You are on page 1of 19

Current Pharmaceutical Design, 2004, 10, 2499-2517 2499

Regulation of Myometrial Smooth Muscle Functions

F. Hertelendy1,* and T. Zakar2

1
Department of Obstetrics, Gynecology and Women’s Health, Saint Louis University School of Medicine, St. Louis,
Missouri, USA and 2Mothers and Babies Research Centre and Department of Obstetrics and Gynaecology, University of
Newcastle, Newcastle, Australia

Abstract: Regulation of myometrial functions during gestation, labor and birth are in the forefront of research in
reproductive sciences. The complexity of the problem is reflected by our scant understanding of the intimate cellular and
molecular events underlying these phenomena, despite extensive efforts spanning several decades. Unlike other smooth
muscles, the myometrium is, to a large extent, under hormonal control. Of these, the steroid hormones, progesterone and
estrogen, play dominant roles in terms of uterine growth, the maintenance of quiescence during gestation and the
preparation of the uterus for labor and delivery. In addition to steroid hormones, there are a number of factors that
modulate myometrial contractility (oxytocin, prostaglandins, endothelin, platelet activating factor) and relaxation
(corticotropin releasing hormone, prostacyclin, nitric oxide). Although notable advances have been made towards
understanding some of the key steps in receptor signaling that define the actions of these factors, a good deal of new
information is needed to fully understand this fundamental life process. Pharmaceutical agents have been used extensively
to induce labor or to prolong pregnancy in the case of preterm labor that represents the major cause of perinatal morbidity
and mortality. Because preterm labor is a syndrome of multiple etiologies, pharmacologic agents will have to be targeted
accordingly. This review attempts to present a critical overview of these topics.

Key Words: myometrium, parturition, preterm labor, progesterone, oxytocin, prostaglandins, cytokines, endothelin, nitric
oxide, CRH, cell signaling, mechanical stretch, tocolysis.

INTRODUCTION leads to increased tension, resulting from the activation of


myosin light chain kinase (MCLK) by the calcium-calmo-
The myometrium, forming the muscular wall of the
dulin complex. MLCK, as in other smooth muscle cells,
uterus, has two principal functions. By growth and stretch, it
phosphorylates the light chain of myosin, allowing it to
provides the protective environment for the developing fetus
interact with actin. The actomyosin complex, in turn, can
and during labor, by powerful contractions, it lends much of
convert the chemical energy of ATP to the mechanical
the mechanical force to expel the products of conception.
energy necessary for myometrial contraction. Calcium may
Although the regulation of neither of these myometrial func- also enter the intracellular compartment by way of receptor
tions is fully understood, there is good evidence that endo- regulated mechanisms. A ligand activated receptor can
crine, paracrine, as well as mechanical factors, are involved.
increase [Ca2+]i by enhancing calcium entry, as well as
The aim of this review is to provide the reader with an
promoting calcium release from intracellular stores. Such a
overview of our current understanding of potential mechan-
mechanism has been amply demonstrated to operate in
isms that maintain the myometrium in a relatively quiescent
myometrial cells. Conversely, G-protein-coupled receptor
state, refractory to various uterotonic substances during
(GPRC) activation may transmit the signal via Gαs,
much of pregnancy, as well as the cellular and molecular
activating the appropriate isoforms of adenylate cyclase
events that may account for the conversion to an active, (AC), causing a rise in cAMP levels.
contractile organ at the onset of labor, responsive to various
uterotonins. Even though the relaxing effect of cAMP on myome-
trium is well established (tocolysis using β2-adrenergic
Although a number of factors are believed to be involved
receptor agonists that activate the AC/cAMP system is based
in the maintenance of quiescence during gestation, there is
general consensus that progesterone, the principal gestational on this principle), the molecular mechanism underpinning
hormone, plays a dominant role in this process and there is this phenomenon is still uncertain. It is known, however, that
cAMP-induced activation of protein kinase A (PKA I and
growing evidence that a functional withdrawal of proges-
PKA II), in association with a family of A-kinase anchor
terone precedes the evolution of myometrial activation
leading to labor. The onset of labor is heralded by phasic proteins [1,2], is involved in the phosphorylation of various
myometrial contractions that are driven by action potentials. target proteins (calcium channels, MLCK, IP3 receptors in
the sarcoplasmic reticulum, etc.) leading to myometrial
The ensuing influx of calcium via voltage-gated channels
smooth muscle relaxation. Elevated levels of cAMP found in
the pregnant myometrium may also be amplified by proges-
*Address Correspondence to that Author at the Department of Obstetrics, terone-inhibited phosphodiesterase activity, suppressing
Gynecology and Women’s Health, Saint Louis University School of cAMP breakdown.
Medicine, St. Louis, Missouri, USA; E-mail: hertelf@slu.edu

1381-6128/04 $45.00+.00 © 2004 Bentham Science Publishers Ltd.


2500 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

The intimate mechanism responsible for the switch from Cells that are targets of progesterone express receptor
a quiescent state to an active, contractile state at the end of proteins (PRs) that bind the hormone with high specificity.
pregnancy, i.e. the mechanism underlying the onset of labor The progesterone-receptor complex then adopts a confor-
in the human, remains largely obscure. However, because the mation that enables it to function as a transcription factor. In
steroid hormones, progesterone and estradiol, directly or this process, the active PR-progesterone complex forms
indirectly regulate the biosynthesis of key proteins involved dimers and binds to palindromic recognition sequences,
in uterine physiology, we shall first address these issues in called PRE (AGAACANNNTGTTCT is the ideal consensus
some detail, followed by the reviewing of a number of sequence), present in the promoter regions of progesterone
factors that modulate myometrial smooth muscle functions responsive genes. This interaction stimulates the transcrip-
during gestation and parturition. Several recent reviews, tion of the target genes. In the absence of progesterone, the
which the reader may wish to peruse [3-5], have addressed receptor is bound to chaperone proteins, predominantly
the regulation of parturition at term and preterm. HSP90, and is inactive as a transcription factor.
The above description, however, is a much simplified
PROGESTERONE
overview of the molecular mechanisms of progesterone
Almost half a century ago Arpad Csapo [6] recognized action. First, two forms of PRs exist, PR-A and PR-B. PR-A
that uterine quiescence during pregnancy results from the is an N-terminally truncated variant of PR-B, and the two
blocking effect of progesterone on myometrial contractility. receptor isoforms can associate to homo- and hetero-dimers.
The progesterone “block” is lifted at term, and the The transcriptional activity of the three receptor dimers is
developing uterine activity soon culminates in the delivery of different and varies in a target gene- and target cell-specific
the fetus. Progesterone has also been implicated in the fashion [13]. Second, PRs undergo multiple phosphorylation
maintenance of the closed cervix, and the removal of the after ligand binding which may modulate their transcrip-
progesterone block is considered critical for the opening of tional activity [14]. Third, PRs bind to promoters as part of a
the birth canal. Consistent with this model, progesterone multiprotein complex. This complex includes the basic
levels in the maternal plasma are high during pregnancy and transcription factors (TBP, polymerase-II, TAFs), coactiva-
fall shortly before labor in many mammalian species. Early tors such as CBP/p300, and the steroid receptor coactivator
withdrawal of progesterone, for example by removing the proteins and also corepressor proteins (reviewed in ref. [15]).
ovaries of pregnant rats or mice, causes preterm delivery, The expression of the cofactors varies in different tissues and
while progesterone administration prolongs gestation beyond physiological states, and their contribution to progesterone-
term. In other species including the human, however, labor dependent regulation may vary according to target genes.
and delivery occur without a fall of circulating progesterone The activity of some cofactors may also be modulated by
levels. Attempts to identify alternative mechanisms, such as protein phosphorylation. Fourth, the PR may interact with
increased target tissue metabolism [7] or the presence of a other transcription factors without binding to the PRE
local progesterone binding substance in the fetal membranes sequence, repressing their activity. This transrepression is
[8], failed to show a drop in tissue progesterone levels before mutual with RelA(p65) [16], while PR represses the activity
the onset of labor. Therefore, progesterone withdrawal of the glucocorticoid and estrogen receptors [13, 17, 18].
before labor in women is believed to be “functional”, Furthermore, PR activity on a PRE-controlled gene may be
involving a mechanism that affects the myometrium in a way inhibited by the AP1 component, c-fos, in a cell line-specific
analogous to a drop in circulating or tissue progesterone fashion [19]. These regulatory interactions have been
levels. The existence of functional progesterone withdrawal identified in cultured cells after co-transfection with reporter
is supported by the observations that administration of the constructs and vectors overexpressing the transcription
progesterone antagonist mifepristone (RU486) increases factors under study. They indicate a great variety of possible
myometrial contractility in early pregnancy [9, 10] and PR modulations at the functional level. Work to translate
promotes cervical dilatation and delivery at term [11, 12]. these basic findings to physiological and pathophysiological
Thus, progesterone is required for the maintenance of situations is still in the initial phase. In the next sections, we
pregnancy and the myometrium is released from the relaxing will review the data relevant to the progesterone dependent
influence of the hormone before labor and delivery occur. regulation of human myometrial function during pregnancy
and labor and will discuss the possible mechanisms of func-
In order to understand the way progesterone suppresses
tional progesterone withdrawal, in view of the information
myometrial contractile activity during most of pregnancy
available on the molecular mechanisms of PR regulation.
and influences the timing of labor in women, two distinct,
but related questions need to be addressed. The first one
Connexin43
relates to the mechanisms by which progesterone suppresses
uterine activity during pregnancy, and the second one The PR is expressed in the pregnant human uterus [20-
addresses the mechanisms that cause functional progesterone 22], therefore progesterone may directly control myometrial
withdrawal before labor. There are no satisfactory answers to function by regulating the expression of progesterone
these questions yet, but some intriguing possibilities are responsive genes. Among these, much attention has been
emerging in view of recent research. devoted to the gene encoding connexin43 (Cx43). The Cx43
protein is a major component of myometrial connexons,
Progesterone Receptor-Mediated Mechanisms which are pore structures between myometrial cells allowing
cell-to-cell communication via diffusible intracellular com-
Progesterone, being a steroid hormone, exerts its effects ponents. Clusters of connexons form gap junctions, which
by influencing the activity of progesterone responsive genes. provide the structural basis of intercellular communication
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2501

necessary for the coordinated, phasic contractions of labor. role of the OTR/oxytocin system in the mechanism of labor
In support of this assumption, the density of myometrial gap onset and maintenance.
junctions has been shown to increase in labor [23-25], and
Cx43 mRNA [26] and protein expression [27] have been The time course of OTR expression in the uterus
found to increase in the uterine muscle in late gestation and corresponds to the pattern of progesterone withdrawal in late
pregnancy [40]. This is best documented in mice [41], where
during labor. Negative regulation of Cx43 gene activity by
progesterone during pregnancy, but not during labor, would failure of parturition in a prostaglandin FP-receptor deficient
strain is due to the lack of luteolysis at the end of the normal
be consistent with functional progesterone withdrawal at the
gestational period. In these animals, high progesterone levels
gene expression level.
are maintained and the OTR is not induced in the uterus at
The effect of progesterone on myometrial cell Cx43 term. Ovariectomy results in a drop of circulating proges-
expression has been studied in cell culture experiments. terone concentrations, induction of OTR and delivery of the
Andersen et al. [28] and Zhao et al. [29] showed that the pups. The oxytocin receptor gene promoter, however, does
synthetic progestin medroxyprogesterone acetate repressed not contain a PRE in mice or any other species studied so
Cx43 gene transcription (measured by nuclear run-on far, including the human [40, 42]. This suggests that the
analysis), mRNA abundance and protein abundance progesterone dependent suppression is either indirect or
(measured by Northern and immunoblot analyses, respec- mediated by a hitherto unrecognized progesterone-sensitive
tively) in confluent primary myocyte cultures from non- regulatory sequence of the OTR gene. In a study where the
pregnant uteri and uterine leiomyomas. At 100 nM progestin effects of sex steroids were examined in human uterine cell
concentration, repression of all three parameters was partial. cultures, Adachi and Oku have indeed found that proges-
Repression by progesterone of Cx43 protein levels and terone suppressed the stimulatory effect of estrogen on OTR
Cx43-gap junction density has also been reported in primary levels measured in a binding assay [43]. On the other hand,
myocyte cultures from term pregnant women [30]. These administration of mifepristone, a progesterone receptor
data strongly suggest that progesterone exerts an inhibitory antagonist, in early human pregnancy has been reported to
effect on Cx43 gene expression in the human myometrium. increase spontaneous uterine contractions without enhancing
The promoter of the human Cx43 gene has been cloned, the sensitivity to oxytocin [9]. In this study, uterine contrac-
sequenced [31] and analyzed functionally by transfection tions were recorded using a pressure transducer inserted into
studies with myometrial cells [29, 31, 32]. The 3.0 kilobase the uterine cavity. Similar results were reported by Webster
upstream sequence of the Cx43 promoter contains a TATA- et al. [44], who monitored intrauterine pressure through a
box sequence, several AP-1, AP-2 and SP-1 regulatory balloon catheter inserted into the uterus of early pregnant
elements, 6 half palindromic estrogen regulatory elements women. The women were treated with epostane, a 3-beta
(EREs) and half palindromic PREs. A reporter construct hyroxysteroid dehydrogenase inhibitor, in a dosage that
containing a thymidine kinase promoter and a PRE upstream reduced maternal serum progesterone concentrations by 77%
of the chloramphenicol acetyltransferase (CAT) gene has and estrogen concentrations by 45% without affecting
been transfected into primary myometrial cells [29]. The cortisol levels. Epostane increased uterine activity, but
construct exhibited progestin-stimulated CAT expression subsequent oxytocin injections showed no increase in
indicating that the cells contained functional PRs. In these oxytocin sensitivity. Selinger et al. conducted a similar study
cells, however, progestin suppressed Cx43 transcription, on mid-trimester pregnancies, also showing no effect of
which argues against the involvement of the half PRE sites epostane treatment on myometrial oxytocin sensitivity [45].
in the regulation. Interaction of the PR with transcription In these women, epostane reduced the levels of progesterone,
factors controlling Cx43 expression via the AP-1 or SP-1 but not of estrogen or cortisol. Thus, there is no consistent
sites is a possibility to be explored, because these sites and clinical or experimental evidence implicating progesterone in
factors are involved in the control of the human myometrial the control of OTR expression or function in the pregnant
Cx43 gene [31-33]. The nature and changes of these human uterus. More data are available on the non-genomic
interactions may be informative regarding the molecular actions of progesterone and its metabolites on OTR function,
mechanisms that maintain the progesterone-repression of the but this evidence, as discussed below, is also controversial.
Cx43 gene during pregnancy and withdraw progesterone
action functionally at labor. Prostaglandins (PGs)
The Oxytocin Receptor (OTR) Prostaglandins are important paracrine regulators of
uterine function in normal and pathological pregnancies and
Oxytocin is a potent and specific stimulant of uterine are used clinically to induce abortion and stimulate parturi-
contractions (see below). Oxytocin receptor expression in the tion. Their involvement in myometrial regulation is discus-
myometrium, measured as specific binding of radioactive sed later; here we summarize those aspects that link intrau-
oxytocin to crude membrane fractions, increases about 12- terine PGs to the action of progesterone. Pharmacological
fold from early pregnancy (13-17 weeks) to term, and a progesterone withdrawal by mifepristone [9] or epostane in
further increase occurs during labor [34-36]. A similar surge early or mid-gestation [45, 46] increases the sensitivity of the
of OTR protein and mRNA abundance accompanies the uterus to prostaglandins. This synergism may explain the
increase of hormone binding activity [37]. The up-regulation efficiency of combined mifepristone/PG-analog treatments
of myometrial OTR expression results in enhanced to terminate early pregnancies [47]. The way mifepristone
sensitivity of the uterus to oxytocin at term [38], which, increases PG-sensitivity, however, is unclear. For example,
according to Mitchell and Schmid [39], argues for a vital Norman et al. [10] found that the mifepristone-enhanced
2502 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

