Vous êtes sur la page 1sur 37

Received Date : 04-Feb-2014

Accepted Article Revised Date : 14-Mar-2014

Accepted Date : 28-Mar-2014

Article type : Review Article

Oxytocin: its mechanism of action and receptor signalling in the myometrium

Sarah Arrowsmith PhD, Department of Cellular and Molecular Physiology, Institute of Translational

Medicine, University of Liverpool, Liverpool, UK

Susan Wray, PhD, Department of Cellular and Molecular Physiology, Institute of Translational

Medicine, University of Liverpool, Liverpool, UK

Corresponding author Dr Sarah Arrowsmith

Department of Cellular and Molecular Physiology

Institute of Translational Medicine

University of Liverpool

Liverpool

UK

Telephone: +44 (0)151 794 5341

Fax: +44 (0)151 794 5321

Email: s.arrowsmith@liv.ac.uk

This article has been accepted for publication and undergone full peer review but has not
been through the copyediting, typesetting, pagination and proofreading process, which may
lead to differences between this version and the Version of Record. Please cite this article as
doi: 10.1111/jne.12154
This article is protected by copyright. All rights reserved.
Abstract:
Accepted Article
Oxytocin is a nonapeptide hormone that has a central role in the regulation of parturition and

lactation. In this review, we address oxytocin receptor (OTR) signalling and its role in the

myometrium during pregnancy and in labour. The OTR belongs to the rhodopsin-type (Class 1) of the

G-protein coupled receptor (GPCR) superfamily and is regulated by changes in receptor expression,

receptor desensitisation and local changes in oxytocin concentration. Receptor activation triggers a

number of signalling events to stimulate contraction, primarily by elevating intracellular calcium

(Ca). This includes; inositol-tris-phosphate-mediated store calcium release, store-operated Ca entry

and voltage-operated Ca entry. We discuss each mechanism in turn and also discuss Ca-independent

mechanisms such as Ca sensitisation. Since oxytocin induces contraction in the myometrium, both

the activation and the inhibition of its receptor have long been targets in the management of

dysfunctional and preterm labours respectively. . We discuss current and novel OTR agonists and

antagonists and their use and potential benefit in obstetric practice. In this regard, we highlight the

following clinical scenarios: dysfunctional labour, postpartum haemorrhage and preterm birth.

Short running title: Oxytocin receptor signalling in the myometrium

Key words: Oxytocin, vasopressin, myometrium, labour, tocolysis

Oxytocin overview

In 1906, Sir Henry Dale discovered that extracts from the human posterior pituitary gland were able

to contract the uterus of a pregnant cat (1). The peptide responsible, termed oxytocin, was later

sequenced and synthesised by du Vigneaud in 1953 (2).

This article is protected by copyright. All rights reserved.


Oxytocin has both central and peripheral actions, with roles in many physiological and pathological
Accepted Article processes including reproduction, for example; parturition (which is the focus of this review),

lactation, maternal behaviour, erectile dysfunction and ejaculation. Oxytocin can also modulate

social behaviour via increasing empathy, trust and pair bonding (3). It is a nine amino acid

hypophysial neuropeptide hormone, synthesised by the magnocellular neurons of the supraoptic

and paraventricular nuclei of the hypothalamus, whose axons terminate within the posterior lobe of

the pituitary gland. It is produced as a pre-propeptide which is subjected to a number of

modifications to produce the mature peptide. These modifications take place during axonal

transport towards the terminals, from where it is released via exocytosis into the circulation in

response to a variety of stimuli (4-6). Oxytocin is also synthesised by peripheral tissues including the

lining of the uterus (decidua/uterine epithelium during pregnancy), corpus luteum, amnion and

placenta, as well as by the testes.

Oxytocin’s effects on myometrium

As stimulation of the uterus is one of oxytocin’s longest known functions, its role in parturition in

humans and animals has been studied extensively. It is therefore somewhat surprising that the exact

mechanism of how this stimulation is accomplished has not yet been clearly defined. The oxytocin

receptor (OTR) belongs to the rhodopsin-type (Class 1) of the G-protein coupled receptor (GPCR)

superfamily. In the uterus, the Gαq/11 protein couples to phospholipase -β (PLC-β) which controls the

hydrolysis of phosphoinositide-bis-phoshate (PIP2) into inositol-tris-phosphate (IP3) and

diacylglycerol (DAG). In turn, these control the mobilisation of Ca from intracellular stores

(sarcoplasmic reticulum, SR) and activation of protein kinase type C (PKC), respectively. Release of Ca

from the SR brings about smooth muscle contractions via stimulation of Ca-dependent calmodulin

which in turn, activates myosin light chain kinase (MLCK). Subsequent phosphorylation of the

This article is protected by copyright. All rights reserved.


regulatory myosin light chains by MLCK brings about cross-bridge cycling and generation of force (7)
Accepted Article (see figure).

