium. J Reprod Fertil 52:87–94

253. Avrech OM, Golan A, Weinraub Z, Bukovsky I, Caspi E 1991Mifepristone (RU486) alone or in combination with a prostaglandin

544 CHALLIS ET AL. Vol. 21, No. 5

Page 32: PDF (454 KB)

analogue for termination of early pregnancy: a review. Fertil Steril56:385–393

254. Csapo AL 1969 The four direct regulatory factors of myometrialfunction.. In: Knight J, Wolstenholme GEW (eds) Progesterone: ItsRegulatory Effect on the Myometrium. Churchill, London, 13

255. Babischkin JS, Pepe GJ, Albrecht ED 1997 Estrogen regulation ofplacental P450 cholesterol side-chain cleavage enzyme messengerribonucleic acid levels and activity during baboon pregnancy. En-docrinology 138:452–459

256. Riley SC, Bassett NS, Berdusco ET, Yang K, Leystra-Lantz C,Luu-The V, Labrie F, Challis JRG 1993 Changes in the abundanceof mRNA for type 1 3bhydroxysteroid dehydrogenase/D5—D4isomearase in the human placenta and fetal membranes duringpregnancy and labor. Gynecol Obstet Invest 35:199–203

257. Riley SC, Walton JC, Luu-The Labrie F, Challis JRG 1992 Im-munohistochemical localization of 3b-hydroxy-5-ene-steroid de-hydrogenase/D53D4 isomerase in human placenta and fetal mem-branes. J Clin Endocrinol Metab 75:956–961

258. Mitchell BF, Challis JRG 1988 Estrogen and progesterone metab-olism in human fetal membranes. In: Mitchell BF (ed) The HumanFetal Membranes: Structure and Function. Perinatology Press, NewYork, pp 5–28

259. Milewich L, Grant NF, Schwarz BE, Chen GT, MacDonald PC1977 Initiation of human parturition. VIII. Metabolism of proges-terone by fetal membranes of early and late human gestation.Obstet Gynecol 50:45–48

260. Mitchell BF, Cruickshank B, McLean D, Challis JRG 1982 Localmodulation of progesterone production in human fetal mem-branes. J Clin Endocrinol Metab 55:1237–1239

261. Erb G, Purdy RH, Lye SJ, Morrow RJ, MacLusky NJ, Circulatingand amniotic fluid sex steroid concentrations in human term preg-nancy: does a change in steroid 5a-reduction signal the onset oflabor? Steroids, in press

262. Grazzini E, Guillon G, Mouillac B, Zingg HH 1998 Inhibition ofoxytocin receptor function by direct binding of progesterone. Na-ture 392:509–512

263. Karalis K, Goodwin G, Majzoub JA 1996 Cortisol blockade ofprogesterone: a possible molecular mechanism involved in theinitiation of human labor. Nat Med 2:556–560

264. Casey ML, MacDonald PC 1996 Transforming growth factor-binhibits progesterone-induced enkephalinase expression in humanendometrial stromal cells. J Clin Endocrinol Metab 81:4022–4027

265. Stevens Y, Challis JRG, Lye SJ 1998 Corticotropin-releasing hor-mone receptor subtype 1 (CRH-R1) is significantly upregulated atthe time of labor in the human myometrium. J Clin EndocrinolMetab 83:4107–4115

266. Sparey C, Robson S, Bailey J, Lyall F, Europe-Finner GN 1999 Thedifferential expression of myometrial connexin-43, cyclooxygen-ase-1 and -2, and Gsa proteins in the upper and lower segments ofthe human uterus during pregnancy and labor. J Clin EndocrinolMetab 84:1705–1710

267. Teoh T-G, Chen Z-Q, Qi S-L, Lye SJ 1997 Paradoxical expressionof inhibitory and stimulatory prostanoid receptors in the humanmyometrium during labour. J Soc Gynecol Invest 4:565 (Abstract)

268. Teoh T-G, Orsini A, Chen Z-Q, Lye SJ 1997 Differential expressionof connexins 43 and 26 in the human myometrium during preg-nancy and labour. J Soc Gynecol Invest 4:342 (Abstract)

269. Wikland M, Lingwood BE, Wiqvist N 1984 Myometrial responseto prostaglandins during labour. Gynecol Obstet Invest 17:131–138

270. Leppert PC 1998 Proliferation and apoptosis of fibroblasts andsmooth muscle cells in rat uteri cervix throughout gestation and theeffect of the antiprogesterone anapristone. Am J Obstet Gynecol178:713–725

271. Mahendroo MS, Cala KM, Russell DW 1996 5a-reduced andro-gens play a key role in murine parturition. Mol Endocrinol 10:380–392

272. Mahendroo MS, Porter A, Russell DW, Word RA 1999 The par-turition defect in steroid 5a-reductase type 1 knockout mice is dueto impaired cervical ripening. Mol Endocrinol 13:981–992

273. Ledger WL, Webster MA, Anderson ABM, Turnbull AC 1985Effect of inhibition of prostaglandin synthesis on cervical softeningand uterine activity during ovine parturition resulting from pro-

gesterone withdrawal induced by epostane. J Endocrinol 105:227–233

274. Mitchell MD 1984 The mechanism(s) of human parturition. J DevPhysiol 6:107–118

275. Chard T 1977 Oxytocin. In: Martini L, Besser GM (eds) In: ClinicalNeuroendocrinology. Academic Press, New York, pp 569–583

276. Soloff MS 1988 The role of oxytocin in the initiation of labour andoxytocin-prostaglandin interactions. In: McNellis D, Challis JRG,MacDonald PC, Nathanielsz PW, Roberts JM (eds) The Onset ofLabour: Cellular and Integrative Mechanisms. Perinatology Press,Ithaca, NY, pp 191–203

277. Zingg HH, Lefebvre DL 1988 Oxytocin and vasopressin gene ex-pression during gestation and lactation. Mol Brain Res 4:1–6

278. Fuchs AR 1985 Oxytocin secretion and milk ejection in the human.In: Amico JA, Robinson AG (eds) In: Oxytocin in Animal Parturi-tion. Excerpta Medica, Amsterdam, pp 200–206

279. Chard T 1989 Fetal and maternal oxytocin in human parturition.Am J Perinatol 6:145–152

280. Fuchs AR, Fuchs F, Husslein R, Soloff MS, Fernstrom MJ 1982Oxytocin receptors and human parturition. A dual role for oxytocinin the initiation of labor. Science 215:1396–1398

281. Gross GA, Imamura T, Luedke CE, Vogt SK, Olson LM, NelsonDM, Sadovsky Y, Muglia LJ 1998 Opposing actions of prostaglan-dins and oxytocin determine the onset of murine labor. Proc NatlAcad Sci USA 95:11871–11875