myometrial contractions, monitored by intrauterine pressure relaxing myometrial strips from women in labor. The
recordings, were not attenuated by indomethacin in a dose importance of the decreased sensitivity of the myometrium
that reduced decidual PGF2α production. The obvious to CGRP at labor is highlighted by the observations of Florio
explanation that progesterone controls myometrial PG- et al. [61], who reported that maternal CGRP levels do not
sensitivity by influencing FP-receptor or other PG-receptor change in labor. Accordingly, Dong et al. [60] detected a
expression is supported by evidence in rats and mice [48-51]. marked decrease of immunoreactive (60 kDa) CGRP-
Data on pregnant human myometrium, on the other hand, is receptor protein levels in laboring myometrium and a
very limited [52]. Cell and tissue culture studies may concomitant decrease of CGRP (Type-1)-receptor mRNA
establish unequivocally whether progesterone controls PG- measured by polymerase chain reaction using primers that
receptor expression in the pregnant human uterus. hybridize to a cognate receptor-cDNA sequence [62]. The
group has since characterized the steroid responsiveness of
Substantial evidence suggests that progesterone up-
the myometrial CGRP-receptor[63]. Using a monoclonal
regulates the expression of 15-hydroxyprostaglandin dehyd-
antibody that appears to recognize a new variant of the
rogenase (PGDH) in uterine smooth muscle, placenta and the
CGRP-receptor, called CGRP-B receptor [64], they have
chorion membrane. The PGDH enzyme catalyzes the key
step of prostaglandin catabolism and has a major influence concluded that progesterone stimulates CGRP receptor
on the tissue level of uterotonic prostaglandins such as PGE2 protein-B expression dose dependently almost six-fold in
and PGF 2α. Since PGs can diffuse between adjacent tissues, non-laboring myometrial explants. Estradiol alone is
inhibitory, but together with progesterone stimulates further
PGDH activity in the myometrium, as well as in the
juxtaposed chorion laeve and in the nearby placenta, can the level of the receptor protein. It will be important to
compare the effects of steroids on tissues obtained before vs.
affect the myometrium. Greenland et al . studied thoroughly
during labor and to monitor specific mRNA levels once the
the steroid regulation of the human PGDH promoter in
CGRP-B receptor mRNA is characterized. Based on the
primary myometrial and endometrial (stromal) cell cultures
available data, the myometrial CGRP-B receptor is a
[53]. They have transfected the cells with a series of PGDH
promising candidate to study functional progesterone
promoter-reporter constructs and detected a marked stimu-
lation of reporter expression by progesterone. To achieve withdrawal at the gene expression level.
stimulation, however, it was necessary to overexpress the
Progesterone Receptors
progesterone receptors by co-transfecting a PR-expression
vector. Overexpressed PR-A and PR-B were both active in Changing PR abundance in the myometrium can
the assays, and a cAMP-analog synergized with progesterone potentially influence the progesterone responsiveness of the
in the presence of PR-B but not of PR-A. Notably, the tissue and underpin, at the molecular level, the concept of
endogenous PR, present in the endometrial cells, was unable functional progesterone withdrawal. Early studies using
to mediate the stimulatory effect of progesterone on PGDH ligand-binding assays and an enzyme-linked immunoassay
promoter-driven reporter expression. Furthermore, no (EIA) found that PR levels were markedly lower in the
consensus PRE was identified in the 2.3 kilobase 5’ flanking pregnant than in the non-pregnant myometrium [20, 65, 66].
sequence of the human PGDH gene [53]. Thus, the At the same time, a characteristically large proportion of the
physiological significance of these PGDH promoter-trans- binding sites was associated with the nuclear fraction in
fection experiments is unclear. In vivo, myometrial PGDH pregnant myometrium, which was concordant with the
activity and protein abundance are unchanged during human persistent high tissue progesterone concentrations during
pregnancy, but decrease at term and preterm labor [54]. pregnancy. Subsequently, several groups have confirmed the
Patel et al. characterized the steroid regulation of PGDH predominant nuclear localization of PR in gravid
activity in primary cultures of chorionic and placental myometrium using immunohistochemistry [21, 33, 67].
trophoblasts [55-57]. Using a variety of natural and synthetic Attempts to assess the tissue abundance of PR by scoring the
progestins, glucocorticoids and hormone antagonists, they intensity of the immunohistochemical staining provided
have concluded that progesterone up-regulates and cortisol conflicting results. How et al. [21] reported a decrease of
down-regulates the activity of PGDH in these cells. The staining with advancing gestation and with term and preterm
regulation appears to be mediated by the progesterone and labor, while Roh et al. [67] detected no change of PR
glucocorticoid receptors and involves changes in PGDH abundance with term labor and decreased abundance in post-
mRNA levels. In the absence of identified consensus or cell- term pregnancies. Patient numbers were rather low in both
specific PRE and GRE sequences in the PGDH promoter, studies (between 3 and 10 per group) lowering the power of
these steroid effects are likely indirect and require further the semi-quantitative scoring even further. Using the
characterization to assess their physiological roles. quantitative EIA methodology, Rezapour et al. [22]
examined myometrial PR levels in a larger cohort of women
Calcitonin Gene-Related Peptide (CGRP) and its (n=18-19 per group). Significantly, these investigators took
Receptors biopsies not only from the lower uterine segment, where the
incision for cesarean delivery and the sampling of
The 37-amino acid neuropeptide, CGRP, is a potent myometrium are usually performed, but also from the fundus
vasodilator present in the maternal plasma in increasing of the uterus. The double sampling enabled them to detect
concentrations as pregnancy advances [58, 59]. Dong et al. regional differences in myometrial PR expression. They have
[60] have shown that CGRP is also an effective inhibitor of found that PR abundance increases in the fundus but not in
the spontaneous contractions of myometrial strips isolated the lower segment myometrium at term labor. Thus,
from non-laboring women, but it is much less effective in according to this set of data, labor is associated with
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2503

increased PR expression in the upper segment but not in the emerging [77-79]. Of the three, the team of Charpigny et al.
lower segment of the uterus. In oxytocin-resistant labor, examined the highest number of genes, 2352, using two
however, PR levels did not increase in the fundus and macroarrays. They found that only 4% of the screened genes
decreased in the lower segment as compared to the term not- exhibited altered expression with advancing gestation and
in-labor group. Winkler et al. [68] used EIA to measure PR labor at term. Remarkably, many developmental, prolife-
levels in the lower segment myometrium during the course ration-related and cell-adhesion-associated genes were
of labor. This study detected a transient decrease of PR massively down-regulated during late gestation, and a lower
abundance during active labor, reaching a nadir at 2-3.9 cm number of pro-inflammatory, contraction- and apoptosis-
cervical dilation. The temporary and regional differences in related genes were up-regulated relatively modestly during
PR levels may indicate a dynamic spatio-temporal regulation late pregnancy or labor. Results from the other two studies
of uterine PR levels including, for example, the changing were in general agreement with these findings. Clearly,
cellular composition of the uterine tissue [69] at term. functional progesterone withdrawal in the period leading up
However, differences in investigative techniques may also to term labor is associated with the repression of many genes
contribute to the variability. involved in pregnancy maintenance possibly giving way to
To assess the functional consequences of changing PR pro-inflammatory processes at labor. Traditional approaches
levels, it is necessary to consider the isoform distribution of will be needed to establish which of the regulated genes are
PRs in the various parts of the uterus and during the various controlled directly by progesterone.
stages of pregnancy. The antibodies used in the EIA and New findings by Condon et al. [15] suggest a plausible
immunohistochemistry measurements discussed above did mechanism by which a number of progesterone-controlled
not differentiate between PR-A and PR-B, although both PR genes can be functionally down-regulated in term myomet-
isoforms are expressed in the pregnant human myometrium rium. These investigators have reported that the levels of
[21]. As pointed out earlier, the transcriptional activity of the three PR coactivators, SRC-2, -3 and CBP decrease in the
two PRs differs in a promoter- and cell-specific fashion; human (fundal) myometrium in labor. The decrease of both
however, the human PR-A, in general, has a dominant mRNA and protein levels was detected. Since the coacti-
negative effect over other steroid receptors including PR-B vators participate in the actions of a variety of transcription
[13, 18]. The inhibitory action of PR-A over PR-B has also factors in addition to PR [80], reduced levels may suppress
been confirmed in transfection studies using myometrial cell the expression of many genes depending on the specificity of
cultures from term pregnant women [70]. Mesiano et al. transcription factor interactions. Moreover, these coactiva-
have recently examined the abundance of PR-A and PR-B tors have intrinsic protein acetyltransferase activity, which is
mRNAs in myometrial biopsies obtained before and during involved in general chromatin remodeling, for example, by
term labor [71]. They have found that both PR-A and PR-B histone acetylation. In agreement with this, Condon et al.
mRNA levels increase in labor, but the increase of PR-A found decreased levels of acetylated histone H3 in laboring
mRNA abundance is more marked resulting in an elevated myometria, indicating a relatively closed chromatin struc-
PR-A/PR-B mRNA ratio. Interestingly, the PR-A/PR-B ture. Treatment of pregnant mice with trichostatin A (TSA),
mRNA ratio correlated in the same laboring tissues with the a histone deacetylase inhibitor, increased histone H3
level of HOXA 10 mRNA, which is an mRNA species acetylation and significantly prolonged pregnancy. In a cell
down-regulated by progesterone in non-pregnant myometrial culture study, however, histone hyperacetylation by TSA
cell cultures [72]. This observation suggests that the PR- blocked the PR-dependent transcription of a mouse
A/PR-B mRNA ratio may be relevant functionally. Further mammary tumor virus (MMTV) promoter-driven reporter
data on PR-A/PR-B protein levels and expression studies on [81]. Collectively, these data indicate that protein or histone
the PR-A/PR-B dependent regulation of progesterone acetylation remodels chromatin and modulates progesterone
responsive genes are needed to corroborate the possibility action in a promoter-specific manner.
that preferential PR-A expression is a molecular mechanism
of functional progesterone withdrawal in laboring human None-Genomic Actions of Progesterone
myometrium.
Strips of pregnant myometrium, when suspended
As discussed in the preceding sections, a major difficulty vertically in an organ bath and perifused with oxygenated
impeding the study of progesterone action during pregnancy and temperature-controlled nutrient buffer, start to contract
is that no myometrially expressed human genes were found spontaneously within 50-90 minutes. Usually one or two
that are well-characterized structurally and functionally, have contractions occur per 10 minutes, which may continue for
critical roles in pregnancy, and respond to progesterone several hours. Connected to an isometric recording device,
robustly in vivo and in vitro. There are candidate progesterone this arrangement is often used as an model of phasic myo-
responsive genes in addition to those discussed, such as metrial activity characteristic of labor. Progesterone added to
HOXA 10 [72], TGFβ1, TGFβ receptor type 1 [73, 74], or the perifusion medium diminishes the spontaneous contrac-
hemoxygenase-1 and 2 [75], although the involvement of the tions within minutes [82, 83]. The inhibition is reversible
hemoxygenases in maintaining uterine quiescence has been and stops within minutes after progesterone is washed out.
questioned [76]. Large-scale functional genomic studies, Detailed analysis of the contraction parameters showed that
which screen hundreds or thousands of genes for differential progesterone decreased the amplitude and the work area of
expression, may uncover new candidates for progesterone- the contractions and increased the contraction frequency and
controlled genes in the myometrium. From three recent the tone of the muscle [84, 85]. The effects of the steroid
studies using functional genomic approaches, an overall were also dose dependent, and importantly, were observed
pattern of labor-related myometrial gene expression is only at supraphysiological progesterone concentrations
2504 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