The above mechanism, i.e. release of Ca from intracellular stores and the Ca-dependent activation of

calmodulin and MLCK is considered the canonical pathway of uterine stimulation by oxytocin.

However when examined more carefully, it is clear that the mechanism of oxytocin’s action is more

complex (8, 9).

Ca entry

There is evidence to suggest that Ca from extracellular sources also contributes to the oxytocin-

induced rise in intracellular free Ca ([Ca]i) as the oxytocin-stimulated rise in [Ca]i is reduced in the

absence of external Ca in immortalized myometrial cells and intact human myometrium (10). A

number of Ca influx mechanisms could be responsible for this.

Voltage-operated Ca channels (VOCCs), also referred to as L-type Ca channels, are the major Ca

channels involved in Ca entry in the myometrium and are essential for phasic, spontaneous activity

(11). Changes in membrane potential such as during action potentials result in opening of these

channels and Ca entry. However, there are conflicting data regarding the contribution of VOCC

activity with oxytocin stimulation in myometrial cells. Early studies showed that oxytocin elicits a

small increase in the inward current which would lead to the opening of L-type channels and thus

increase Ca entry (12). In freshly dispersed myometrial cells from pregnant rats, Arnaudeau et al .,

(1994) (13) also found that the Ca channel blocker oxodipine decreased the calcium transient and

sustained rise in [Ca]i elicited by oxytocin and concluded that Ca influx via VOCCs participates in the

global Ca response induced by oxytocin. The extent of their contribution however has been

questioned. Inoue et al., (1992) showed inhibition of L-type current by oxytocin in pregnant rat

myomterial cells (14), and data from an immortalised myometrial-derived cell line suggest that

oxytocin-stimulated calcium influx is independent of VOCCs, as inhibition of these channels using

This article is protected by copyright. All rights reserved.


nifedipine did not have any effect on the increase in [Ca]i under oxytocin-stimulation (15). However
Accepted Article the latter study can be criticised as the immortalised cells no longer express VOCCs (16). We suggest

it is likely that oxytocin affects L-type channel activity indirectly, such as via the opening of other

cation channels (e.g. Ca-activated Cl- channels) or capacitative Ca entry, which would then lead to

depolarisation and subsequent VOCC opening.

Capacitative Ca entry

There is now substantial evidence to support the hypothesis that oxytocin augments Ca entry via a

process known as capacitative Ca entry (or store-operated Ca entry, SOCE) which is distinct from

voltage operated Ca entry, and describes a process whereby depletion of internal Ca stores is

coupled to Ca entry pathways. It can be stimulated by inhibition of Ca store pumps (i.e. SERCA) e.g.

by thapsigargin or by signal-mediated store Ca release, such as following agonist stimulation (17). A

number of downstream signal effectors resulting from GPCR activation, including IP3 and DAG, are

known to contribute to store-operated Ca entry, both of which are activated in the myometrium in

response to oxytocin. Evidence for SOCE in myometrium comes from a number of studies showing

that the rise in [Ca]i following agonist stimulation was greater in the presence of extracellular Ca

compared to zero-Ca conditions (suggestive of Ca influx). This Ca influx was also insensitive to

nifedipine and therefore was independent of VOCCs. It was blocked by PLC inhibition (18) and by

SKF96365, a Ca channel inhibitor previously used in studies of SOCE (19) and was stimulated by

depletion of the Ca store following inhibition of SERCA (15). Furthermore, the expression of the

transient receptor potential (Trp) superfamily of proteins, notably TRPC subfamily, which are known

to mediate non-selective cation and Ca entry in response to a variety of stimuli in a number of cell

types (20, 21), have been detected in myometrium (22, 23), which further suggests that pathways

for SOCE exist in the myometrium and that they contribute to the mechanism of Ca influx during

oxytocin stimulation.