282. Muglia LJ 2000 Genetic analysis of fetal development and partu-rition control in the mouse. Pediat Res 47:437–443

283. Honnebier MBOM, Figueroa JP, Rivier J, Vale W, Nathanielsz PW1989 Studies on the role of oxytocin in late pregnancy in the pregnantrhesus monkey: plasma concentrations of oxytocin in the maternalcirculation throughout the 24-h day and the effect of the syntheticoxytocin antagonist [1-b-Mpa(b-(CH2)5) 1.(Me(Tyr2,Orn8)] oxytocinon spontaneous nocturnal myometrial contractions. J Dev Physiol12:225–232

284. Riemer RK, Goldfien AC, Goldfien A, Roberts JM 1986 Rabbituterine oxytocin receptors and in vitro contractile response: abruptchanges at term and the role of eicosanoids. Endocrinology 119:699–709

285. Soloff MS, Alexandrova M, Fernstrom MJ 1979 Oxytocin recep-tors: triggers for parturition and lactation? Science 204:1313–1315

286. El Alj A, Bonoris E, Cynober E, Germain G 1990 Heterogeneity ofoxytocin receptors in the pregnant rat myometrium near parturi-tion. Eur J Pharmacol 186:231–238

287. Honnebier MBOM, Myers TR, Figueroa JP, Nathanielsz PW 1989Variation in myometrial response to intravenous oxytocin admin-istration at different times of the day in the pregnant rhesus mon-key. Endocrinology 125:1498–1503

288. Lefebvre DL, Giaid A, Bennett H, Lariviere R, Zingg HH 1992Oxytocin gene expression in rat uterus. Science 256:1553

289. Lefebvre DL, Lariviere R, Zingg HH 1993 Rat amnion: a novel siteof oxytocin production. Biol Reprod 48:632–639

290. Chibbar R, Miller FD, Mitchell BF 1993 Synthesis of oxytocin inamnion, chorion and decidua may influence the timing of humanparturition. J Clin Invest 91:185–192

291. Mitchell BF, Cross J, Hobkirk R, Challis JRG 1984 Formation ofunconjugated estrogens from estrone sulfate by dispersed cellsfrom human fetal membranes and decidua. J Clin EndocrinolMetab 58:845–849

292. Chibbar R, Wong S, Miller FD, Mitchell BF 1995 Estrogen stim-ulates oxytocin gene expression in human chorio-decidua. J ClinEndocrinol Metab 80:567–572

293. Richard S, Zingg HH 1990 The human oxytocin gene promoter isregulated by estrogens. J Biol Chem 265:6098–6103

294. Zhuge R, Li S, Chen TH, Hsu WH 1995 Oxytocin induced abiphasic increase in the intracellular CA21 concentration of porcinemyometrial cells: participation of a pertussis toxin-insensitive G-protein, inositol 1,4,5-trisphosphate-sensitive Ca21 pool, andCa21 ion channels. Mol Reprod Dev 41:20–28

295. Skinner K, Challis JRG 1985 Changes in the synthesis and me-tabolism of prostaglandins by human fetal membranes and deciduaat labor. Am J Obstet Gynecol 151:519–523

296. Sugimoto Y, Yamasaki A, Segi E, Tsuboi K, Aze Y, Nishimura T,Oida H, Yoshida N, Tanaka T, Katsuyarna M, Hasumoto K, Mu-

October, 2000 PARTURITION 545

Page 33: PDF (454 KB)

rata T, Hurata M, Ushikubi F, Negishi M, Ichikawa A, NarumiyaS 1997 Failure of parturition in mice lacking the prostaglandin Freceptor. Semin Reprod Endocrinol 27710:681–683

297. Langenbach R, Morham SG, Tiano HF, Loftin CD, Ghanayem BI,Chulada PC, Mahler JF, Lee CA, Goulding EH, Kluckman KD,Ledford A, Lee CA 1995 Prostaglandin synthase 1 gene disruptionin mice reduces arachidonic acid-induced inflammation and indo-methacin-induced gastric ulceration. Cell 83:483–492

298. Morham SG, Langenbach R, Loftin CD, Tiano HF, VouloumanosN, Jennette JC, Mahler JF, Kluckman KD, Ledford A, Lee CA 1995Prostaglandin synthase 2 gene disruption causes severe renal pa-thology in the mouse. Cell 83:473–482

299. Romero R, Munoz H, Gomez R, Parra M, Polanco M, Valverde V,Hasbun J, Garrido J, Ghezzi M, Mazor M, Tolosa JE, Mitchell MD1996 Increase in prostaglandin bioavailability precedes the onset ofparturition. Prostaglandins Leukot Essent Fatty Acids 54:187–191

300. Brown NL, Alvi SA, Elder MG, Bennett PR, Sullivan MH 1998 Aspontaneous induction of fetal membrane prostaglandin produc-tion precedes clinical labour. J Endocrinol 157:R1–R6

301. Keirse MJNC, Turnbull AC 1973 E prostaglandins in amnioticfluid during late pregnancy and labor. J Obstet Gynaecol Br Com-monw 80:970–973

302. Clark JD, Lin L-L, Kriz RW, Ramesha CA, Sultzman LA, Lin AY,Milona N, Knopf JL 1991 A novel arachidonic acid-selective cys-tolic PLA2 contains a Ca21-dependent translocation domain withhomology to PKC and GAP. Cell 65:1043–1051

303. Rajabi MR, Cybulsky AV 1995 Phospholipase A2 activity is in-creased in guinea pig uterine cervix in late pregnancy and at par-turition. Am J Physiol 269:E940–E947

304. Skannal DG, Brockman DE, Eis ALW, Xue S, Siddiqi TA, MyattL 1997 Changes in activity of cytosolic phospholipase A2 in humanamnion at parturition. Am J Obstet Gynecol 177:179–184

305. Skannal DG, Eis ALW, Brockman DE, Siddiqi TA, Myatt L 1997Immunohistochemical localization of phospholipase A2 isoformsin human myometrium during pregnancy and parturition. Am JObstet Gynecol 176:878–882

306. Uozumi N, Kume K, Nagase T, Nakatani N, Ishii S, Tashiro F,Komagata Y, Maki K, Ikuta K, Ouchi Y, Migazaki , Shimizu T1997 Role of cytosolic phospholipase A2 in allergic response andparturition. Nature 390:618–622

307. Xue S, Slater DM, Myatt L 1996 Induction of both cystosolic phos-pholipase A2 and prostaglandin H synthase-2 by interleukin-1b inWISH cells is inhibited by dexamethasone. Prostaglandins 51:107–124