(IC50=225µM). (Progesterone concentration is 0.2-0.3µM in effects of progesterone, 5β-DHP and other steroids on ligand
uterine tissue [86] and approximately 2µM in venous uterine binding and calcium signal generation. Nearly all tested
blood [87]). The rapidity of action and the requirement of steroids inhibited ligand binding and calcium signaling by all
non-physiological hormone concentrations suggested that receptors to a degree dependent on the particular receptor
progesterone inhibited the myometrial contractions by a non- and cell line. The effective concentration range was 10-
genomic, receptor-independent mechanism. Further support 500µM. The steroid effects were clearly non-genomic and
for this suggestion was provided by steroid specificity were related to membrane composition and/or properties
studies, which showed that the progesterone receptor [94]. Overall, the results did not support the possibility of
antagonist mifepristone (RU 486) did not block the action of direct interaction of the steroids with the receptor proteins.
progesterone. Moreover, progesterone metabolites, especially Finally, Astle et al. re-examined the effect of 5β-DHP on
the 3α-hydroxy-5α-reduced progesterone derivative and 5β- oxytocin binding and (human) OTR-dependent signaling
dihydro-progesterone, were even more effective than [95]. The data in this report demonstrated that 5β-DHP (up
progesterone (IC50 values: 35µM and 81µM, respectively) to 100µM) had no effect on the binding of oxytocin to the
[83, 88]. According to work by Fu et al. [89], 15µM and OTR either in membranes from pregnant human myomet-
150µM progesterone elevated the cAMP content of the rium or in CHO cells expressing OTR. Furthermore, 5β-
perifused myometrial strips and increased the release of DHP (1µM) had no influence on the inward calcium current
cAMP into the medium, suggesting that the anti-contractile in a whole-cell patch-clamp arrangement with primary
effect was mediated at least in part by cAMP. Further, human myometrial cells.
Kofinas et al. [86] have shown that pharmacological concen- Despite the controversy surrounding the actions of
trations of progesterone inhibited the activity of cAMP-
progesterone and 5β-DHP on OTR-signalling, clinical data
phosphodiesterase in a cell-free assay with myometrial
show that progesterone is effective in preventing preterm
extracts. Thus, progesterone may cause cAMP accumulation
birth in high-risk pregnancies. Recently, the results of a
by directly inhibiting the breakdown of the cyclic nucleotide.
multicenter clinical trial have been published, where 463
Following up on early observations [90], Fu et al. high risk women were enrolled in a double-blind placebo-
examined the influence of progesterone on oxytocin- controlled design [96]. The treatment group (310 partici-
stimulated myometrial contractions [91]. Addition of 16µM pants) received weekly injections containing 250 mg of 17α-
progesterone together with oxytocin (10 or 100 mU/ml) hydroxyprogesterone caproate, which was an effective dose
decreased the amplitude and the activity area of the in previous pilot studies (reviewed in ref. [96]). High risk
contractions, increased the frequency of the contractions and was determined as spontaneous preterm delivery in a
elevated the tonus of the muscle. These effects were similar previous pregnancy. The treatments started at 16-20 weeks
to those seen on the spontaneously contracting myometrium. of pregnancy and resulted in a significant reduction of the
At the same time, progesterone prevented the tachyphylaxis incidence of preterm deliveries before the 37th (term), 35th
of the oxytocin response normally seen after longer exposure and 32nd weeks of gestation. In an other clinical study
to the peptide. The physiological significance of the anti- involving 142 women with high risk pregnancy [97], vaginal
tachyphylactic effect is not clear; however, it indicates that suppositories containing 100 mg progesterone were
progesterone interfered with OTR-dependent signalling administered weekly from 25-27 weeks of gestation. The
under the experimental conditions employed. Grazzini and design was also randomized, double blind and placebo-
co-workers reported a direct interaction between the human controlled (treatment group: n=72). Preterm birth rate and
OTR and the progesterone metabolite 5β-dihydro-proges- delivery rate before 34 gestational weeks were significantly
terone (5β-DHP) [92]. The interaction was demonstrated in lower in the treatment group. Uterine activity, monitored by
CHO (Chinese hamster ovary) cells stably transfected with an external tocodynamometer, has been also reduced in the
the human OTR. The binding of oxytocin to membranes progesterone-treated group. These clinical studies strongly
from the transfected cells was inhibited by 5β-DHP with a Ki suggest that the prophylactic use of progesterone may
of 32 nM. Moreover, 5β-DHP inhibited the oxytocin-induced decrease the probability of preterm birth in a high risk group
inositol phosphate production in the CHO cells with a very of pregnant women, and importantly, may improve birth
low Ki (0.25nM). Progesterone was ineffective in this outcomes [96]. The mode of action of progesterone is
system, but had analogous inhibitory actions on the rat OTR. unknown, but it is tempting to speculate that the non-
The data by Grazzini et al. indicated that 5β-DHP was an genomic pharmacological action(s) of progesterone play a
effective oxytocin antagonist in addition to its inhibitory role opposing the consequences of any functional progeste-
effect on the spontaneous myometrial contractions discussed rone withdrawal that may occur pathologically before term.
above. Subsequently, Thornton et al. [88] tested the influ-
ence of 5β-DHP on oxytocin-augmented myometrial contrac- ESTROGEN
tions. They found that 100µM 5β-DHP in the medium
decreased the oxytocin sensitivity of the myometrium, In the sheep [98] or the rat [99], the fall of progesterone
measured as activity area or peak contraction force, by about levels is followed by a sharp rise in estrogen concentrations
10-fold. This suggested that 5β-DHP might be a tocolytic in the maternal plasma at term. Thus, progesterone
withdrawal is accompanied by estrogen activation in these
oxytocin antagonist. The encouraging results prompted
further studies to explore the interaction 5β-DHP with the animal species, and the hormonal changes are believed to
bring about labor and delivery by acting together. In the
OTR. Burger et al. [93] transfected several G-protein-
coupled receptors, including the human OTR, into various human, progesterone and estradiol concentrations increase
gradually in the maternal plasma during the second and third
cell lines including CHO cells. They have measured the
trimesters and show no change in this tendency at term
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2505

before labor onset [100]. The ratio of estrogen to progeste- overexpressing vectors are co-transfected into cell lines in
rone concentrations in the maternal saliva also remains order to perform functional analyses. The regulatory
unchanged during the last twenty weeks of pregnancy, interactions discovered in these models indicate several
reflecting the unchanged ratio of unconjugated (free) mechanistic possibilities that are in line with the concept of
hormone levels in the plasma [101]. Furthermore, estradiol functional estrogen activation. For example, the change of
levels do not change within human myometrium in labor ER-isoform and cofactor levels, increased growth factor
[86]. Collectively, these data suggest that estrogen activa- signaling, altered chromatin structure and changing trans-
tion, like progesterone withdrawal, must be functional in cription factor transrepression profile may all potentially
human pregnancy at term. The symptoms of the recently increase the estrogen sensitivity of a target cell. Further work
described congenital aromatase deficiency syndrome [102] is needed to determine which of the possible regulatory
are in agreement with this assumption. The estrogens of mechanisms are involved in the physiological and patho-
human pregnancy (including estradiol and estriol) are physiological regulation of the myometrium by estrogens.
produced by placental aromatase, which converts fetal One critical issue is to identify functionally important estro-
adrenal androgens to estrogens. Aromatase deficiency, gen responsive genes in the pregnant human myometrium.
caused by the recessive autosomal mutations of the CYP19 Information about some of these genes is summarized below.
gene, results in low estrogen levels in pregnancy, virilization
of the female fetus and the mother, but no reported disorder Connexin 43
of the parturition process. Maternal estrogens, of course, are
Estrogen treatment of cultured human myometrial cells
present in these pregnancies, but their concentrations are
grossly abnormal. It is reasonable to conjecture that in these increases the level of Cx43 protein [28, 30, 33, 109], sugges-
patients, the maternal estrogen is sufficient to activate the ting that the Cx43 gene is estrogen responsive. The human
estrogen receptors in the uterus, and any change in estrogen Cx43 gene promoter contains 6 half-ERE elements [31];
however, no evidence suggests so far that these sequences
action at term is the result of regulation at the receptor or
are involved in the up-regulation of Cx43 gene activity in
post-receptor levels.
uterine myocytes. Nuclear run-on, DNAse I footprinting, gel
mobility shift and transfection assays indicated that the AP1
Estrogen Receptor-Dependent Regulation
and SP1 sites within the proximal Cx43 promoter were
The pregnant human myometrium is an estrogen target important positive regulatory elements, but these sites and
tissue expressing estrogen receptors (ERs) [20]. The ERs the corresponding transcription factors did not mediate the
bind the hormone with high affinity and specificity, then estrogen stimulation, even when the ER was overexpressed
undergo a conformational change and gain ability to function in primary myometrial cells [32, 33]. Thus, the mechanism
as a transcription factor. Estrogen responsive genes contain a of the estrogen stimulation of human Cx43 expression
nucleotide sequence within the promoter (the ideal remains unknown. An interesting possibility has been raised
palindrome is AGGTCANNNTGACCT), which binds the by new work from Oltra et al. [110], who isolated a small
active ER dimer resulting in transcriptional activation. These zinc finger protein from an estrogen induced rat myometrium
steps are akin to those described for progesterone and the expression library. In cotransfection assays, this protein,
PRs. Likewise, the outline of the ER-dependent gene called Ini, enhanced the estrogen response of the Cx43 gene
regulation is very simplified. There are two ER isoforms, in a cell specific fashion. The reporter construct used in the
ERα and ERβ, produced by separate genes. In vitro gene experiments contained the proximal 145bp region of the
transfer experiments suggested that the two ERs have human Cx43 gene promoter, which does not possess half
distinct modes of action on the same promoters and have palindromic EREs. Ini is a widely expressed and highly
different ligand activation properties [103]. Moreover, ERβ, conserved transcriptional cofactor also found in humans. The
in general, is a transrepressor of ERα [103]. These potential involvement of Ini in regulating the estrogen
differences also exhibited cell line-dependent variations. sensitivity of the Cx43 or other genes in the pregnant human
Further, the two receptor isoforms showed opposing actions myometrium deserves further study.
on a promoter containing an AP1 site [104]. Growth factors,
such as EGF, and cAMP activate ERα independently of the The Oxytocin/OTR-System
steroid ligand [105], which represents an additional level of
regulation involving the phosphorylation of ERα by the Estrogen stimulates myometrial OTR gene expression in
MAP-kinase system [106], and possibly by PKA. ERα ovariectomized rats [111]. Primary cultures of myometrial
cells from non-pregnant women also respond to estrogen
interacts with a large number of cofactors when it performs
with increased OTR expression, as determined by a ligand-
its gene-regulatory function (reviewed by McDonnel and
binding assay [43]. The 1.8 kb upstream promoter region of
Norris [107]). The cofactors can be either activators or
repressors and are involved in chromatin remodeling by the human OTR gene contains 3 half-palindromic EREs
histone acetylation, and also in establishing a link between [42], but these elements do not mediate estrogen stimulation
the receptor and the basic transcription factors. Finally, ER is in transfection assays even when the cells are cotransfected
with ER-expressing vectors [112]. Thus, the mechanism by
subject to transrepression by other transcription factors, such
which estrogens regulate the OTR gene is unclear. Estrogen
as PR-A (discussed above), and NF-κB p65 [108].
may stimulate the oxytocin responsiveness of human
The bulk of data summarized above on the molecular myometrial cells at a post-receptor step. This was suggested
regulation of ER function has been obtained in gene by Phaneuf et al. [113], who measured the activation of
transfection experiments. In these experiments, estrogen- phospholipase C (PLC) by oxytocin, fluoroaluminate (a G-
responsive promoter-reporter constructs and various ER- protein activator) and PGF2α in primary (non-pregnant)
2506 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

myometrial cell cultures. Estrogen pretreatment increased the expression at the time of labor. Nevertheless, there is still no
efficiency of all three PLC activators, and tamoxifen par- information about myometrial ER expression during
tially reversed the estrogen effect suggesting an involvement gestation, prior to labor onset.
of the ERs. The immediate target of estrogen regulation in
Mesiano et al. [71] have recently examined myometrial
the signal transduction system has not been identified.
ER expression using the sensitive and highly specific real-
The oxytocin gene is expressed in the human decidua and time quantitative RT-PCR technique. These investigators
the fetal membranes [114]. Oxytocin mRNA levels increase have found a significant increase of ERα mRNA abundance
in chorio-decidua explants incubated with 1 and 10 nM in myometrial biopsies with term labor. The levels of ERβ
estradiol, and the stimulation is partially abolished by co- mRNA, which encodes the dominant negative ERβ isoform,
incubation with tamoxifen [114, 115]. Estrogens, therefore, were much lower and constant in the same cohort of
may potentially stimulate myometrial contractions indirectly myometrial biopsies. Interestingly, OTR mRNA levels also
by increasing the production of oxytocin in the decidua, increased with labor, and ERα mRNA abundance showed
which is the tissue lining the internal surface of the pregnant significant positive correlation with OTR mRNA abundance
uterus. The human oxytocin gene contains several imperfect in non-laboring tissues. These results, if confirmed by
half-palindromic EREs, and one of these, located at approxi- protein measurements, may support the possibility of ERα-
mately 160 bases upstream of the transcriptional start site, mediated estrogen activation and OTR induction before or
confers estrogen responsiveness to a reporter-construct in during labor.
neuroblastoma cells overexpressing ER [116]. This regula-
tory element is highly conserved and appears to mediate Functional Progesterone Withdrawal and Estrogen
regulation by several transcription factors including orphan Activation: Concepts in Need of Further Testing
receptors, like SF-1 and COUP-TF [40]. Interestingly, ER
The hypothesis of functional progesterone withdrawal
was reported to bind this element with very low affinity [40],
and estrogen activation was formulated as an extension to
which raises the possibility that the stimulation of the
humans of the reproductive endocrinology of animal species,
oxytocin gene by estrogens is not direct, but mediated by
other estrogen-controlled transcription factors or by proteins where circulating progesterone levels drop and estrogen
levels rise before labor onset. Its central postulates are that
that anchor the ER to the regulatory DNA sequence. No such
the responsiveness of the human myometrium to progeste-
factors or mechanisms have been demonstrated so far.
rone decreases and to estrogen increases in term pregnancies.
Similarly, no information is available about the time course
Despite several decades of investigations, however, no
of decidual oxytocin expression during pregnancy or with
conclusive evidence for, or against, the existence of such
labor.
mechanisms has been found. The slow progress is due to
It is likely that estrogen regulates more genes in the major obstacles to research in this area of medical science.
pregnant human myometrium than just Cx43 and OTR. For Tissue availability is limited to small biopsy samples excised
example, CGRP-B receptor protein levels increase in from the myometrium during elective or emergency cesarean
estrogen-treated myometrial explants from term pregnant section. Myometrial cells in primary culture show instable
women [63], and TGFβ1 secretion and TGFβ receptor type-1 phenotype, and immortalized pregnant myometrial cell lines
abundance increases and decreases, respectively, in (non- [119] are still not generally available and characterized.
pregnant) human myometrial cell cultures [74] treated with There are only a few genes known to be under progesterone
estradiol. Given the high number of differentially expressed and/or estrogen control in pregnant myometrial cells, but
genes in the human myometrium in late pregnancy [79], remarkably, reporter construct assays and transfection
other estrogen responsive genes may be identified in the studies were not successful in determining the molecular
future. mechanisms of the steroid regulation of these genes. Without
this information, gestational age-dependent changes of
ER in the Pregnant Human Myometrium steroid responsiveness cannot be examined at the single gene
level. Cross-talk between the PR and ER pathways has also
The level and the activity of ERs may influence the
been proposed, especially the inhibition of ER-signaling by
estrogen responsiveness of the myometrium. This important
PRs [17], by low PR-A to PR-B ratio [71], or possibly by
possibility has prompted several groups of investigators to
PR-A-transrepression, as discussed. The involvement of
measure ER expression in the pregnant myometrium.
these mechanisms in the physiological actions of progeste-
Hormone-exchange assays indicated the presence of ERs in
the nuclear and cytoplasmic fractions of term pregnant rone or estrogen in pregnancy remains to be tested. If the
progesterone and/or estrogen receptor pathways play a
myometria [20, 117, 118], and immmunohistochemistry
central role in the regulation of myometrial gene expression
demonstrated the presence of ERα protein in myometrial cell
during pregnancy, molecular studies of hormone action will
nuclei [21]. Others detected faint and diffuse ER-staining by
immunohistochemistry [67] and low but measurable levels likely lead to the discovery of new drug targets and drugs.
of ERα protein by EIA [22]. The majority of studies found Hopefully, a new generation of drugs targeting key steps in
no difference in ER localization and abundance with normal the maturation of the myometrium to a contractile phenotype
will effectively prevent or treat various forms of
term or preterm labor; however, Winkler et al. [68] reported
that ERα protein levels transiently decreased in the dysregulated myometrial function such as preterm labor,
dystocia, or post-term pregnancy.
myometrium during the early phase of active labor at term.
Overall, available data do not support the possibility that Having reviewed the regulatory functions of female sex
functional estrogen activation occurs by increased ER steroids we shall now turn to what we prefer to term as
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2507