This article is protected by copyright. All rights reserved.


SR Ca release
Accepted Article
Release of intracellular Ca from stores during oxytocin-induced contractions was suggested from

studies of human myometrium (10). It is difficult however to directly demonstrate SR Ca release in

any tissue (24). In freshly dispersed uterine myocytes however, our group has succeeded in loading

the SR with the low affinity Ca sensitive indicative mag-Fluo-4, and studying the effect of agonists on

luminal SR Ca (25). Combining this with cytosolic loading with the high affinity Ca indicator, fura-2,

allows the simultaneous investigation of changes in both lumenal store and cytosolic Ca

concentrations. Using these methods in rat myometrial cells, both a decrease in luminal SR [Ca] and

a transient rise in cytosolic [Ca] was observed following oxytocin application in zero Ca conditions (9,

25, 26). Thus oxytocin can cause release of SR Ca and a lowering of SR Ca content, which, as

indicated above, will trigger capacitative Ca entry. The dynamics of Ca signalling however will be

different under normal physiological conditions (i.e. presence of external Ca), with a more sustained

rise in cytosolic [Ca] being reported (9), which is indicative of Ca release and entry, followed by L-

type Ca channel entry.

Ca efflux

Another mechanism whereby agonists can affect Ca signals and hence contraction, is by modulating

the efflux of Ca across the plasma membrane. In myometrium this occurs via Na-Ca exchange and

the plasmalemmal Ca-ATPase (27). There is limited evidence to suggest that oxytocin has effects on

Ca extrusion mechanisms via inhibition of Ca-ATPases and Ca efflux from cells, prolonging the

elevation of [Ca]i (28, 29). Given the potential importance of these mechanisms to the ability of

oxytocin to modulate uterine Ca signals and contractility, further studies of these aspects of oxytocin

action are called for. Cytoplasmic calcium will also be lowered after oxytocin stimulation by the SR

Ca-ATPase (SERCA) (25). The effect of oxytocin on SERCA activity appears not be have studied but

inhibition of SERCA leads to increased ca signalling and force in human myometrium (30).

This article is protected by copyright. All rights reserved.


Ca sensitisation
Accepted Article
In uterine smooth muscle cells, the force of contraction depends on the balance between the

activities of MLCK and myosin light chain phosphatase (MLCP) as these dictate the extent of myosin

phosphorylation and hence contraction: Phosphorylation of the regulatory light chains by MLCK is

responsible for bringing about contraction whilst dephosphorylation by MLCP brings about

relaxation (see figure). Oxytocin binding to its receptor also triggers the activation of Rho proteins (a

family of Ras homology proteins) which regulate acto-myosin interactions and Ca sensitivity of the

contractile proteins in smooth muscle. In the myometrium, RhoA is activated primarily via activation

of Gα12/13 proteins, which in turn triggers the activation of Rho kinase (ROCK). ROCK then inhibits

MLCP by phosphorylation of its myosin-binding subunit (MYPT1) (31, 32) thereby also inhibiting the

dephosphorylation of myosin light chains (see figure and (33)). This produces a greater level of light

chain phosphorylation and tension at a given level of Ca. Phosphorylation of myosin to augment

contraction in this Ca-independent manner is known as Ca sensitisation (34). In addition to ROCK,

PKC activated by DAG following oxytocin receptor activation, can also affect MLCP activity either by

phosphorylation of the phosphatase directly, or via CPI-17, a smooth muscle specific inhibitor of

MLCP (35). However to what extent sensitisation is a mechanism of oxytocin stimulation of uterine

smooth muscle cells is debatable. While some data exists to support it, including data from human

studies, (9, 36, 37), these have either not measured [Ca]i or not done so under steady state

conditions. When examined directly, inhibition of Rho-ROCK pathway in the uterus produced only

moderate effects on [Ca]i and force (38), suggesting that unlike vascular smooth muscles, Ca

sensitisation, at least via the ROCK pathway, is little used in myometrium and is unlikely therefore to

play a major role in oxytocin’s action.