308. Xue S, Brockman DE, Slater DM, Myatt L 1995 Interleukin-1binduces the synthesis and activity of cystosolic phospholipase A2and the release of prostaglandin E2 in human amnion-derivedWISH cells. Prostaglandins 49:351–369

309. Olson DM, Mijovic JE, Sadowsky DW 1995 Control of humanparturition. Semin Perinatol 19:52–63

310. Smith WL, DeWitt DL 1996 Prostaglandin endoperoxide H syn-thases-1 and -2. Adv Immunol 62:167–215

311. Smith WL, Garavito RM, DeWitt DL 1996 Prostaglandin endo-peroxide H synthases (cyclooxygenases)-1 and -2. J Biol Chem271:33157–33160

312. Xu X-M, Hajibeige A, Tazawa R, Loose-Mitchell D, Want L-H, WuKK 1995 Characterization of human prostaglandin H synthasegenes. In: Samuelsson B, Paoletti R (eds) Advances in Prostaglan-din, Thromboxane, and Leukotriene Research. Raven Press, NewYork, pp 105–107

313. Baguma-Nibasheka M, Nathanielsz PW 1998 In vivo administra-tion of nimesulide, a selective PGHS-2 inhibitor, increases in vitromyometrial sensitivity to prostaglandins while lowering sensitivityto oxytocin. J Soc Gynecol Invest 5:296–299

314. Poore KR, Young IR, Hirst JJ 1999 Efficacy of the selective pros-taglandin synthase type 2 inhibitor nimesulide in blocking basalprostaglandin production and delaying glucocorticoid-inducedpremature laborin sheep. Am J Obstet Gynecol 180:1244–1253

315. Sawdy RJ, Slater DM, Fisk N, Edmonds DK, Bennett PR 1997 Useof a cyclo-oxygenase type-2 selective non-steroidal anti-inflamma-tory agent to prevent preterm delivery. Lancet 350:265–266

316. Needleman P, Turk J, Jakschik BA, Morrison AR, Lefkowith JB1986 Arachidonic acid metabolism. Annu Rev Biochem 55:69–102

317. Mitchell MD, Romero R, Edwin SS, Trautman MS 1995 Prosta-glandins and parturition. Reprod Fertil Dev 7:623–632

318. Ensor CM, Yang J-Y, Okita RT, Tai H-H 1990 Cloning and se-quence analysis of the cDNA for human placental NAD1-depen-dent 15-hydroxyprostaglandin dehydrogenase. J Biol Chem 265:14888–14891

319. Xun CQ, Ensor CM, Tai H-H 1991 Regulation of synthesis andactivity of NAD1-dependent 15-hydroxyprostaglandin dehydro-genase (15-PGDH) by dexamethasone and phorbol ester in humanerythroleukemia (HEL) cells. Biochem Biophy Res Commun 177:1258–1265

320. Challis JRG, Patel FA, Pomini F 1999 Prostaglandin dehydroge-nase and the initiation of labor. J Perinat Med 27:26–34

321. Senior J, Sangha RK, Baxter GS, Marshall K, Clayton JK 1992 Invitro characterization of prostanoid FP-, DP-, IP- and TP-receptorson the non-pregnant human myometrium. Br J Pharmacol 107:215–221

322. Schaefers H-J, Goppelt-Struebe M 1996 Interference of cortico-steroids with prostaglandin E2 synthesis at the level of cyclooxy-genase-2 mRNA expression in kidney cells. Biochem Pharmacol52:1415–1421

323. Goppelt-Struebe M 1997 Molecular mechanisms involved in theregulation of prostaglandin biosynthesis by glucocorticoids. Bio-chem Pharmacol 53:1389–1395

324. Wang L-H, Hajibeigi A, Xu X-M, Loose-Mitchell D, Wu KK 1993Characterization of the promoter of human prostaglandin H syn-thase-1 gene. Biochem Biophy Res Commun 190:406–411

325. Inoue H, Kosaka T, Miyata A, Hara S, Yokoyama C, NanayamaT, Tanabe T 1995 Structure and expression of the human prosta-glandin endoperoxide synthase 2 gene. In: Samuelsson B, et al. (eds)Advances in Prostaglandin, Thromboxane and Leukotriene Re-search. Raven Press, New York, pp 109–111

326. Tazawa R, Xu X-M, Wu KK, Wang L-H 1994 Characterization ofthe genomic structure, chromosomal location and promoter of hu-man prostaglandin H synthase-2 gene. Biochem Biophy Res Com-mun 203:190–199

327. Kniss DA 1999 Cyclooxygenases in reproductive medicine andbiology. J Soc Gynecol Invest 6:285–292

328. Dudley DJ, Collmer D, Mitchell MD, Trautman MS 1996 Inflam-matory cytokine mRNA in human gestational tissues: implicationsfor term and preterm labor. J Soc Gynecol Invest 3:328–335

329. Dudley DJ, Trautman MS, Mitchell MD 1993 Inflammatory me-diators regulate interleukin-8 production by cultured gestationaltissues: evidence for a cytokine network at the chorio-decidualinterface. J Clin Endocrinol Metab 76:404–410

330. McKay LI, Cidlowski JA 1999 Molecular control of immune/inflammatory responses: interactions between nuclear factor-kBand steroid receptor-signaling pathways. Endocr Rev 20:435–459

331. Economopoulos P, Sun M, Purgina B, Gibb W 1996 Glucocorti-coids stimulate prostaglandin H synthase type-2 (PGHS-2) in thefibroblast cells in human amnion cultures. Mol Cell Endocrinol117:141–147

332. Zakar T, Olson DM 1989 Dexamethasone stimulates arachidonicacid conversion to prostaglandin E2 in human amnion cells. J DevPhysiol 12:269–272

333. Zakar T, Olson DM 1995 Studies on glucocorticoid hormone ac-tions in the regulation of human amnion PGHS. Reprod Fertil Dev7:517–520

334. Zakar T, Hirst JJ, Mijovic JE, Olson DM 1995 Glucocorticoidsstimulate the expression of prostaglandin endoperoxide H syn-thase-2 in amnion cells. Endocrinology 136:1610–1619

335. Keirse MJNC, Turnbull AC 1976 The fetal membranes as a pos-sible source of amniotic fluid prostaglandins. Br J Obstet Gynaecol83:146–151

336. Keirse MJNC 1990 Eicosanoids in human pregnancy and partu-rition. In: Mitchell MD (ed) Eicosanoids in Reproduction. CRCPress, Boca Raton, FL, pp 199–222