factors modulating myometrial smooth muscle functions different in the upper and lower segments and remained
during pregnancy and labor. unchanged during pregnancy and labor. Furthermore,
receptor expression was generally lower in the pregnant than
Prostaglandins in the non-pregnant myometrium. The various types of PG-
receptors have been identified and characterized originally in
Prostaglandins have long been considered to play impor-
functional studies, using smooth muscle preparations for
tant roles in uterine physiology and pathology, based on their bioassays and a series of PGs, PG analogs and PG
ability to induce abortion as well as parturition at term, to
antagonists as effectors [130, 131]. Such a pharmacological
treat postpartum hemorrhage and to affect myometrial
approach has been applied subsequently to the pregnant
contractility. In addition, the progressive increase in amnio-
myometrium by Senior et al. [132]. The results suggest that
tic fluid PG levels prior to labor and the ability of PG in the muscle bath experiments, the inhibitory PGE2 response
synthesis inhibitors to prolong gestation have further was mediated by the EP 2 receptor, while the excitatory PGE2
underscored this belief. Due to the ubiquity of arachidonic response was triggered by the EP3 receptor, since the other
acid (AA) as a component of membrane phospholipids, PGs excitatory PGE2 receptor, EP1, was not detectable using a
are readily synthesized, depending on the presence and specific EP 1 antagonist. Stimulation by PGF2α was mediated
activity of the cyclooxygenase enzymes (COX-1&2)
by the excitatory FP receptor. PGI2 relaxed the myometrium
catalyzing the rate-limiting step in the metabolism of AA,
acting through the IP receptor, while the mechanism of PGI 2-
and the specific synthases that convert PGH2, the common
evoked contractions was unclear. PGD2 also exhibited a
precursor, to the bioactive end products. Of the two cyclo-
biphasic effect on myometrial contractility, like PGE2 and
oxygenase isoenzymes, COX-1 is expressed constitutively PGI2. The relaxing action of PGD2 was effected by the DP
and apparently does not change during gestation and labor receptor, and the excitation was possibly mediated by the
[120]. The expression of COX-2 mRNA in association with cross-stimulation of FP receptors by the DP-agonists, as
labor is less clear cut. Some investigators found a decrease,
suggested by Fernandes and Crankshaw [133]. Thromboxane
while others reported no change or an increase in COX-2
A2 analog was excitatory, acting through the TP receptor.
message and protein. It would appear that such inconsistent
Thus, the pharmacological characterization revealed that
findings maybe due in part to the site of myometrial tissue
multiple PG-receptors participate in the contractile and
excision. Specimens collected during labor at cesarian
relaxing effects of prostaglandins on the myometrium.
section from the lower segment of the uterus yielded up to
15-fold higher levels of COX-2 mRNA than similar tissue The pharmacological classification of PG receptors has
samples collected from non-laboring patients [121]. It seems, been confirmed by the molecular cloning of the PG receptor
therefore, that the major PGs, prostacyclin (PGI2) and PGE 2, cDNAs and genes and also by studies of the coupled
produced by the myometrium are directed towards cervical signalling pathways. The reader is referred to excellent
ripening rather than enhancing myometrial contractions. This comprehensive and specialized review articles for this body
may also explain in part the relaxant action of PGI2 on of information [134, 135]. In the human myometrium,
myometrial smooth muscle. Once again, the choice of Matsumoto et al. [136] detected EP3 and FP mRNAs by
experimental approach and the qualitative or quantitative northern blotting of polyA+ RNA. The abundance of both
nature of employed methodology may contribute to the mRNAs was lower in pregnancy than in the non-pregnant
variability of reported observations. It is also important to state. Using semi-quantitative RT-PCR, Brodt-Eppley and
note that measuring the message and protein levels may only Myatt measured FP and EP2 mRNA abundance in the
reflect the transcriptional and translational status of the cell. myometrium at different times of gestation [52]. Overall,
Prostaglandin levels will depend on the activity of the they detected a gestational age-dependent decrease in the
biosynthetic enzymes, as well as the presence and activity of (relative) abundance of both mRNAs. In addition, FP mRNA
PGDH, the principal PG-metabolizing enzyme. Thus, levels increased with term labor, but preterm labor had no
dynamic changes in bioactive PG levels can occur if the effect on the myometrial levels of EP2 or FP mRNAs. The
balance is upset from either direction, e.g. in the case of processing of the primary EP3 gene transcript is especially
intrauterine infection attended by a robust induction of COX- complex, giving rise to alternatively spliced C-terminal
2 (see under cytokines). Moreover, it should be emphasized receptor variants. Six variants have been identified so far,
that the myometrium is exposed to PGs produced by and the EP3 subtypes are coupled to various signaling
neighboring intrauterine tissues such as the decidua, the systems that can increase or decrease intracellular cAMP
chorion laeve and the amnion. PG biosynthesis and levels, stimulate phosphatidyl-inositol turnover and activate
degradation in these tissues have been studied extensively the MAP-kinase pathway [137, 138]. These variants,
during the past decades, predominantly because of the therefore, are capable of transmitting both smooth muscle
possible effects of the generated PGs on myometrial contracting and relaxing signals. The expression of EP3-II,
functions. The results have been summarized often and the EP3-III, EP 3-V, and EP3-VI mRNAs has already been confirmed
interested reader is referred to some of the more recent in the human myometrium [137, 139]. In pregnancy,
reviews for additional information [122-126]. myometrial EP3-II mRNA levels decreased and EP3-VI mRNA
levels increased as compared to the non-pregnant state [139].
How do PGs act to modulate myometrial functions? The
The significance of the EP3 variant distribution and changes
first step involves the recognition by specific receptors. in the modulation of myometrial contractility by PGs still
Crude particulate preparations from myometrial homo-
remains to be established.
genates contain high affinity PGE and PGF2α binding sites
indicating the presence of membrane receptors [127-129]. The molecular characterization of the myometrial PG
The number and affinity of these binding sites were not receptors and the coupled signaling pathways is an area of
2508 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

intensive research. The work is expected to identify human parturition represents an inflammatory process. This
prospective drug targets and new drugs for the control of assumption is supported by the observation that leukocytes
uterine activity in pathological conditions such as preterm infiltrate gestational tissues and may also be the major
labor. Examples are the FP receptor and the new FP receptor source of cytokines in the uterus [150]. Further, increased
antagonist THG113, which effectively delayed delivery in an expression of the cytokines IL-1, IL-6, IL-8 and TNF has
animal model of preterm birth [140]. The development of been documented in the myometrium and cervix in
various selective PG-receptor type- and subtype-specific association with parturition [69,151]. Here we shall focus on
agonists and antagonists currently in progress is likely to IL-1, the prototypical proinflammatory cytokine, in terms of
offer new hope for the better treatment of preterm labor, its action on the myometrium..
early cervical maturation or recurrent preterm birth.
The labor-promoting action of IL-1 has been attributed at
The postreceptor actions of PGs are still uncertain. It is least in part to the induction of cPLA2 and COX-2 and the
generally believed that the heptahelical G-protein coupled ensuing rise in PG production by uterine tissues, including
receptor of the uterotonic PGF2α transmits the signal via the the myometrium [121,152-155]. IL-1 signaling in various
phosphoinositide cycle, generating the two intracellular cell types involves multiple pathways leading to the
signaling molecules, IP3 and DAG. IP3, in turn, mobilizes expression of more than a hundred different genes. In intact
calcium from intracellular stores, while DAG is an human myometrial cells, IL-1β binds with high affinity
endogenous activator of PKC. The latter is involved inter (Kd=2 x 10-10M) [156]. Although two IL-1 receptors, each
alia in AA release by way of activating PLA2, thus having a single membrane-spanning domain, have been
contributing to the provision of ample precursor pool. identified, Scatchard analysis indicated the presence of a
However, it is not known if such a mechanism is of single class of binding sites in human myometrial cells. Post
physiological significance. Comparing the effects of PGF2α receptor steps in the signaling pathways leading to COX-2
with oxytocin in primary cultures of human myometrial expression involve the activation of the MAP kinase family
cells, Molnár and Hertelendy [141] observed that both of enzymes, of which p38 MAP kinase is functionally linked
agonists were equipotent in raising [Ca2+]i in the presence of to COX-2 mRNA and protein expression, because SB
1mM extracellular calcium. However, in calcium free 203580, a selective inhibitor of p38 action blocked the effect
medium only oxytocin could increase [Ca2+]i, albeit to a of IL-1 in primary cultures of human myocytes [157,158].
considerably lesser extent, confirming a previous report by However, the role of jun-terminal kinase (JNK), which
Anwer and Sanborn using rat myometrial cells [142]. responds robustly to IL-1 stimulation, is uncertain and a
Further, at concentrations which raised [Ca2+]i, PGF2α failed crosstalk between MAP kinases is not uncommon in various
to activate the phosphoinositide cycle, responding only at other systems. Another signaling pathway that leads to
micromolar concentrations unlikely to be encountered in vivo. activation and nuclear translocation of NF-κB has also been
This observation, combined with the finding that the calcium described in an immortalized human myometrial cell line
channel blocker verapamil inhibited PGF2α-promoted rise in [159]. The nuclear transcription factor NF-κB in its inactive
[Ca2+]i, led to the conclusion that the primary effect of PGF2α form is localized in the cytosol bound to the inhibitory
in myometrial cells is to promote calcium entry. Prostacyc- protein IκB. Upon stimulation with IL-1, IκB is rapidly
lin, on the other hand, activates the AC/cAMP system which, degraded allowing the translocation of NF-κB to the nucleus
as pointed out elsewhere, is involved in myometrial where it can interact with one or both of the NF-κB binding
relaxation and is down regulated at the onset of labor [143]. motifs present in the promoter of human COX-2 gene.
Stimulation of myometrial cells caused a rapid degradation
It has also been suggested that PGs may function as
of IκB (as assessed by immunoblotting) and nuclear binding
ionophores [144], or by altering membrane fluidity [145],
of NF-κB (as demonstrated by electrophoretic mobility shift
leading ultimately to increased calcium entry. More recent
assay) [159]. Inhibition of IκB degradation prevented IL-1
studies indicate that PGs may activate the peroxisome-
induced COX-2 mRNA and protein expression, as well as
proliferation-activated receptor signaling pathway affecting
PGI2 synthesis. Combined stimulation of myocyte cultures
gene transcription [146]. Gene transcription may also be
modulated by PGs in an autocrine fashion via intracellular with TNF greatly potentiated PGE2 and PGI 2 production, but
receptors identified in the perinuclear envelope [147]. another cytokine, interferon-γ, significantly suppressed IL-
induced COX-2 expression and PG production [160].
Collectively, PGs, in conjunction with their numerous
receptors that have been characterized, activate multiple Interestingly, IL-1-induced activation of MAPK, COX-2
signaling pathways to bring about their multifaceted actions expression and PG synthesis was also partially inhibited by
in uterine smooth muscle cells. Clearly, more work is needed pertussis toxin, a widely used tool to probe for the
involvement of certain G-proteins in signal transduction
to elucidate the intimate mechanisms of action of the
[161]. Thus, IL-1 receptor that does not belong to the G-
principal PGs and other bioactive eicosanoids in association
with the pregnant and parturient uterus. protein coupled receptors with seven membrane spanning
domains, interacts in some fashion with the latter. In a
fascinating recent study, Tribe et al. [162] investigated the
Cytokines
effects of IL-1β on the expression of sarcoplasmic reticulum
The inflammatory cytokines, chiefly IL-1, TNF and IL-6, calcium ATPase (SERCA) 2b and Ca2+ fluxes in primary
have long been linked to intrauterine infection-driven cultures of myometrial cells isolated from tissue specimens
preterm labor [148]. There is accumulating evidence, obtained at cesarian section from women not in labor. They
however, that pro-inflammatory cytokines are also involved found a significant increase in SERCA 2b protein expression
in normal term labor [149]. In fact, it has been proposed that and calcium mobilization after 24 h treatment with 10ng/ml
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2509

IL-1β. The rise in [Ca2+]i was apparently due to increased without affecting a rise in [Ca2+]i. However, oxytocin-
release, as well as entry of Ca2+; the latter independent of induced phosphorylation of MLCK was attenuated, as was
altered activity of voltage-gated calcium channels. oxytocin-induced phosphorylation of MLC phospatase,
which inhibits the activity of the enzyme. There is also
In conclusion, proinflammatory cytokines, especially IL-
evidence that the expression of Rho-kinase is increased in
1β, are powerful inducers of COX-2 and PG production in
human myometrium. Interleukin’s multifunctional character the myometrium during pregnancy [175, 176].
is reflected by the activation of multiple signaling pathways, In summary, the oxytocin/OTR system plays an impor-
which may include mechanisms that influence intracellular tant, though perhaps not a decisive role in the regulation of
calcium levels and, hence, myometrial contractility. myometrial function in the context of labor and delivery.

Oxytocin Endothelin
Even though oxytocin has been widely used in clinical The vasoconstrictor, endothelin (ET-1), is a potent
practice for the pharmacologic induction or augmentation of stimulant of uterine smooth muscle contraction. The action
labor [163], its physiologic role in the initiation of parturition of ET-1 is mediated by the A-type receptors (ETA) coupled
has been disputed, owing to a lack of its demonstrable to the PLC-phosphoinositide-Ca2+-signaling pathway, with
increase in plasma levels prior to the onset of labor. More the ensuing phosphorylation of MLC and force generation
recently, however, an increase in oxytocin mRNA and [177, 178]. ETA-receptors in the myometrium increase as
peptide has been demonstrated in human and rat uterine pregnancy progresses towards labor [178, 179], supporting a
tissues [114, 164]. This finding, together with the striking physiologic role for ET-1 in the regulation of parturition.
increase in oxytocin receptors (OTR) at term and, ET-1 has also been found to stimulate the expression of
particularly, during labor, lends support to a paracrine role COX-2 and PG production in human myometrial cells [180].
for oxytocin in the regulation of myometrial function Recent studies provide experimental evidence for the
associated with the birth process. Since oxytocin sensitivity operation of an additional signaling pathway activated by
of the myometrium parallels the progressive rise in OTRs ET-1 [181]. Stimulation of human myometrial cells activated
during gestation, it suggests that it is the OTR that is of an atypical protein kinase C isoform (PKCξ) that is not
primary importance in promoting myometrial contractility regulated by DAG or calcium, but by phospholipidic
[165]. This notion is supported by the efficacy of various mediators, products of phosphatidylinositol 3-kinase (PI3-
OTR antagonists to suppress uterine contractions and their kinase) [182]. Inhibition of PI3-kinase by LY-294002 or by
clinical applications as tocolytic agents (see below). It an antisense against PKCξ significantly suppressed ET-1-
should be noted, however, that OTR antagonists can only induced myometrial contractions [181]. Activated PKCζ
delay, but not block parturition, indicative of the operation of associates with the actin component of the cytoskeleton of
additional pathways responsible for the initiation and human myometrial cells isolated near term and may regulate
maintenance of parturition. the phosphorylation or binding properties of actin. Interest-
ingly, ET-1-activated PKC may also interact with the
OTR belongs to the superfamily of the heptahelical, G-
signaling pathway of inflammatory cytokines (IL-1β, TNFα)
protein-coupled receptors. Ligand binding activates multiple
which regulate the multifunctional nuclear transcription
signaling pathways. Of these, the activation of phospholipase
factor NF- κB [183, 184]. Finally, ET-1 stimulates myomet-
Cβ (PLCβ), which breaks down PIP2 to IP3 and DAG, has
rial cell proliferation [185,186], perhaps contributing as a
been amply demonstrated in human [166-168] and animal
paracrine regulator to uterine growth during pregnancy.
[169, 170] myometrial tissues and cells. The ensuing rise in
IP3 releases Ca 2+ from the sarcoplasmic reticulum, causing a
sudden rise in intracellular Ca2+ ([Ca2+]i), whereas DAG Platelet Activating Factor (PAF)
activates protein kinase C (PKC). Activation of PLA2, e.g. by inflammatory cytokines,
Another important function of oxytocin is the promotion leads not only to the release of AA and the subsequent
of PG production by stimulation of arachidonic acid (AA) generation of prostanoids modulating myometrial functions,
release by the sequential hydrolysis of DAG and by G- but can also give rise to PAF, by way of the acetyl-CoA
protein-coupled activation of PLA2, that may also be acetyltransferase catalyzed acetylation of phosphatidyl-
activated by oxytocin-induced increase in [Ca2+]i. In choline. (In addition to this “remodeling pathway” in the
addition, oxytocin-induced activation of the MAP kinase central nervous system and some other tissue, a “de novo”
pathway [171], coupled to the activation of cytosolic PLA2 pathway can also generate PAF via the synthesis of 1-O-
(cPLA2), should also enhance the release of AA, the alkyl-2-acetylglycerol, which is then converted to PAF by a
common precursor of prostaglandins and other eicosanoids. specific dithiothreitol-insensitive CDP-choline-1-alkyl-2-
Prolonged exposure of human myometrial cells to oxytocin acetyl-sn-glycerocholine-phosphotrans-ferase [187]).
has been shown to promote COX-2 gene transcription and PAF produced in endothelial cells may act in a paracrine
sustained PG production [172]. Such a signaling pathway for fashion on myometrial cells. Stimulation of myometrial
oxytocin has also been demonstrated in bovine uterine cells contractions by PAF has been observed both in vivo and in
[173, 174]. Finally, oxytocin can also enhance myometrial vitro [188]. Its putative role, such as premature rupture of the
contraction by a calcium-independent mechanism involving membranes and preterm labor, particularly in complicated
the RhoA/Rho-kinase cascade [175]. Studies using Y-27632, pregnancies, is supported by the detection of PAF in amnio-
a specific inhibitor of Rho-kinase, have demonstrated an tic fluid [189]. PAF receptor antagonists have been shown to
inhibition of oxytocin-induced myometrial contraction delay parturition in rats [190]. In human myometrial cells, as
2510 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

in endothelial cells, PAF activates the phosphoinositide cycle is a multifunctional serine protease, affecting a wide variety
after binding to specific G-protein-coupled receptors, of cellular functions [202]. It is generated by catalytic
causing an increase in [Ca2]i due, in part, to the release of cleavage from its precursor, prothrombin, by the activated
bound intracellular calcium [191]. This response to PAF was factor Xa. Most of its actions are mediated by protease
inhibited by L-659,989, a PAF-receptor antagonist. Stimula- activated receptors (PARs), belonging to the superfamily of
tion of eicosanoid production may also contribute to the heptahelical G-protein-coupled receptors. Activation of PAR
uterotonic action of PAF [192]. Thus, there is a close analogy involves the enzymatic cleavage of the amino-terminal
between the action of ET-1 and PAF on myometrium. This is exodomain of the G-protein-coupled PAR, revealing a new
further underscored by the possible involvement of PAF in amino-terminal that acts as a tethered ligand, binding intra-
PKC-mediated cytoskeletal rearrangement as demonstrated molecularly to the body of the receptor initiating transmem-
in endothelial cells [193]. brane signaling. Synthetic peptides coinciding with the
amino acid sequence of the new amino-terminal (SFLLRN),
Arachidonic Acid (AA) referred to as TRAP (thrombin receptor activating peptide),
can activate PAR independently of thrombin without the
AA is distributed throughout the body as a polyunsatu- splitting off of the N-terminal segment of the receptor.
rated fatty acid component of membrane phospholipids. Its
Unlike other G-protein-coupled receptors, PAR activation is
physiological role in the myometrium and various other
irreversible and the receptor is rapidly internalized and
tissues has been attributed to its acting as the principal
degraded. Replenishment is from preformed intracellular
precursor of prostaglandins and other biologically active
stores. Like oxytocin and other uterotonins, thrombin-
eicosanoids. There is good evidence, however, that AA itself activated PAR is coupled to the phosphoinositide signaling
may serve as a cell signaling molecule [194]. AA, when cascade, generating IP 3 and DAG as intracellular messengers
added to various cell types, including human myometrial with the subsequent rise in [Ca2+]i and activation of PKC. In
cells [195], has been shown to stimulate the release of
addition, however, thrombin activates a number of other
intracellular calcium, even in the presence of inhibitors that
signaling pathways involving, e.g., the MAP kinase family
block the conversion to eicosanoids.
of enzymes, PI3 kinase, JAK-2/STAT-1, etc. (reviewed in
Stimulation of [3H]-AA-labeled human myometrial cells [203]).
with PGF2a caused a rapid release of AA, chiefly from
A novel aspect of thrombin action in human myometrial
phosphatidylethanolamine, indicative of PLA2 activation,
cells has been described recently [204]. Stimulation of
whereas oxytocin liberated AA mainly from phosphoino-
primary cultures of human myocytes with thrombin upregu-
sitides, suggestive of G-protein-coupled PLC activation. AA
lated COX-2 expression and PGE2 production in a time- and
may also participate in intracellular signaling by activating a
dose-dependent manner. These responses were completely
specific PKC that is distinct from the “classic” DAG-
abolished by actinomycin D, indicative of transcriptional
sensitive enzyme [196]. During labor, AA levels in amniotic regulation of COX-2 expression, but were not affected by
fluid increase several-fold and intramniotic administration of antagonists to PLC, PKC and ERK. However, SB203580, a
AA was shown to induce abortion during the second
selective inhibitor of p38 MAP kinase, abrogated thrombin
trimester [197].
action. Thus, the action of thrombin on human myometrial
cells is analogous to that of oxytocin in terms of induction of
Thrombin
COX-2, activation of the phosphoinositide cycle and
Intrauterine bleeding, which is estimated to occur in 15- promotion of contractility. Intracellular signaling, however,
20% of pregnancies, has been recognized by clinicians as a may follow diverging pathways.
risk factor for placental abruption, spontaneous abortion and Thrombin in the plasma is rapidly bound to antithrombin
preterm birth. These pathological conditions are associated (AT) in a 1:1 ratio and the formed TAT complex can be
with increased uterine contractions. Indeed, addition of accurately measured, giving quantitative values for thrombin
whole blood to rat myometrial strips promoted contractility generation. Using this analytical approach, Uszynski [205]
that was attributed to thrombin, a known stimulant of smooth reported a 10-fold increase in TAT levels at the end of
muscle contractions [198, 199]. Subsequent studies by the pregnancy and a further transient 15-fold increase after
same group have demonstrated the presence of prothrombin separation and expulsion of the placenta. Increased TAT
in rat myometrium and its expression in endometrium and levels have also been reported in preterm labor and preterm
placenta, in the absence of bleeding [200]. Importantly, premature rupture of membranes (PPROM), as well as in
thrombin-induced tension in rat myometrium was much patients with small-for-gestational-age fetuses [206, 207]. It
enhanced in pregnant myometrium compared to tissue from has been suggested that the plasma level of TAT may serve
non-pregnant animals. This could be attributed to a to predict PPROM [208].
corresponding rise in [Ca2+]i, and to a 10-fold increase in
thrombin receptor mRNA [201]. These observations, and the Stretch
well-established contractile effect of thrombin in a variety of
other smooth muscle preparations, support the notion of a Mechanoreception is the most broadly evolved signal
functional role for thrombin in the regulation of myometrial recognition in living organisms, from bacteria (turgor
contractions. pressure induces the expression of osmoregulatory genes), to
plants (geotropism) and the animal kingdom [209, 210]. It
The physiologic function of thrombin has long been has been known for a long time that mechanical stretch
attributed to its role as a key intermediate in the coagulation
imposed on skeletal, cardiac or smooth muscle, leads to
cascade. More recently, it has become evident that thrombin
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2511

hypertrophy [211]. In 1965, Csapo et al. placed closed increases exponentially towards the end of pregnancy, but its
liquid-filled rubber balloons into single horns of the physiological role in pregnancy maintenance and parturition
bicornuate uteri of ovariectomized rabbits and observed that remains an enigma. CRH binds to G-protein-coupled recep-
the stretched horns increased in weight compared to the tors, of which two subtypes, R1 and R1, have been identified
contralateral unstretched uterine horns. Based on RNA/DNA and cloned. In addition, a number of isoforms of both
ratio and myometrial contractility they concluded that the receptors, resulting from alternative gene splicing, are known.
weight increase was largely due to de novo protein synthesis Importantly, R1 receptors are significantly upregulated in
[212, 213]. Similar observations were reported using human myometrium at the time of labor, lending support to
continuous distension of non-pregnant rat uteri [214]. the notion that the CRH/R1 system is involved in myo-
Interestingly, mechanical stretch of uterine muscle not only metrial activity. Because CRH-activated receptors are
induces growth, but also prevents postpartum involution coupled to Gαs and the generation of the myometrial relaxant,
undergoing at the same time by the unstretched contralateral cAMP, a role in the maintenance of uterine quiescence,
horn [215]. Using the rat model, the Toronto group extended particularly during the rapid expansion of uterine volume,
these observations in a series of elegant experiments has been suggested. CRH may also promote uterine
(reviewed in [216]), and demonstrated the expression of a relaxation by activating the NO/cGMP signaling pathway
cassette of proteins involved in myometrial activation at [226]. The inhibition of basal IL-1β- and oxytocin-
term, including OTR, Cx43, and PGF2α receptor (FP), that stimulated PGE2 production, as demonstrated in human
could be reproduced by mechanical stretch of the non-gravid myometrial cell cultures [227], could potentiate the relaxant
horn, provided that progesterone withdrawal had occurred. effect. The paradoxical rise in CRH/R1 levels at labor may
Importantly, progesterone administration prevented both the promote the birth process by relaxing the lower uterine
expression of these proteins and the onset of parturition. segment, rather than potentiating uterotonin-driven contrac-
They concluded that the regulation of these key proteins is tions in the fundus [228]. The myometrial relaxant effect
under both hormonal and mechanical factors. In subsequent may also be obtunded by the downregulation of Gαs at the
experiments, Lye and his colleagues have demonstrated that onset of labor [229]. In addition, the effect of CRH may be
the transcription factor AP-1 is upregulated in rat myomet- reduced due to phosphorylation of CRH receptor by
rium shortly before the onset of labor [217]. They also oxytocin-activated PKC [230]. All in all, further evidence is
observed the activation of the MAP kinase family of required in support of a physiological role for CRH in the
enzymes (ERK, JNK, p38) in stretched rat myometrial regulation of myometrial functions.
smooth muscle cells, leading to the expression of the early
gene c-fos [218]. Using selective inhibitors they demons- Nitric Oxide (NO)
trated that the activity of all three MAP kinases are required
NO (originally thought to act only as a potent vasodilator
for the upregulation of c-fos mRNA and that the
released from the endothelium) is a multifunctional signaling
mechanotransduction is linked upstream to tyrosine kinase
molecule widely distributed in the body, affecting a variety
activation. Less is known about the molecular aspects of
of cellular functions, including myometrial smooth muscle
mechanotransduction in human myometrium. Nonetheless,
contractility (for review, see [231]). NO is generated from
several lines of evidence support the concept of stretch-
the amino acid, L-arginine, during its conversion to L-
induced activation of the uterus. The incidence of preterm
labor is markedly higher in multifetal pregnancies, polyhydr- citrulline by NO synthase (NOS). There are three NOS
amnios or somatomegaly in singleton pregnancies [219, isozymes, the Ca-dependent constitutively expressed eNOS
and nNOS and the cytokine-inducible iNOS that does not
220]. Yet in a recent study, Lyall et al. [221] found no
require a rise in Ca2+ for activity. In most systems, NO
difference between the expression of Gαs, gap junction
activates the guanylate cyclase/cGMP signaling pathway,
proteins or PGE2 receptors in mechanically stretched human
leading to relaxation. Several lines of evidence, using
myometrial cells isolated from singleton and multiple
pregnant uteri. Other investigators reported increased Cx43 various NO donors, indicate that in human non-pregnant,
laboring and non-laboring myometrium, NO-induced relaxa-
and c-jun and c-fos expression in term-pregnant human
myometrium [33]. Upregulation of COX-2 and prostaglandin tion is independent from cGMP [232-234]. Interestingly,
human myometrial strips are much less sensitive to the
production appear to be the most consistent responses to
myometrial stretch in both animal and human studies [222, relaxing effect of cGMP than precontracted bovine trachea
smooth muscle [235]. Further, the inconsistent reports failed
223].
to provide concrete evidence to support the role of NO in
In summary, mechanogenetic regulation of protein maintaining uterine quiescence. In one study, Norman et al.
synthesis in the myometrium during gestation and at term [236] found an increase in the constitutive NOS expression
may play important functional roles in both pregnancy in the human myometrium during pregnancy, while a
maintenance (by promoting uterine growth) and parturition subsequent report by the same group failed to observe any
(by inducing the synthesis of key proteins). change in NOS activity in the uterus and placenta during
parturition [237], contrary to the findings of Bansal et al.
Corticotropin Releasing Hormone (CRH) [238], who reported a decline in myometrial NOS expression
associated with labor and delivery. Dennes et al. [239], on
CRH, a hypothalamic peptide regulating pituitary ACTH the other hand, reported that myometrial NOS mRNA
secretion, is also expressed locally in a variety of tissues, expression did not change during gestation or with the onset
including the human placenta and myometrium (reviewed in of labor, nor was iNOS induced by cytokines in cultured
[224, 225]. Its production and release into the circulation cells [240].
2512 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