337. Olson DM, Skinner K, Challis JRG 1983 Prostaglandin output inrelation to parturition by cells dispersed from human intrauterinetissues. J Clin Endocrinol Metab 57:694–699

338. Hirst JJ, Teixeira FJ, Zakar T, Olson DM 1995 Prostaglandin en-doperoxide-H synthase-1 and -2 messenger ribonucleic acid levels

546 CHALLIS ET AL. Vol. 21, No. 5

Page 34: PDF (454 KB)

in human amnion with spontaneous labor onset. J Clin EndocrinolMetab 80:517–523

339. Hirst JJ, Teixeira FJ, Zakar T, Olson DM 1995 Prostaglandin Hsynthase-2 expression increases in human gestational tissues withspontaneous labour onset. Reprod Fertil Dev 7:633–637

340. Lopez-Bernal A, Hansell DJ, Khong TY, Keeling JW, TurnbullAC 1989 Prostaglandin E production by the fetal membranes inunexplained preterm labour and preterm labour associated withchorioamnionitis. Br J Obstet Gynaecol 96:1133–1139

341. Lopez-Bernal A, Hansell DJ, Alexander S, Turnbull AC 1987Prostaglandin E production by amniotic cells in relation to term andpreterm labour. Br J Obstet Gynaecol 94:864–869

342. Slater DM, Berger L, Newton R, Moore G, Bennett PR 1994 Therelative abundance of type 1 and type 2 cyclo-oxygenase mRNA inhuman amnion at term. Biochem Biophy Res Commun 193:304–308

343. Teixeira FJ, Zakar T, Hirst JJ, Guo F, Machin G, Olson DM 1993Prostaglandin endoperoxide H synthase (PGHS) activity increaseswith gestation and labour in human amnion. J Lipid Mediat 6:515–523

344. Gibb W, Sun M 1996 Localization of prostaglandin H synthasetype 2 protein and mRNA in term human fetal membranes anddecidua. J Endocrinol 150:497–503

345. Price TM, Kauma SW, Curry Jr TE, Clark MR 1989 Immunohis-tochemical localization of prostaglandin endoperoxide synthase inhuman fetal membranes and decidua. Biol Reprod 41:701–705

346. Slater DM, Berger L, Newton R, Moore G, Bennet PR 1995 Ex-pression of cyclooxygenase Types 1 and 2 in human fetal mem-branes at term. Am J Obstet Gynecol 172:77–82

347. Gibb W, Riopel L, Collu R, Ducharme JR, Mitchell MD, LavoieJC 1988 Cyclooxygenase products formed by primary cultures ofcells from human chorion laeve: influence of steroids. Can J PhysiolPharmacol 66:788–793

348. Casey ML, MacDonald PC 1988 Decidual activation: the role ofprostaglandins in labor. In McNellis D, MacDonald PC, ChallisJRG, Nathanielsz PW, Roberts JM (eds) The Onset of Labor: Cellularand Integrative Mechanisms. Perinatalogy Press, Ithaca, NY, p141–156

349. Teixeira FJ, Zakar T, Hirst JJ, Guo F, Sadowsky DW, Machin G,Demianczuk N, Resch B, Olson DM 1994 Prostaglandin endo-peroxide-H synthase (PGHS) activity and immunoreactive PGHS-1and PGHS-2 levels in human amnion throughout gestation, at termand during labor. J Clin Endocrinol Metab 78:1396–1402

350. Mitchell MD, Branch DW, Lundin-Schiller S, Romero R, DaynesRA, Dudley DJ 1991 Immunologic aspects of preterm labor. SeminPerinatol 15:210–224

351. Sullivan MH, Roseblade CK, Elder MG 1991 Metabolism of pros-taglandin E2 on the fetal and maternal sides of intact fetal mem-branes. Acta Obstet Gynecol Scand 70:425–427

352. Mijovic JE, Zakar T, Nairn TK, Olson DM 1998 Prostaglandinendoperoxide H synthase (PGHS) activity and PGHS-1 and -2ribonucleic acid abundance in human chorion throughout gesta-tion and with preterm labor. J Clin Endocrinol Metab 83:1358–1367

353. Mijovic JE, Zakar T, Nairn TK, Olson DM 1997 Prostaglandinendoperoxide-H synthase-2 expression and activity increases withterm labour in the human chorion. Am J Physiol 272:E832–E840

354. Okazaki T, Casey ML, Okita JR, MacDonald PC, Johnston JM1981 Initiation of parturition. XII. Biosynthesis and metabolism ofprostaglandins in human fetal membranes and uterine decidua.Am J Obstet Gynecol 139:373–381

355. Nakla S, Skinner K, Mitchell BF, Challis JRG 1986 Changes inprostaglandin transfer across human fetal membranes obtainedafter spontaneous labor. Am J Obstet Gynecol 155:1337–1341

356. Cheung PYC, Challis JRG 1989 Prostaglandin E2 metabolism in thehuman fetal membranes. Am J Obstet Gynecol 161:1580–1585

357. van Meir CA, Ramirez MM, Matthews SG, Calder AA, KeirseMJNC, Challis JRG 1997 Chorionic prostaglandin catabolism isdecreased in the lower uterine segment with term labor. Placenta18:109–114

358. Molnar M, Hertelendy F 1990 PGF2a and PGE2 binding to ratmyometrium during gestation, parturition, and postpartum. Am JPhysiol 258:E740–E747

359. Hayflick L 1961 The establishment of a line (WISH) of humanamnion cells in continuous cultivation. Exp Cell Res 23:14–20

360. Hansen WR, Sato T, Mitchell MD 1998 Tumour necrosis factor-alpha stimulates increased expression of prostaglandin endoper-oxide H synthast type 2 mRNA in amnion-derived WISH cells. JMol Endocrinol 20:221–231

361. Hulkower KI, Otis ER, Li J, Ennis BW, Cugier DJ, Bell RL, CarterGW, Glaser KB 1997 Induction of prostaglandin H synthase-2 andtumor necrosis factor a in human amnionic WISH cells by variousstimuli occurs through distinct intracellular mechanisms. J Phar-macol Exp Ther 280:1065–1074

362. Casey ML, Cox SM, Word RA, MacDonald PC 1990 Cytokines andinfection-induced preterm labor. Reprod Fertil Dev 2:499–509

363. Denison FC, Kelly RW, Calder AA, Riley SC 1998 Cytokine se-cretion by human fetal membranes, decidua and placenta at term.Hum Reprod 13:3560–3565

364. Habenicht AJR, Goerig M, Grulich J, Rothe D, Gronwald R, LothU, Schettler G, Kommerell B, Ross R 1985 Human platelet-drivengrowth factor stimulates prostaglandin synthesis by activation andby rapid de novo synthesis of cyclooxygenase. J Clin Invest 75:1381–1387

365. Mitchell MD, Edwin SS, Lundin-Schiller S, Silver RM, SmotkinD, Trautman MS 1993 Mechanism of interleukin-1b stimulation ofhuman amnion prostaglandin biosynthesis: mediation via a novelinducible cyclooxygenase. Placenta 14:615–625

366. Dudley DJ, Trautman MS, Araneo BA, Edwin SS, Mitchell MD1992 Decidual cell biosynthesis of interleukin-6: regulation by in-flammatory cytokines. J Clin Endocrinol Metab 74:884–889

367. Dudley DJ, Trautman MS, Edwin SS, Lundin-Schiller S, MitchellMD 1992 Biosynthesis of interleukin-6 by cultured human chorionlaeve cells: regulation by cytokines. J Clin Endocrinol Metab 75:1081–1086

368. Jones CA, Finlay-Jones JF, Hart PH 1997 Type-1 and type-2 cy-tokines in human late-gestation decidual tissue. Biol Reprod 57:303–311

369. Kniss DA, Zimmerman PD, Garver CL, Fertel RH 1997 Interleu-kin-1 receptor antagonist blocks interleukin-1-induced expressionof cyclooxygenase-2 in endometrium. Am J Obstet Gynecol 177:559–567

370. Adcock IM, Newton R, Barnes PJ 1997 NF-kB involvement inIL-1b-induction of GM-CSF and COX-2: inhibition by glucocorti-coids does not require 1-kB. Biochem Soc Trans 25:154S

371. Albert TJ, Su HC, Zimmerman PD, Iams JD, Kniss DA 1994Interleukin-1b regulates the inducible cyclooxygenase in amnion-derived WISH cells. Prostaglandins 48:401–416

372. Kniss DA, Iams JD 1998 Regulation of parturition update, Endo-crine and panacrine effectors of term and preterm labor. Clin Peri-natol 25:819–836

373. Belt AR, Baldassare JJ, Molnar M, Romero R, Hertelendy F 1999The nuclear transcription factor NF-kB mediates interleukin-1b-induced expression of cyclooxygenase-2 in human myometrialcells. Am J Obstet Gynecol 181:359–366

374. Wang Z, Tai H-H 1998 Interleukin-1b and dexamethasone regulategene expression of prostaglandin H synthase-2 via the NF-kB path-way in human amnion derived WISH cell. Prostaglandins LeukotEssent Fatty Acids 59:63–69

375. Whittle WL, Gibb W, Challis JRG 2000 The characterization ofhuman amnion epithelial and mesenchymal cell culture; the cel-lular expression activity and glucocorticoid regulation of prosta-glandin synthesis. Placenta 21:894–401

376. Gibb W, Lavoie JC 1990 Effects of glucocorticoids on prostaglandinformation by human amnion. Can J Physiol Pharmacol 68:671–676

377. Newman SP, Flower RJ, Croxtall JD 1994 Dexamethasone sup-pression of IL-1b-induced cyclooxygenase 2 expression is not me-diated by lipocortin-1 in A549 cells. Biochem Biophy Res Commun202:931–939

378. DeWitt DL, Meade EA 1993 Serum and glucocorticoid regulationof gene transcription and expression of the prostaglandin H syn-thase-1 and prostaglandin H synthase-2 isozymes. Arch BiochemBiophys 306:94–102

379. Samet JM, Fasano MB, Fonteh AN, Chilton FH 1995 Selectiveinduction of prostaglandin G/H synthase I by stem cell factor anddexamethasone in mast cells. J Biol Chem 270:8044–8049

380. Potestio FA, Zakar T, Olson DM 1988 Glucocorticoids stimulate

October, 2000 PARTURITION 547

Page 35: PDF (454 KB)

prostaglandin synthesis in human amnion cells by a receptor-mediated mechanism. J Clin Endocrinol Metab 67:1205–1210

381. Sun M, Ramirez MM, Challis JRG, Gibb W 1996 Immunohisto-chemical localization of glucocorticoid receptor in human fetalmembranes and decidua at term and preterm delivery. J Endocrinol149:243–248

382. Sawdy RJ, Dennes JB, Allport V, Slater DM, Elder MG, SullivanMH, Bennett PR 1999 Region and labour-dependent synthesis ofprostaglandin E2 by human fetal membranes. Placenta 20:181–184

383. Olson DM, Opavsky MA, Challis JRG 1983 Prostaglandin syn-thesis by human amnion is dependent upon extracellular calcium.Can J Physiol Pharmacol 61:1089–1092

384. Olson DM, Skinner K, Challis JRG 1983 Estradiol-17b and 2-hydroxyestradiol-17b-induced differential production of prosta-glandins by cells dispersed from human intrauterine tissues atparturition. Prostaglandins 25:639–651

385. Sander J, Myatt L 1990 Regulation of prostaglandin E2 synthesis inhuman amnion by protein kinase C. Prostaglandins 39:355–363

386. Schatz F, Gurpide E 1983 Effects of estradiol on prostaglandin F2a

levels in primary monolayer cultures of epithelial cells from humanproliferative endometrium. Endocrinology 113:1274–1279

387. Mitchell MD, MacDonald PC, Casey ML 1984 Stimulation ofprostaglandin E2 synthesis in human amnion cells maintained inmonolayer culture by a substance(s) in amniotic fluid. Prostaglan-dins Leukot Med 15:399–407

388. Casey ML, Mitchell MD, MacDonald PC 1987 Epidermal growthfactor-stimulated prostaglandin E2 production in human amnioncells: specificity and nonesterified arachidonic acid dependency.Mol Cell Endocrinol 53:169–176

389. Warrick C, Skinner K, Mitchell BF, Challis JRG 1985 Relationbetween cyclic adenosine monophosphate and prostaglandin out-put by dispersed cells from human amnion and decidua. Am JObstet Gynecol 153:66–71

390. Divers Jr WA, Wilkes MM, Babaknia A, Yen SSC 1981 An increasein catecholamines and metabolites in the amniotic fluid compart-ment from middle to late gestation. Am J Obstet Gynecol 139:483–486

391. DiRenzo GC, Venincasa MD, Bleasdale JE 1984 The identificationand characterization of beta-adrenergic receptors in human amniontissue. Am J Obstet Gynecol 148:398–405

392. Casper RF, Lye SJ 1986 Myometrial desensitization to continuousbut not to intermittent b-adrenergic agonist infusion in the sheep.Am J Obstet Gynecol 154:301–305

393. Schlegel W, Demers LM, Hildebrandt-Stark HE, Behrman HR,Greep RO 1974 Partial purification of human placental 15-hydroxyprostaglandin dehydrogenase: kinetic properties. Prosta-glandins 5:417–433