In a more recent study, Batra et al. [241], using identical channel blocker, nicardipine, for short term tocolysis,
methodologies, compared the cellular distribution, activity Larman et al. [248] found no differences with regard to
(formation of 14C-citrulline from 14C-arginine) and prolongation of pregnancy.
expression (Western blot) of NOS in human myometria and
Calcium Channel Blockers
vagina with those of the rat. They were able to detect only
very low NOS activity in human myometrium compared to Calcium channel blockers (nicardipine, nifedipine) act by
that measured in the corresponding tissue of the rat. preventing calcium entry into myocytes, as well as suppres-
Surprisingly, vaginal NOS activity was 4-5-fold higher than sing intracellular calcium release. In a comprehensive meta-
that found in myometrium of either species. In view of these analysis, Tsatsaris et al. [249] compared nifedipine with β-
observations, the authors suggested that, in human preg- mimetic therapy. The results indicated that the calcium
nancy, the paracrine action of NO derived from the fetal antagonist was more effective in prolonging pregnancy
membranes and placenta may contribute to the maintenance beyond 48h than β-adrenergic agonists, with fewer side
of uterine quiescence. It may be pertinent to note here that, effects, but without affecting neonatal mortality rate.
compared to mouse macrophages, the iNOS gene of human
Non-Steroidal anti-Inflammatory Drugs (NSAID)
macrophages is hyporesponsive to induction by lipopoly-
saccharide (LPS) + interferon-γ + IL-1β cocktail [242]. Indomethacin, an inhibitor of both COX-1 and 2 activity,
However, the same regimen effectively induces iNOS gene has been the most extensively evaluated NSAID for tocolysis.
in human hepatocytes [243]. Attempts to induce the expres- Although indomethacin, or a related drug, sulindac, have
sion of iNOS in human myometrial cells with cytokines and been shown in placebo controlled trials to be effective in
LPS have failed (F. Hertelendy, unpublished). Hence, it prolonging pregnancy for 48h or longer, there are undesir-
appears that the regulation and activity of NOS enzymes in able side effects that include, in addition to the well
both species are cell specific. documented constriction of ductus arteriosus, intraventricu-
lar hemorrhage, olygohydramnios, pulmonary dysplasia and
Tocolysis necrotizing enterocolitis [250, 251]. In a randomized
controlled prospective trial, indomethacin and nylhidrin were
Early diagnosis of preterm labor, a complex syndrome of
found to be equipotent in delaying delivery [252]. Since
multiple etiologies, is critical for the implementation of
labor has been associated with upregulation of COX-2,
appropriate clinical management in an attempt to delay or, if
selective inhibitors targeted at this enzyme may eventually
possible, prevent premature birth, the leading cause of
prove to be more effective tocolytics with fewer side effects.
neonatal morbidity and mortality in developed countries. Indeed, a small recent trial (n=24) found celecoxib to be as
There are a variety of drugs available for pharmacologic
effective as indomethacin in prolonging pregnancy, but
intervention to suppress myometrial contractions and the
without constricting the ductus arteriosus [253]. Evidently,
literature is replete with reports and reviews of clinical trials
larger, well designed multi center trials with clearly defined
evaluating the efficacy, as well as shortcomings, of such
end points are required to evaluate the possible benefits of
therapies. In general, first line (acute) tocolytic therapy may
selective COX-2 inhibitors vs. traditional tocolytics.
prolong pregnancy for 48h or longer, allowing the
administration of corticosteroids to enhance lung maturation Oxytocin Receptor Antagonists
and, if available, for transfer to a tertiary medical facility for It was pointed out elsewhere in this review that OTR
high risk pregnancy. Prolonged tocolytic therapy has little or density in the myometrium increases strikingly as pregnancy
no beneficial effect and may be harmful for both mother and
progresses to term. Hence, the idea that an OTR blocker
fetus.
could prolong pregnancy and delay delivery in uncomplicated
Beta Mimetics preterm labor appeared a promising avenue to pursue. Atosi-
ban, a drug that binds to OTRs (though even more avidly to
Beta Mimetics (Isosuprine, salbutamol, terbutaline, feno- arginine vasopressin receptors) offered such a promise.
terol, hexoprenaline, nylidrin, ritodrin). These drugs activate
However, after large scale multi center, multinational trials
β1 and β2 adrenergic receptors, raising cAMP levels resulting comparing it to placebo or the beta-agonist terbutalin, it was
in reduced intracellular Ca2+ concentration and, ultimately,
concluded that atosiban was not more efficacious in terms of
myometrial contractions. Although beta mimetics have been clinical significance than placebo or terbutalin, although its
used extensively, there is substantial evidence linking such
safety profile was superior to that of the beta agonist [254-
therapy with adverse effects for both mother and fetus
257].
without clinically significant improvement in perinatal or
neonatal outcomes [244,245]. Nitric Oxide
Magnesium Sulfate The rationale to use NO donors stems from animal
experiments, which showed that such compounds relaxed,
Magnesium sulfate acts at high concentrations by and NOS inhibitors promoted, uterine activity. In human,
displacing calcium from the endoplasmic reticulum. It is a
however, the putative role of NO in myometrial relaxation is
popular tocolytic and anticonvulsant agent in the USA, even controversial (see above under Nitric Oxide). Duckitt and
though there is no adequate supporting evidence for its use Thornton [258] have recently reviewed the results of several
for the prevention of preterm labor following acute tocolysis trials, comparing the effects of transdermally or intrave-
[246]. In one trial (n=156) there was no difference between
nously administered nitroglycerine, with placebo or no
magnesium sulfate therapy and placebo in terms of preterm treatment vs. beta receptor agonists (ritodrine, albuterol) and
delivery [247]. Comparing i.v. magnesium with the calcium magnesium sulfate. They found no differences between NO
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2513

donors and placebo or no treament, concluding: “ The nitric CBP = CREB (cAMP-response element-binding
oxide donors appear to have a more favorable side effect protein)-binding protein
profile than some other tocolytic agents but in the absence of
cGMP = Guanosine-3’-5’-cyclic monophosphate
beneficial effects on perinatal morbidity and mortality, their
routine administration cannot be advocated”. Larger, better- COUP-TFs = Chicken ovalbumin upstream promoter-
controlled trials may uncover additional information that associated transcription factors
may modify this view and include NO-donors among the DAG = 1,2-diacyl-glycerol
therapeutic modalities for the treatment of preterm labor.
DHP = Dihydro-progesterone
Taken together, there are no satisfactory tocolytic drugs
that have influenced the outcome of preterm labor during the EGF = Epidermal growth factor
past three decades in the USA. Because preterm labor is EP1, -2, -3, -4
multifactorial, combination therapies may offer some receptors = PGE2 receptor type-1, -2, -3, -4
advantages over single tocolytic therapies. But, ultimately,
elucidating the etiologies of preterm labor may offer more ERK = Extracellular signal-regulated protein
promising avenues for the design of new classes of tocolytic kinase
drugs. FP receptor = PGF2α receptor

CONCLUSION Gαs = G-protein alpha-s

Pregnancy maintenance and parturition are regulated by HOXA-10 = Gene 10 of the homeotic selector
complex interactions of endocrine, paracrine and mechano- complex A
genetic factors. The unraveling of these mechanisms has HSP90 = Heat-shock protein 90
posed a monumental task for reproductive biologists. Much
IL-1, IL-1β = Interleukin-1 (beta)
of our knowledge relies on data from animal experiments,
the results of which cannot always be extrapolated to the IL-6 = Interleukin 6
human. Because of ethical considerations, studies on human IL-8 = Interleukin 8
subjects are limited and information is gathered mainly from
experiments using tissue explants or cultured cells. Whereas IP receptor = Prostacyclin receptor
the molecular mechanism of myometrial smooth muscle IP3 = Myo-inositol 1,4,5-trisphosphate
activity has been quite well mapped out, the intimate
mechanism responsible for converting the quiescent pregnant JAK-2 = Janus tyrosine kinase-2
uterus to the parturient organ remains largely elusive. What LPS = Lipopolysaccharide
has emerged over the last decade or so are the multiple levels
of regulation involving key steps in receptor signaling MAP-kinase,
leading to gene regulation. Compounding the complexity of MAPK = Mitogen-activated protein kinase
normal parturition at term are the multiple etiologies of MLC = Myosine light chain
preterm labor, such as intrauterine infection, psychosocial
stress, multifetal pregnancy, drug abuse, poor prenatal care, NO = Nitric oxide
malnutrition, placental abruption, etc. Future work will have PGD2 = Prostaglandin D2
to focus on early recognition of these risk factors, coupled
PGE2 = Prostaglandin E2
with the development of reliable diagnostic tests. This
should allow early intervention aimed at intercepting key PGF2α = Prostaglandin F2α
upstream steps that would prevent the evolution of labor by, PGH2 = Prostaglandin endoperoxide H2
for example, blocking the synthesis of key proteins involved
in the activation of the myometrium. Additional difficulty in PIP2 = Phosphatidyl inositol-4, 5-bisphopsphate
designing new drugs targeted at key steps in cell signaling SF-1 = Steroidogenic factor-1
pathways may lie in the intrinsic promiscuity of key
enzymes and their multiple isoforms (e.g., PKC, tyrosine SRC-2, -3 = Steroid receptor coactivator-2, -3
kinase, IκB/NF-κB system, MAPK, etc.). A concerted effort TBP = TATA box-binding protein
by pharmacologists, biologists, chemists and clinicians may
bring us closer to understanding the intricate regulation of RT-PCR = Reverse transcriptase-coupled
labor and birth, that may allow the design of new approaches polymerase chain reaction
for the diagnosis and prevention of preterm birth and other STAT-1 = Signal transducer and activator of
pathologies of this reproductive process. transcription-1
TAFs = TBP(TATA-binding protein)-associated
ABBREVIATIONS
factors
AP1, AP2 = Activator protein complex -1, -2 TGF-β1 = Transforming growth factor beta-1
ATP = Adenosine-5’-triphosphate
TNF, TNFα = Tumor necrosis factor (alpha)
cAMP = Adenosine-3’,5’-cyclic monophosphate
TP receptor = Thromboxane A2 receptor
2514 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

REFERENCES [41] Sugimoto Y, Yamasaki A, Segi E, Tsuboi K, Aze Y, Nishimura T,


et al. Science 1997; 277: 681.
References 259-261 are related articles recently published in [42] Inoue T, Kimura T, Azuma C, Inazawa J, Takemura M, Kikuchi T,
Current Pharmaceutical Design. et al. J Biol Chem 1994; 269: 32451.
[43] Adachi S, Oku M. J Smooth Muscle Res 1995; 31: 175.
[1] Colledge M, Scott JD. Trends Cell Biol 1999; 9: 216. [44] Webster MA, Pattison NS, Phipps SL, Gillmer MD. Br J Obstet
[2] Moss SB, Getton GL. Trends Endocrinol Metab 2001; 12: 434. Gynaecol 1985; 92: 957.
[3] Challis JRG, Lye SJ. In The physiology of reproduction, ed. 2, [45] Selinger M, Mackenzie IZ, Gillmer MD, Phipps SL, Ferguson J. Br
Knobil, E, Neil, J.D, Eds, Raven Press: New York USA 1994; J Obstet Gynaecol 1987; 94: 1218.
pp.985-1031. [46] Webster MA, Phipps SL, Gillmer MD. Br J Obstet Gynaecol 1985;
[4] Challis JRG, Matthews SG, Gibb W, Lye SJ. Endocrine Rev 2000; 92: 963.
21: 514.
[47] Avrech OM, Golan A, Weinraub Z, Bukovsky I, Caspi E. Fertil
[5] Terzidou V, Bennett PR. Curr Opin Obstet Gynecol 2002; 14: 105. Steril 1991; 56: 385.
[6] Csapo A. Am J Anat 1956; 98: 273. [48] Brodt-Eppley J, Myatt L. Biol Reprod 1998; 59: 878.
[7] Mitchell BF, Wong S. Am J Obstet Gynecol 1993; 168: 1377. [49] Ou CW, Chen ZQ, Qi S, Lye SJ. Am J Obstet Gynecol 2000; 182:
[8] Kossmann JC, Bard H, Gibb W. Biol Reprod 1982; 27: 320. 919.
[9] Swahn ML, Bygdeman M. Br J Obstet Gynaecol 1988; 95: 126. [50] Dong YL, Yallampalli C. Biol Reprod 2000; 62: 533.
[10] Norman JE, Kelly RW, Baird DT. Hum Reprod 1991; 6: 740. [51] Cook JL, Shallow MC, Zaragoza DB, Anderson KI, Olson D. M
[11] Frydman R, Lelaidier C, Baton-Saint-Mleux C, Fernandez H, Vial Biol Reprod 2003; 68: 579.
M, Bourget P. Obstet Gynecol 1992; 80: 972. [52] Brodt-Eppley J, Myatt L. Obstet Gynecol 1999; 93: 89.
[12] Lelaidier C, Baton C, Benifla JL, Fernandez H, Bourget P, [53] Greenland KJ, Jantke I, Jenatschke S, Bracken KE, Vinson C,
Frydman RJ. Obstet Gynaecol 1994; 101: 501. Gellersen B. Endocrinology 2000; 141: 581.
[13] Vegeto E, Shahbaz MM, Wen DX, Goldman ME, O'Malley BW, [54] Giannoulias D, Patel FA, Holloway AC, Lye SJ, Tai HH, Challis
McDonnell D. P Mol Endocrinol 1993; 7: 1244. JR. J Clin Endocrinol Metab 2002; 87: 1345.
[14] Kazmi SM, Visconti V, Plante RK, Ishaque A, Lau C. [55] Patel FA, Clifton VL, Chwalisz K, Challis JR. J Clin Endocrinol
Endocrinology 1993; 133: 1230. Metab 1999; 84: 291.
[15] Condon JC, Jeyasuria P, Faust JM, Wilson JW, Mendelson CR. [56] Patel FA, Challis JR. J Clin Endocrinol Metab 2002; 87: 700.
Proc Natl Acad Sci USA 2003; 100: 9518. [57] Patel FA, Funder JW, Challis JR. J Clin Endocrinol Metab 2003;
[16] Kalkhoven E, Wissink S, van der Saag PT, van der Burg B. J Biol 88: 2922.
Chem 1996; 271: 6217. [58] Stevenson JC, Macdonald DW, Warren RC, Booker MW,
[17] Katzenellenbogen BS. J Soc Gynecol Investig 2000; 7: S33. Whitehead MI. Br Med J4 (Clin. Res. Ed.) 1986; 293: 1329.
[18] Giangrande PH, Pollio G, McDonnell D. P J Biol Chem 1997; 272: [59] Yallampalli C, Chauhan M, Thota CS, Kondapaka S, Wimalawansa
32889. SJ. Trends Endocrinol Metab 2002; 13: 263.
[19] Shemshedini L, Knauthe R, Sassone-Corsi P, Pornon A, [60] Dong YL, Fang L, Kondapaka S, Gangula PR, Wimalawansa SJ,
Gronemeyer H. EMBO J 1991; 10: 3839. Yallampalli C. J Clin Invest 1999; 104: 559.
[20] Khan-Dawood FS, Dawood MY. Am J Obstet Gynecol 1984; 150: [61] Florio P, Margutti A, Apa R, Miceli F, Pezzani I, Degli Uberti EC,
501. Petraglia FJ. Soc Gynecol Investig 2001; 8: 165.
[21] How H, Huang ZH, Zuo J, Lei ZM, Spinnato JA, 2nd Rao CV. [62] Aiyar N, Rand K, Elshourbagy NA, Zeng Z, Adamou JE, Bergsma
Obstet Gynecol 1995; 86: 936. DJ, et al. Biol Chem 1996; 271: 11325.
[22] Rezapour M, Backstrom T, Lindblom B, Ulmsten U. Obstet [63] Dong YL, Wimalawansa S, Yallampalli C. Am J Obstet Gynecol
Gynecol 1997; 89: 918. 2003; 188: 466.
[23] Garfield RE, Hayashi RH. Am J Obstet Gynecol 1981; 140: 254. [64] Chauhan M, Thota CS, Kondapaka S, Wimalawansa S, Yallampalli
[24] Balducci J, Risek B, Gilula NB, Hand A, Egan JF, Vintzileos AM. C. Peptides 2003; 24: 65.
Am J Obstet Gynecol 1993; 168: 1609. [65] Giannopoulos G, Tulchinsky D. J Clin Endocrinol Metab 1979; 49:
[25] Kilarski WM, Rezapour M, Backstrom T, Roomans GM, Ulmsten 100.
U. Acta Obstet Gynecol Scand 1994; 73: 377. [66] Padayachi T, Pegoraro RJ, Rom L, Joubert SM. J Steroid Biochem
[26] Chow L, Lye SJ. Am J Obstet Gynecol 1994; 170: 788. Mol Biol 1990; 37: 509.
[27] Sparey C, Robson SC, Bailey J, Lyall F, Europe-Finner G. N J Clin [67] Roh CR, Lee BL, Oh WJ, Whang JD, Choi DS, Yoon BK, et al. J
Endocrinol Metab 1999; 84: 1705. Korean Med Sci 1999; 14: 552.
[28] Andersen J, Grine E, Eng CL, Zhao K, Barbieri RL, Chumas JC, et [68] Winkler M, Kemp B, Classen-Linke I, Fischer DC, Zlatinsi S,
al. Am J Obstet Gynecol 1993; 169: 1266. Neulen J, et al. J Soc Gynecol Investig 2002; 9: 226.
[29] Zhao K, Kuperman L, Geimonen E, Andersen J. Biol Reprod 1996; [69] Thomson AJ, Telfer JF, Young A, Campbell S, Stewart CJ,
54: 607. Cameron IT, et al. Hum Reprod 1999; 14: 229.
[30] Ambrus G, Rao CV. Endocrinology 1994; 135: 2772. [70] Pieber D, Allport VC, Hills F, Johnson M, Bennett PR. Mol Hum
[31] Geimonen E, Jiang W, Ali M, Fishman GI, Garfield RE, Andersen Reprod 2001; 7: 875.
J. J Biol Chem 1996; 271: 23667. [71] Mesiano S, Chan EC, Fitter JT, Kwek K, Yeo G, Smith R. J Clin
[32] Echetebu CO, Ali M, Izban MG, MacKay L, Garfield RE. Mol Endocrinol Metab 2002; 87: 2924.
Hum Reprod 1999; 5: 757. [72] Cermik D, Karaca M, Taylor HS. J Clin Endocrinol Metab 2001;
[33] Geimonen E, Boylston E, Royek A, Andersen J. J Clin Endocrinol 86: 3387.
Metab 1998; 83: 1177. [73] Chegini N, Rong H, Dou Q, Kipersztok S, Williams RS. J Clin
[34] Fuchs AR, Fuchs F, Husslein P, Soloff MS, Fernstrom MJ. Science Endocrinol Metab 1996; 81: 3215.
1982; 215: 1396. [74] Hatthachote P, Gillespie J. I Endocrinology 1999; 140: 2533.
[35] Fuchs AR, Fuchs F, Husslein P, Soloff MS. Am J Obstet Gynecol [75] Acevedo CH, Ahmed AJ. Clin Invest 1998; 101: 949.
1984; 150: 734. [76] Barber A, Robson SC, Lyall F. Am J Pathol 1999; 155, 831.
[36] Rezapour M, Backstrom T, Ulmsten U. Steroids 1996; 61: 338. [77] Aguan K, Carvajal JA, Thompson LP, Weiner CP. Mol Hum
[37] Kimura T, Takemura M, Nomura S, Nobunaga T, Kubota Y, Inoue Reprod 2000; 6: 1141.
T, et al. Endocrinology 1996; 137: 780. [78] Chan EC, Fraser S, Yin S, Yeo G, Kwek K, Fairclough RJ, et al. J
[38] Fuchs AR, Fuchs F. Br J Obstet Gynaecol 1984; 91: 948. Clin Endocrinol Metab 2002; 87: 2435.
[39] Mitchell BF, Schmid BJ. Soc Gynecol Investig 2001; 8: 122. [79] Charpigny G, Leroy MJ, Breuiller-Fouche M, Tanfin Z, Mhaouty-
[40] Ivell R, Walther N. Mol Cell Endocrinol 1999; 151: 95. Kodja S, Robin P, et al. Biol Reprod 2003; 68: 2289.
[80] Giles RH, Peters DJ, Breuning MH. Trends Genet 1998; 14: 178.
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2515

[81] Wilson MA, Ricci AR, Deroo BJ, Archer TK. J Biol Chem 2002; [122] Mitchell MD, Romero RJ, Edwin SS, Trautman MS. Reprod Fertil
277: 15171. Dev 1995; 7: 623.
[82] Lofgren M, Holst J, Backstrom T. Acta Obstet Gynecol Scand [123] Challis JR, Lye SJ, Gibb W. Ann. N. Y. Acad Sci USA 1997; 828:
1992; 71: 28. 254.
[83] Perusquia M, Jasso-Kamel J. Life Sci 2001; 68: 2933. [124] Challis JRG, Matthews SG, Gibb W, Lye S. J Endocr Rev 2000;
[84] Fu X, Rezapour M, Lofgren M, Ulmsten U, Backstrom T. Obstet 21: 514.
Gynecol 1993; 82: 23. [125] Challis JR, Sloboda DM, Alfaidy N, Lye SJ, Gibb W, Patel FA, et
[85] Lofgren M, Backstrom T. Acta Obstet Gynecol Scand 1994; 73: al. Reproduction 2002; 124: 1.
186. [126] Keelan JA, Blumenstein M, Helliwell RJ, Sato TA, Marvin KW,
[86] Kofinas AD, Rose JC, Koritnik DR, Meis PJ. J Reprod Med 1990; Mitchell MD. Placenta 2003; 24 Suppl A, S33.
35: 1045. [127] Fukai H, Den K, Sakamoto H, Kodaira H, Uchida F, Takagi S.
[87] Davidson BJ, Murray RD, Challis JR, Valenzuela GJ. Am J Obstet Endocrinol Jpn 1984; 31: 565.
Gynecol 1987; 157: 54. [128] Adelantado JM, Lopez Bernal A, Turnbull AC. Br J Obstet
[88] Thornton S, Terzidou V, Clark A, Blanks A. The Lancet 1999; 353: Gynaecol 1988; 95: 348.
1329. [129] Giannopoulos G, Jackson K, Kredentser J, Tulchinsky D. Am J
[89] Fu X, Backstrom T, Ulmsten U. Gynecol Obstet Invest 1998; 45: Obstet Gynecol 1985; 153: 904.
242. [130] Kennedy I, Coleman RA, Humphrey PP, Levy GP, Lumley P.
[90] Barnafi L, Larraguibel R. Acta Endocrinol (Copenh) 1974; 76: 172. Prostaglandins 1982; 24: 667.
[91] Fu X, Rezapour M, Lofgren M, Ulmsten U, Backstrom T. Obstet [131] Coleman RA, Smith WL, Narumiya S. Pharmacol Rev 1994; 46:
Gynecol 1993; 82: 532. 205.
[92] Grazzini E, Guillon G, Mouillac B, Zingg HH. Nature 1998; 392: [132] Senior J, Marshall K, Sangha R, Clayton JK. Br J Pharmacol 1993;
509. 108: 501
[93] Burger K, Fahrenholz F, Gimpl G. FEBS Lett 1999; 464: 25. [133] Fernandes B, Crankshaw D. Eur J Pharmacol 1995; 283: 73.
[94] Gimpl G, Wiegand V, Burger K, Fahrenholz F. Prog Brain Res [134] Narumiya S, Sugimoto Y, Ushikubi F. Physiol Rev 1999; 79: 1193.
2002; 139: 43. [135] Olson DM, Zaragoza DB, Shallow MC, Cook JL, Mitchell BF,
[95] Astle S, Khan RN, Thornton S. Br J Obstet Gynecol 2003; 110: Grigsby P, et al. Placenta 2003; 24: Suppl A, S47.
589. [136] Matsumoto T, Sagawa N, Yoshida M, Mori T, Tanaka I,
[96] Meis PJ, Klebanoff M, Thom E, Dombrowski MP, Sibai B, Mukoyama M, et al. Biochem Biophys Res Commun 1997; 238:
Moawad AH, et al. N Engl J Med 2003; 348: 2379. 838.
[97] da Fonseca EB, Bittar RE, Carvalho MH, Zugaib M. Am J Obstet [137] Kotani M, Tanaka I, Ogawa Y, Suganami T, Matsumoto T, Muro
Gynecol 2003; 188: 419. S, et al. J Clin Endocrinol Metab 2000; 85: 4315.
[98] Challis JR. Nature 1971; 229: 208. [138] Kotani M, Tanaka I, Ogawa Y, Usui T, Mori K, Ichikawa A, et al.
[99] Puri CP, Garfield RE. Biol Reprod 1982; 27: 967. Mol Pharmacol 1995; 48: 869.
[100] Tulchinsky D, Hobel CJ, Yeager E, Marshall JR. Am J Obstet [139] Wing DA, Goharkhay N, Hanna M, Naidu YM, Kovacs BW, Felix
Gynecol 1972; 112: 1095. JC. J Soc Gynecol Investig 2003; 10: 124.
[101] Darne J, McGarrigle HH, Lachelin GC. Br J Obstet Gynaecol [140] Peri KG, Quiniou C, Hou X, Abran D, Varma DR, Lubell WD, et
1987; 94: 227. al. Semin Perinatol 2002; 26: 389.
[102] Grumbach MM, Auchus RJ. J Clin Endocrinol Metab 1999; 84: [141] Molnár M, Hertelendy F. J Clin Endocrinol Metab 1990; 85: 3468.
4677. [142] Anwer K, Sanborn BM. Endocrinology 1989; 124: 17.
[103] Hall JM, McDonnell D. P Endocrinology 1999; 140: 5566. [143] Lopez Bernal A, Rivera J, Europe-Finner GN, Phaneuf S, Asboth
[104] Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner G. Adv Exp Med Biol 1995; 395: 435.
PJ, et al. Science 1997; 277: 1508. [144] Carsten ME, Miller JD. Am J Obstet Gynecol 1987; 157: 1303.
[105] El-Tanani MK, Green CD. Mol Endocrinol 1997; 11: 928. [145] Deliconstantinos G, Fotiou SJ. Endocrinol 1986; 110: 395.
[106] Lu Q, Ebling H, Mittler J, Baur WE, Karas RH. FEBS Lett 2002; [146] Hertz R, Berman I, Keppler D, Bar-Tana J. Eur J Biochem 1996;
516: 1. 235: 242.
[107] McDonnell DP, Norris JD. Science 2002; 296: 1642. [147] Bhattacharya M, Peri K, Ribeiro-da-Silva A, Almazan G, Shichi H,
[108] McKay LI, Cidlowski JA. Mol Endocrinol 1998; 12: 45. Hou X, et al. J Biol Chem 1999; 274: 15719.
[109] Di WL, Lachelin GC, McGarrigle HH, Thomas NS, Becker DL. [148] Romero R, Mazor M. Clin Obstet Gynecol 1988; 31: 533.
Mol Hum Reprod 2001; 7: 671. [149] Melly RW. Rev Reprod 1996; 1: 89.
[110] Oltra E, Pfeifer I, Werner R. Endocrinology 2003; 144: 3148. [150] Young A, Thomson AJ, Ledingham MA, Jordan F, Greer IA,
[111] Zingg HH, Rozen F, Breton C, Larcher A, Neculcea J, Chu K, et al. Norman JE. Biol Reprod 2002; 66: 445.
Adv Exp Med Biol 1995; 395: 395. [151] a) Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F,
[112] Kimura T, Saji F, Nishimori K, Ogita K, Nakamura H, Koyama M, Greer, et al. Mol Hum Reprod 2003; 9: 41. b). Hou X, Varma DR,
et al. J Mol Endocrinol 2003; 30: 109. Chemtob SJ. Biol Chem 1999; 274: 15719.
[113] Phaneuf S, Europe-Finner GN, MacKenzie IZ, Watson SP, Lopez [152] Mitchell MD, Romero RJ, Edwin SS, Trautman MS. Reprod Fertil
Bernal AJ. Reprod Fertil 1995; 103: 121. Dev. 1995; 7: 623.
[114] Chibbar R, Miller FD, Mitchell BF. J Clin Invest 1993; 91: 185. [153] Todd HM, Dundoo VL, Gerber WR, Cwiak CA, Baldassare JJ,
[115] Chibbar R, Wong S, Miller FD, Mitchell BF. J Clin Endocrinol Hertelendy FJ. Mat Fet Med 1996; 5: 161.
Metab 1995; 80: 567. [154] Pollard JK, Mitchell MD. Am J Obstet Gynecol 1996; 174: 682.
[116] Richard S, Zingg HH. J Biol Chem 1990; 265: 6098. [155] Rauk PN, Chiao JP. Am J Reprod Immunol 2000; 43: 152.
[117] Giannopoulos G, Goldberg P, Shea TB, Tulchinsky D. J Clin [156] Hertelendy F, Romero R, Molnár M, Todd H, Baldassare JJ. Am J
Endocrinol Metab 1980; 51: 702. Reprod Immunol 1993; 30: 49.
[118] Bernard A, Duffek L, Torok I, Kosa Z. Acta Physiol Hung 1988; [157] Belt AR, Baldassare JJ, Romero R, Hertelendy FJ. Soc Gynecol
71: 507. Invest 1998; 5(Suppl.), 64.
[119] Monga M, Ku CY, Dodge K, Sanborn B. M Biol Reprod 1996; 55: [158] Bartlett SR, Sawdy R, Mann GE. J Physiol 1999; 520. 2, 399.
427. [159] Belt AR, Baldassare JJ, Molnár M, Romero R, Hertelendy F. Am J
[120] Slater DM, Dennes W, Campa JS, Poston L, Bennett PR. Mol Hum Obstet Gynecol 1999; 181: 359.
Reprod 1999; 5: 880. [160] Hertelendy F, Molnár M, Romero R. J Soc Gynecol Investig 2002;
[121] Erkinheimo TL, Saukkonen K, Narko K, Jalkanen J, Ylikorkala 9: 215.
OR, Ristamaki K. J Clin Endocrinol Metab 2000; 85: 3468. [161] Hertelendy F, Rastogi P, Molnár M, Romero R. Am J Reprod
Immunol 2001; 45: 142.
2516 Current Pharmaceutical Design, 2004, Vol. 10, No. 20 Hertelendy and Zakar