394. Cheung PYC, Walton JC, Tai H-H, Riley SC, Challis JRG 1990Immunocytochemical distribution and localization of 15-hydroxyprostaglandin dehydrogenase in human fetal membranes,decidua, and placenta. Am J Obstet Gynecol 163:1445–1449

395. Cheung PYC, Walton JC, Tai H-H, Riley SC, Challis JRG 1992Localization of 15-hydroxyprostaglandin dehydrogenase in hu-man fetal membranes, decidua, and placenta during pregnancy.Gynecol Obstet Invest 33:142–146

396. Sangha RK, Walton JC, Ensor CM, Tai H-H, Challis JRG 1994Immunohistochemical localization, mRNA abundance and activityof 15-hydroxyprostaglandin dehydrogenase in placenta and fetalmembranes during term and preterm labor. J Clin EndocrinolMetab 78:982–989

397. van Meir CA, Sangha RK, Walton JC, Matthews SG, KeirseMJNC, Challis JRG 1996 Immunoreactive 15-hydroxyprostaglan-din-dehydrogenase (PGDH) is reduced in fetal membranes frompatients at preterm delivery in the presence of infection. Placenta17:291–297

398. Patel FA, Clifton VL, Chwalisz K, Challis JRG 1999 Steroid reg-ulation of prostaglandin dehydrogenase activity and expression inhuman term placenta and chorio-decidua in relation to labor. J ClinEndocrinol Metab 84:291–299

399. Patel FA, Sun K, Challis JRG 1998 Involvement of 11-hydroxy-steroid dehydrogenase in the regulation of prostaglandin dehy-drogenase activity by cortisol/cortisone in human term placentaand fetal membranes. J Soc Gynecol Invest 5 [Suppl] (Abstract T626)

400. Gibb W, Lavoie J-C, Roux JF 1978 3b-Hydroxysteroid dehydro-genase activity in human fetal membranes. Steroids 32:365–372

401. Grimshaw R, Mitchell BF, Challis JRG 1983 Steroid modulationof pregnenolone to progesterone conversion by human placentalcells in vitro. Am J Obstet Gynecol 145:234–238

402. Cheng L, Kelly RW, Thong KJ, Hume R, Baird DT 1993 The effectsof mifepristone (RU486) on prostaglandin dehydrogenase in de-cidual and chorionic tissue in early pregnancy. Hum Reprod 8:705–709

403. Patel FA, Chwalisz K, Challis JRG, Regulation of prostaglandindehydrogenase (PGDH) activity by cortisol and progesterone mayinvolve paracrine/autocrine interaction and effects on levels ofPGDH mRNA. Program of the 45th Annual Meeting of the Societyfor Gynecologic Investigation, 1998 (Abstract 136)

404. Karalis K, Majzoub JA 1995 Regulation of placental corticotro-phin-releasing hormone by steroids - possible implication in laborinitiation. Ann NY Acad Sci551–555

405. Lennon C, Carlson MG, Nelson DM, Sadovsky Y 1999 In vitromodulation of the expression of 15-hydroxy-prostaglandin dehy-drogenase by trophoblast differentiation. Am J Obstet Gynecol180:690–695

406. Sun K, Yang K, Challis JRG 1997 Differential regulation of 11b-hydroxysteroid dehydrogenase type 1 and 2 by nitric oxide incultured human placental trophoblast and chorionic cell prepara-tion. Endocrinology 138:4912–4920

407. Bedwani JR, Marley PB 1975 Enhanced inactivation of prostaglan-din E2 by the rabbit lung during pregnancy or progesterone treat-ment. Br J Pharmacol 53:547–554

408. Myatt L, Jogee M, Elder MG 1983 Regulation of prostacyclin me-tabolism in human placental cells in culture by steroid hormones.In: Lewis PJ, Moncada S, O’Grady J (eds) Prostacyclin in Pregnancy.Raven Press, New York, pp 119–129

409. Keelan JA, Goodwin V, Mitchell MD 1998 Inhibition of 15-hy-droxysteroid dehydrogenase expression and activity by cytokinesin human placental trophoblasts. J Soc Gynecol Invest 5 [Suppl](Abstract 39)

410. Pomini F, Caruso A, Challis JRG 1999 Interleukin-10 modifies theeffect of interleukin 1-b and tumor necrosis factor a on the activityand expression of prostaglandin H synthase-2 and the NAD1-dependent 15-hydroxyprostaglandin dehydrogenase in culturedterm human villous trophoblast and chorion trophoblast cells.J Clin Endocrinol Metab 84:4645–4651

411. Romero R, Quintero R, Emamian M, Wan M, Grzyboski C, Hob-bins JC, Mitchell MD 1987 Arachidonate lipoxygenase metabolitesin amniotic fluid of women with intra-amniotic infection and pre-term labor. Am J Obstet Gynecol 157:1454–1460

412. Romero R, Wu YK, Mazor M, Hobbins JC, Mitchell MD 1988Amniotic fluid prostaglandin E2 in preterm labor. ProstaglandinsLeukot Essent Fatty Acids 34:141–145

413. Romero R, Hobbins JC, Mitchell MD 1988 Endotoxin stimulatesprostaglandin E2 production by human amnion. Obstet Gynecol71:227–228

414. Bennett PR, Rose MP, Myatt L, Elder MG 1987 Preterm labor:stimulation of arachidonic acid metabolism in human amnion cellsby bacterial products. Am J Obstet Gynecol 156:649–655

415. Lamont RF, Anthony F, Myatt L, Booth L, Furr PM, Taylor-Robinson Dl 1990 Production of prostaglandin E2 by human am-nion in vitro in response to additon of media conditioned by mi-croorganisms associated with chorioamnionitis and preterm labor.Am J Obstet Gynecol 162:819–825

416. Romero R, Mazor M, Wu YK, Avila C, Oyarzun E, Mitchell MD1989 Bacterial endotoxin and tumor necrosis factor stimulate pros-taglandin production by human decidua. Prostaglandins LeukotEssent Fatty Acids 37:183–186

417. Romero R, Brody DT, Oyarzun E, Mazor M, WuYK, Hobbins JC,Durum SK 1989 Infection and labor. III. Interleukin-1: a signal forthe onset of parturition. Am J Obstet Gynecol 160:1117–1123

418. Silver RM, Edwin SS, Trautman MS, Simmons DL, Branch DW,Dudley DJ, Mitchell MD 1995 Bacterial lipopolysaccharide-mediated fetal death. Production of a newly recognized form ofinducible cyclooxygenase (COX-2) in murine decidua in responseto lipopolysaccharide. J Clin Invest 95:725–731