[162] Tribe RM, Moriarty P, Dalrymple A, Hassoni AH, Poston L. Biol [201] Shintani Y, Hirano K, Nishimura J, Nakano H, Kanaide H. Brit J
Reprod 2003; 68: 1842. Pharmacol 2000; 131: 1619.
[163] Owen J. Hauth JC. Clin Obstet Gynecol 1992; 35: 464. [202] Coughlin SR. Nature 2000; 407: 258.
[164] Lefebvre DL, Giaid A, Bennett H, Lariviere R, Zingg HH. Science [203] Macfarlane SR, Seatter MJ, Kanke T, Hunter GD, Plevin R.
1992; 256: 1553. Pharmacol Rev 2001; 53: 245.
[165] Blanks AM, Thornton S. Br J Obstet Gynaecol 2003; 110: Suppl. [204] Molnár M, Rigó J, Romero R, Hertelendy FJ. Soc Gynecol Invest
20, 46. 2001; 8 (Suppl.) 193.
[166] Schrey MP, Cornford PA, Read AM, Steer PJ. Am J Obstet [205] Uszynski M. Eur J Obstet Gynecol Reprod Biol 1997; 75: 127.
Gynecol 1988; 159: 964. [206] Chaiworapongsa T, Espinoza J, Yoshimatsu J, Kim YM, Bujold E,
[167] Molnár M, Hertelendy F. J Clin Endocrinol Metab 1990; 71: 1243. Edwin S, et al. J Matern Fet Neonat Med 2002; 11: 368.
[168] Phaneuf S, Europe-Finner GN, Varney M, MacKenzie IZ, Watson [207] Chaiworapongsa T, Yoshimatsu J, Espinoza J, Kim YM, Yoon BH,
SP, Lopez Bernal AJ. Endocrinol 1993; 136: 497. Romero RJ. Matern Fet Neonat Med 2002; 11: 362.
[169] Marc S, Leiber D, Harbon S. FEBS lett 1986; 201: 9. [208] Rosen T, Schatz F, Kuczynski E, Lam H, Koo AB, Lockwood CJ. J
[170] Monga M, Campbell DF, Sanborn BM. Am J Obstet Gynecol Matern Fet Neonat Med 2002; 11: 11.
1999; 181: 424. [209] Sachs F. Membr Biochem 1986; 6: 173.
[171] Ohmichi M, Koike K, Nohara A, Kanda Y, Sakamoto Y, Zhang [210] Erdos T, Butler-Browne GS, Rappaport L. Biochimie 1991; 73:
ZX. Endocrinology 1995; 136: 2082. 1219.
[172] Molnár M, Rigó J Jr, Romero R, Hertelendy F. Am J Obstet [211] Reynolds SRM, Kaminester. Am J Physiol 1936; 116: 510.
Gynecol 1999; 181: 42. [212] Csapo A, Erdos T, Mattos CR, Gramss E, Moscowitz C. Nature
[173] Asselin E, Drolet P, Fortier MA. Endocrinology 1997; 138: 4798. 1965; 207: 1378.
[174] Fuchs AR, Rust W, Fields MJ. Biol Reprod 1999; 60: 341. [213] Mattos CR, Kempson RL, Erdos T, Csapo A. Fertil Steril 1967; 18:
[175] Tahara M, Morishige K, Sawada K, Ikebuchi Y, Kawagishi R, 545.
Tasaka K, et al. Endocrinology 2002; 143: 920. [214] Douglas AJ, Clarke EW, Goldspink DF. Am J Physiol 1988; 254:
[176] Moran CJ, Friel AM, Smith TJ, Cairns M, Morrison JJ. Mol Hum 543.
Reprod 2002; 8: 196. [215] Wray S. J Physiol 1983; 340: 525.
[177] Word RA, Kamm KE, Stull JT, Casey ML. Am J Obstet Gynecol [216] Lye SJ, Mitchell J, Nashman N, Oldenhof A, Ou R, Shynlova O, et
1990; 162: 1103. al. L Frontiers Horm Res 2001; 27: 165.
[178] Héluy V, Germain G, Fournier T, Ferré F, Breuiller-Fouché M. Eur [217] Mitchell JA, Lye SJ. Biol Reprod 2002; 67: 240.
J Pharmacol 1995; 285: 89. [218] Oldenhof AO, Shynlova OP, Liu M, Langille BL, Lye SJ. Am J
[179] Osada K, Tsunoda H, Miyauchi T, Sugishita Y, Kubo T, Goto K. Physiol 2002; 283: C1530.
Am J Physiol 1997; 272: R541. [219] Gardner MO, Goldenberg RL, Cliver, SP, Tucker JM, Nelson KG,
[180] Molnár M, Hertelendy FJ. Soc Gynecol Invest 1998; 3: (Suppl.) Copper RL. Obstet Gynecol 1995; 85: 553.
102. [220] Lackman F, Capewell, V, Richardson, B, DaSilva, O, Gagnon, R.
[181] Liberto GD, Dallot E, Eude-Le Parco I, Carbol D, Ferré F, Am J Obstet Gynecol 2001; 184: 946.
Breuiller-Fouché M. Am J Physiol 2003; 285: C599. [221] Lyall F, Lye S, Teoh T, Cousins F, Milligan G, Robson S. J Soc
[182] Nakanishi H, Brewer KA, Exton JH. J Biol Chem 1993; 268: 9194. Gynecol Investig 2002; 9: 299.
[183] Diaz-Meco MT, Dominguez I, Sanz L, Dent P, Lozano J, Municio [222] Manabe Y, Yoshimura S, Mori T, Aso T. Prostaglandins 1985; 30:
MM, et al. EMBO J 1994; 210: 287. 141.
[184] Leitges M, Sanz L, Martin P, Duran A, Braun U, Garcia JF, et al. J [223] Leguizamon G, Smith J, Younis H, Nelson M, Sadovsky Y. Am J
Mol Cell 2001; 8: 771. Obstet Gynecol 2001; 184: 117.
[185] Molnár M, Huszár T, Rigó J, Baldassare J, Hertelendy F. J Soc [224] Linton EA, Woodman JR, Asboth G, Glynn RP, Plested CP, López
Gynceol Invest 1999; 6: (Suppl.) 721. Bernal A. Experimental Physiology 2001; 86.2: 273[.
[186] Eude I, Dallot E, Ferré F, Breuiller-Fouché M. Biol Reprod 2002; [225] Hillhouse EW, Grammatopoulos DK. Reproduction 2002; 124:
66: 44. 323.
[187] Renooij W, Snyder F. Biochim Biophys Acta 1981; 663: 545. [226] Aggelidou E, Hillhouse EW, Grammatopoulos DK. Proc Natl Acad
[188] Tetta C, Montrucchio G, Alloatti G, Roffinello C, Emanuelli G, Sci USA 2002; 99: 3300.
Benedetto C, et al. Proc Soc Exp Biol 1986; 183: 376. [227] Grammatopoulos DK, Hillhouse WW. J Clin Endocrinol Metab
[189] Hoffman DR, Romero R, Johnston JM. Am J Obstet Gynecol 1990; 1999; 84: 2204.
162: 525. [228] Benedetto C, Petraglia F, Marozio L,.Chiarolini L, Florio P,
[190] Zhu Y.-P, Hoffman DR, Hwang W-B, Miyaura S, Johnston JM. Genazzani AR, et al. Am J Obstet Gynecol 1994; 171: 126.
Biol Reprod 1991; 44: 39. [229] Europe-Finner GN, Phaneuf S, Tolkovsky AM, Watson SP, López
[191] Molnár M, Hertelendy FJ. Matern-Fetal Med 1992; 1: 1. Bernal A. J Clin Endocrinol Metab 1994; 79: 1835.
[192] Peplow PV, Mikhailidis DP. Prostaglandins Leukot Essent Fatty [230] Grammatopoulos DK, Hillhouse E. W Endocrinology 1999; 140:
Acids 1990; 41: 71. 585.
[193] Bussolino F, Silvagno F, Garbarino G, Costamagna C, Soldi R, [231] Mail H, Longo M, Saade GR, Garfield RE. Current Pharmaceutical
Sanavio F, et al. J Biol Chem 1994; 269: 2877. Design 2003; 9: 359.
[194] Graber R, Sumida C, Nunez EA. J Lipid Mediat Cell Signal 1994; [232] Hoffman P, Stanke-Labesque F, Fanchin R, Dilai N, Pons JC,
9: 91. Ayoubi JM. Hum Reprod 2003; 18: 148.
[195] Hertelendy F, Molnár M, Rigó J, Jr. Mol Cell Endocrinol 1995; [233] Tichenor, S.D, Malmquist, N.A, Buxton I.L. Cell Signal, 2003; 15,
110: 113. 763.
[196] Blobe GC, Khan WA, Hannun YA. Prostaglandins Leukot Essent [234] Buxton IL, Kaiser RA, Malmquist NA, Tichenor S. Br J Pharmacol
Fatty Acids 1995; 52: 129. 2001; 134: 206.
[197] MacDonald PC, Schultz FM, Duenhoelter JH, Gant NF, Jimenez [235] Word RA, Casey ML, Kamm KE, Stull JT. Am J Physiol 1991;
JM, Pritchard JH, et al. Obstet Gynecol 1974; 44: 629. 260: C861.
[198] Elovitz MA, Saunders T, Ascher-Landsberg J, Phillippe M. Am J [236] Norman JE, Thomson AJ, Telfer JF, Young A, Greer IA, Cameron
Obstet Gynecol 2000; 183: 799. IT. Mol Hum Reprod 1995; 5: 175.
[199] Elovitz MA, Ascher-Landsberg J, Saunders T, Phillippe M. Am J [237] Thomson AJ, Telfer JF, Kohnen G, Young A, Cameron IT, Greer
Obstet Gynecol 2000; 183: 674. IA, et al. Hum Reprod 1997; 12: 2546.
[200] Phillippe M, Wolff D, Saunders T, Thomas L, Chapa J. J Soc [238] Bansal RK, Goldsmith PC, He Y, Zaloudek CJ, Ecker JL, Riemer
Gynecol Investig 2002; 9: 276. RI. J Clin Invest 1997; 99: 2502.
Regulation of Myometrial Smooth Muscle Functions Current Pharmaceutical Design, 2004, Vol. 10, No. 20 2517

[239] Dennes WJB, Slater DM, Poston L, Bennett PR. Am J Obstet [252] Kurki T, Eronen M, Lumme R, Ylikorkala O. Obstet Gynecol
Gynecol 1999; 180: 387. 1991; 78: 1093.
[240] Bartlett SR, Bennett PR, Campa JS, Dennes WJ, Slater DM, Mann [253] Stika CS, Gross GA, Leguizamon G, Gerber S, Levy R, Mathur A,
GE, et al. J Physiol 1999; 15: 521. et al. Am J Obstet Gynecol 2002; 187: 653.
[241] Batra S, Losif C, Al-Hijji J, Larsson I. Reprod Biol Endocrinol [254] Romero R, Sibai BM, Sanchez-Ramos L, Valenzuela GJ, Veille JC,
2003; 1: 10. Tabor B, et al. Am J Obste Gynecol 2000; 182: 1173.
[242] Albina JE, J Leukoc Biol 1995; 58: 643. [255] Valenzuela GJ, Sanchez-Ramos L, Romero R, Silver HM, Koltun
[243] Nussler AK, Di SM, Billiar TR, Hoffman RA, Geller DA, Selby R, WD, Millar L, et al. Am J Obstet Gynecol 2000; 182: 1184.
et al. J Exp Med 1992; 176: 261. [256] European Atosiban Study Group, Acta Obstet Gynecol 2001; 80:
[244] Caughey AB, Parer JT. Semin Perinatol 2001; 25: 248. 413.
[245] Gyetvai K, Hannah ME, Hodnett ED, Ohlsson A. Obstet Gynecol [257] The Worldwide Atosiban versus Beta-agonists Study Group. Br J
1999; 94: 869. Obstet Gynecol 2001; 108: 133.
[246] Crowther CA, Moore V, Cochrane Database Syst Rev 2000. [258] Duckitt K, Thornton S. Cochrane Rev 2002; issue 3.
[247] Cox SM, Sherman ML, Leveno KJ. Am J Obstet Gynecol 1990; [259] Gregg AR. Mouse models and the role of nitric oxide in
163: 767. reproduction. Curr Pharm Design 2003; 9(5): 391-8.
[248] Larmon JE, Ross BS, May WL, Dickerson GA, Fisher RG, [260] Maul H, Longo M, Saade GR, Garfield RE. Nitric oxide and its
Morrison JC. Am J Obstet Gynecol 1999; 181: 1432. role during pregnancy: from ovulation to delivery. Curr Pharm
[249] Tsatsaris V, Papatsonis D, Goffinet F, Dekker G, Carbonne B. Design 2003; 9(5): 359-80.
Obstet Gynecol 2001; 97(Pt.2): 840. [261] Thaler CD, Epel D. Nitric oxide in oocyte maturation, ovulation,
fertilization, cleavage and implantation: a little dab'll do ya. Curr
[250] Rosen LJ, Zucker D, Oppenheimer-Gazit V, Yagel S. Am J Obstet
Gynecol 2001; 184: 1. Pharm Design 2003; 9(5): 399-409.
[251] Macones GA, Marder SJ, Clothier B, Stamilio D. M Am J Obstet.

You might also like