419. Romero R, Avila C, Brekus CA, Morotti R 1991 The role of sys-

548 CHALLIS ET AL. Vol. 21, No. 5

Page 36: PDF (454 KB)

temic and intrauterine infection in preterm parturition. Ann NYAcad Sci 622:355–375

420. Romero R, Mazor M, Tartakovsky B 1991 Systemic administrationof interleukin-1 induces preterm parturition in mice. Am J ObstetGynecol 165:969–971

421. Romero R, Wu YK, Sirtori M 1989 Amniotic fluid concentrationsof prostaglandin F2a, 13,14-dihydro-15-keto-prostaglandin F2a

(PGFM) and 11-deoxy-13,14-dihydro-15-keto-11, 16-cyclo-prosta-glandin E2 (PGEM-LL) in preterm labor. Prostaglandins 37:149–161

422. Romero R, Manogue KR, Mitchell MD, Wu YK, Oyarzun E,Hobbins JC, Cerami A 1989 Infection and labor. IV. Cachectin-tumor necrosis factor in the amniotic fluid of women with intra-amniotic infection and preterm labor. Am J Obstet Gynecol 161:336–341

423. Romero R, Avila C, Santhanam U, Sehgal PB 1990 Amniotic fluidinterleukin 6 in preterm labor. J Clin Invest 85:1392–1400

424. Romero R, Ceska M, Avila C, Mazor M, Behnke E, Lindley I 1991Neutrophil attractant/activating peptide-1/interleukin-8 in termand preterm parturition. Am J Obstet Gynecol 165:813–820

425. Keelan JA, Sato T, Mitchell MD 1997 Interleukin (IL)-6 and IL-8production by human amnion: regulation by cytokines, growthfactors, glucocorticoids, phorbol esters, and bacterial lipopolysac-charide. Biol Reprod 57:1438–1444

426. Petraglia F, Sutton S, Vale W 1989 Neurotransmitters and peptidesmodulate the release of immunoreactive corticotropin-releasingfactor from cultured human placental cells. Am J Obstet Gynecol160:247–251

427. Petraglia F, Florio P, Nappi C, Genazzani AR 1996 Peptide sig-naling in human placenta and membranes: autocrine, paracrineand endocrine mechanisms. Endocr Rev 17:156–186

428. MacDonald PC, Casey ML 1993 The accumulation of prostaglan-dins (PG) in amniotic fluid is an after-effect of labor and not in-dicative of a role for PGE2 or PGF2a in the initiation of humanparturition. J Clin Endocrinol Metab 76:1332–1339

429. Challis JRG, Matthews SG, van Meir CA, Ramirez MM 1995Current topic: the placental corticotrophin-releasing hormone-adrenocorticotrophin axis. Placenta 16:481–502

430. Riley SC, Challis JRG 1991 Corticotorphin-releasing hormone pro-duction by the placenta and fetal membranes. Placenta 12:105–119

431. Grino M, Chrousos GP, Margioris AN 1987 The corticotropinreleasing hormone gene is expressed in human placenta. BiochemBiophys Res Commun 148:1208–1214

432. Goland RS, Wardlaw SL, Stark RI, Brown Jr LS, Frantz AG 1986High levels of corticotropin-releasing hormone immunoactvitiy inmaternal and fetal plasma during pregnancy. J Clin EndocrinolMetab 63:1199–1203

433. Campbell EA, Linton EA, Wolfe CDA, Scraggs PR, Jones MT,Lowry PJ 1987 Plasma corticotropin-releasing hormone concen-trations during pregnancy and parturition. J Clin Endocrinol Metab64:1054–1059

434. Goland RS, Wardlaw SL, Blum M, Tropper PJ, Stark RI 1988Biologically active corticotropin-releasing hormone in maternaland fetal plasma during pregnancy. Am J Obstet Gynecol 159:884–890

435. Okamoto E, Takagi T, Makino T, Sata H, Iwata I, Nishino E,Mitsuda N, Sugita N, Otsuki Y, Tanizawa O 1989 Immunoreactivecorticotropin-releasing hormone, adrenocorticotorpin and cortisolin human plasma during pregnancy and delivery and postpartum.Horm Metab Res 21:566–572

436. Goland RS, Jozak S, Warren WB, Conwell IM, Stark RI, TropperPJ 1993 Elevated levels of umbilical cord plasma corticotropin-releasing hormone in growth-retarded fetuses. J Clin EndocrinolMetab 77:1174–1179

437. Wolfe CD, Patel SP, Campbell EA, Linton EA, Anderson J, LowryPJ, Jones MT 1988 Plasma corticotrophin-releasing factor (CRF) innormal pregnancy. Br J Obstet Gynaecol 95:997–1002

438. Laatikainen TJ, Raisanen UJ, Salminen KR 1988 Corticotropin-releasing hormone in amniotic fluid during gestation and labor andin relation to fetal lung maturation. Am J Obstet Gynecol 159:891–895

439. Sasaki A, Shinkawa O, Margioris AN, Liotta AS, Sato S, Mu-rakami D, Go M, Shimizu Y, Hanew K, Yoshinaga K 1987 Im-

munoreactive corticotropin-releasing hormone in human plasmaduring pregnancy, labor and delivery. J Clin Endocrinol Metab64:224–229

440. Warren WB, Patrick SL, Goland RS 1992 Elevated maternalplasma corticotropin-releasing hormone levels in pregnancies com-plicated by preterm labor. Am J Obstet Gynecol 166:1198–1204

441. Korebrits C, Ramirez MM, Watson L, Brinkman E, Bocking AD,Challis JRG 1998 Maternal corticotropin-releasing hormone is in-creased with impending preterm birth. J Clin Endocrinol Metab83:1585–1591

442. Potter E, Behan DP, Fischer WH, Linton EA, Lowry PJ, Vale WW1991 Cloning and characterization of the cDNAs for human and ratcorticotropin releasing factor-binding proteins. Nature 349:423–426

443. Linton EA, Perkins AV, Woods RJ, Eben F, Wolfe CD, Behan DP,Potter E, Vale WW, Lowry PJ 1993 Corticotropin releasing hor-mone-binding protein (CRH-BP): plasma levels decrease duringthe third trimester of normal human pregnancy. J Clin EndocrinolMetab 76:260–262

444. Jones SA, Brooks AN, Challis JRG 1989 Steroids modulate cor-ticotropin-releasing hormone production in human fetal mem-branes and placenta. J Clin Endocrinol Metab 68:825–830

445. Riley SC, Walton JC, Herlick JM, Challis JRG 1991 The localiza-tion and distribution of corticotropin-releasing hormone in thehuman placenta and fetal membranes throughout gestation. J ClinEndocrinol Metab 72:1001–1007

446. Frim DM, Emanuel RL, Robinson BG, Smas CM, Adler GK,Majzoub JA 1988 Characterization and gestational regulation ofcorticotropin-releasing hormone messenger RNA in human pla-centa. J Clin Invest 82:287–292

447. Robinson BG, Emanuel RL, Frim DM, Majzoub JA 1988 Glu-cocorticoid stimulates expression of corticotropin-releasing hor-mone gene in human placenta. Proc Natl Acad Sci USA 85:5244–5248

448. Marinoni E, Korebrits C, Di Iorio R, Cosmi EV, Challis JRG 1998Effect of betamethasone in vivo on placental corticotropin-releasinghormone in human pregnancy. Am J Obstet Gynecol 178:770–778

449. Elliott JP, Radin TG 1995 The effect of corticosteroid administra-tion on uterine activity and preterm labor in high-order multiplegestations. Obstet Gynecol 85:250–254

450. Yeshaya A, Orvieto R, Ben-Shem E, Dekel A, Peleg D, Dicker D,Ben-Rafael Z 1996 Uterine activity after betamethasone adminis-tration for the enhancement of fetal lung maturation. Eur J ObstetGynecol Reprod Biol 67:139–141

451. Jones SA, Challis JRG 1990 Steroid, corticotrophin-releasing hor-mone, ACTH and prostaglandin interactions in the amnion andplacenta of early pregnancy in man. J Endocrinol 125:153–159

452. Nodwell A, Carmichael L, Fraser M, Challis JRG, Richardson B1999 Placental release of corticotrophin-releasing hormone acrossthe umbilical circulation of the human newborn. Placenta 20:197–202

453. Clifton VL, Read MA, Leitch IM, Boura AL, Robinson PJ, SmithR 1994 Corticotropin-releasing hormone-induced vasodilatation inthe human fetal placental circulation. J Clin Endocrinol Metab79:666–669

454. Hobel CJ, Dunkel-Schetter C, Roesch SC, Castro LC, Arora CP1999 Maternal plasma corticotropin-releasing hormone associatedwith stress at 20 weeks’ gestation in pregnancies ending in pretermdelivery. Am J Obstet Gynecol 180:S257–S263

455. Grammatopoulos D, Thompson S, Hillhouse EW 1995 The humanmyometrium expresses multiple isoforms of the corticotropin-releasing hormone receptor. J Clin Endocrinol Metab 80:2388–2393

456. Grammatopoulos DK, Dai Y, Randeva HS, Levine MA, KarterisE, Easton AJ, Hillhouse EW 1999 A novel spliced variant of thetype 1 corticotropin-releasing hormone receptor with a deletion inthe seventh transmembrane domain present in the human preg-nant term myometrium and fetal membranes. Mol Endocrinol 13:2189–2202

457. Karteris E, Grammatopoulos D, Dai Y, Olah KB, Ghobara TB,Easton AJ, Hillhouse EW 1998 The human placenta and fetalmembranes express the corticotropin-releasing hormone receptor1a (CRH-1a) and the CRH-C variant receptor. J Clin EndocrinolMetab 83:1376–1379

October, 2000 PARTURITION 549

Page 37: PDF (454 KB)

458. Grammatopoulos DK, Hillhouse EW 1999 Basal and interleukin-1b-stimulated prostaglandin production from cultured humanmyometrial cells: differential regulation by corticotropin-releasinghormone. J Clin Endocrinol Metab 84:2204–2211

459. Grammatopoulos D, Stirrat GM, Williams SA, Hillhouse EW1996 The biological activity of the corticotropin-releasing hormonereceptor-adenylate cyclase complex in human myometrium is re-duced at the end of pregnancy. J Clin Endocrinol Metab 81:745–751

460. Grammatopoulos DK, Hillhouse EW 1999 Activation of proteinkinase C by oxytocin inhibits the biological activity of the humanmyometrial corticotropin-releasing hormone receptor at term. En-docrinology 140:585–594

461. Smith R 1999 The timing of birth. Sci Am 280:68–75462. McLean M, Bisits A, Davies J, Woods R, Lowry P, Smith R 1995

A placental clock controlling the length of human pregnancy. NatMed 1:460–463

463. Quartero HW, Srivatsa G, Gillham B 1992 Role for cyclic aden-osine monophosphate in the synergistic interaction between oxy-

tocin and corticotrophin-releasing factor in isolated human gesta-tional myometrium. Clin Endocrinol (Oxf) 36:141–145

464. Keirse MJNC 1995 b-Mimetic tocolysis in preterm labour. In: EnkinMW, Renfrew MJ (eds) Pregnancy and Childbirth Module. Coch-rane Database of Systemic Reviews, Cochrane Updates on disc

465. Lye SJ, Dayes BA, Freitag CL, Brooks J, Casper RF 1992 Failureof ritodrine to prevent preterm labor in the sheep. Am J ObstetGynecol 167:1399–1408

466. DeWitt DL 1999 Cox-2-selective inhibitors: the new super aspirins.Mol Pharmacol 55:625–631

467. Orchinik M 1998 Glucocorticoids, stress, and behavior: shifting thetimeframe. Horm Behav 34:320–327

468. Jobe AH, Wada N, Berry LM, Ikegami M, Ervin MG 1998 Singleand repetitive maternal glucocorticoid exposures reduce fetalgrowth in sheep. Am J Obstet Gynecol 178:880–885

469. Challis JRG, Cox DB, Sloboda DM 2000 Regulation of cortico-steroids in the fetus: control of birth and influence on adult disease.Semin Neonatol 4:93–97

Erratum

Figure 1 in the June 2000 Endocrine Reviews article by P. C. White and P. W. Speiser, “Congenital adrenalhyperplasia due to 21-hydroxylase deficiency” (Endocrine Reviews 2000, 21:245–291) contained errors thathave been corrected in the following figure:

FIG. 1. Pathways of steroid biosynthesis. The pathways for synthesis of progesterone and mineralocorticoids (aldosterone), glucocorticoids(cortisol), androgens (testosterone and dihydrotestosterone), and estrogens (estradiol) are arranged from left to right. The enzymatic activitiescatalyzing each bioconversion are written in boxes. For those activities mediated by specific cytochromes P450, the systematic name of theenzyme (“CYP” followed by a number) is listed in parentheses. CYP11B2 and CYP17 have multiple activities. The planar structures of cholesterol,aldosterone, cortisol, dihydrotestosterone, and estradiol are placed near the corresponding labels.

550 CHALLIS ET AL. Vol. 21, No. 5