In addition to oxytocin's direct influences on the force-calcium relationship in the myometrium,

oxytocin may have other roles which modulate myometrial contraction that are independent of the

contractile apparatus. For example, in baboon corpus lutea, oxytocin was shown to increase

This article is protected by copyright. All rights reserved.


expression of the major gap junction protein Connexin 43 (39). In the myometrium, expression of
Accepted Article these gap junctions facilitates the co-ordination and synchronicity of myometrial contractions which

are needed labour (40). More recently, oxytocin was shown to be associated with nuclear

translocation of the transcription factor, NFAT (nuclear factor of activated T cells). NFAT is activated

following a rise in [Ca]i whereby calcinuerin mediates its dephosphorylation and unmasking of a

nuclear localisation signal. In myometrial cells, oxytocin induced a concentration-dependent

translocation of the transcription factor to the nucleus which was inhibited by oxytocin and

calcineurin inhibitors (41), thus highlighting oxytocin's other potential role as a signal for

transcription.

Oxytocin and other tissues

In tissues such as amnion and decidua (uterine epithelium), evidence for OTR signalling has also

been documented: For example, in the amnion, up-regulation of OTR was reported in rabbits at the

end of pregnancy and oxytocin binding to its receptor was significantly increased in human fetal

membranes with labour onset (42, 43). More recently, Terzidou et al., 2011 showed an increase in

OTR mRNA and protein concentrations in human amnion epithelial cells with the onset of labour,

and incubation of these cells with oxytocin simulated an up-regulation of cyclo-oxygenase 2 and

synthesis of prostaglandin E2, via the extracellular signal-regulated protein kinase signal transduction

pathway (44). Oxytocin gene and receptor expression have also been shown in chorion and decidua

in humans (45). Treatment of human decidua with oxytocin resulted in stimulation of Prostaglandin

F2α production (46, 47). The mechanism for this may involve the mitogen-activated protein kinase

(MAPK) system, possibly attributed to the activities of PKC (48). Prostaglandins themselves are key

modulators of myometrial contraction therefore, it suggests a complimentary role for oxytocin/OTR

signalling in uterine tissues exists whereby oxytocin can interact both directly with the myometrium

in stimulating uterine contractions, but also indirectly, via the formation of prostaglandins in other

tissues.

This article is protected by copyright. All rights reserved.


Regulation of myometrial oxytocin and oxytocin receptors
Accepted Article
The regulation of oxytocin’s uterotonic activity can be considered from two aspects; 1) changes in

the concentration of oxytocin in the circulation and 2) changes in OTR expression or its sensitivity.

We will discuss each in turn next.

Oxytocin concentration

A raised plasma oxytocin concentration at some stage during parturition has been detected for most

placental mammals including humans (49, 50). For many animals, immediately prior to labour onset,

oxytocin is released from the pituitary into the maternal circulation. However, there is no good,

consistent evidence to support a significant increase in maternal oxytocin concentration prior to

labour onset in humans (51-53). Thornton et al., (1992) reported that the progress of labour is not

related to an increase in oxytocin concentration and that uterine contractions are not associated

with changes in its plasma concentration (54). A surge in oxytocin towards the second and third

stage of labour has however been reported, but only in a few women (54, 55). In animals it has been

shown that release of oxytocin from the pituitary is pulsatile, most of which occurs during fetal

expulsion (56, 57). Discrete fluctuations (pulses) in oxytocin concentration have been detected in

humans by some but not others (52, 54). The precise measurement of oxytocin concentration in

labour however, has been fraught with technical difficulties, including the accuracy of the methods

of sampling used, particularly when trying to measure short bursts of peptide release, oxytocin

having a short half life and that it undergoes rapid metabolism and clearance (58). All of which are

likely to have given rise to the wide variation and inconsistencies in oxytocin concentrations being

reported. In addition as noted earlier, oxytocin is also produced locally by intrauterine tissues (45).

Thus the close proximity of these tissues to myometrium (and decidua) suggests that a paracrine OT

signalling system within intrauterine tissues may also contribute to labour onset and/or progression.

This would therefore alleviate the need for a significant change in the maternal circulation (59) and

This article is protected by copyright. All rights reserved.


would provide one explanation for the lack of change in maternal circulation concentration
Accepted Article observed.

Interestingly, circulating oxytocin does not appear to be essential for labour since human labour

occurs normally in cases of maternal pituitary gland dysfunction or in the absence of oxytocin arising

from the fetal circulation such as in fetal anencephaly (60). Oxytocin deficient mice (OT -/-) also

deliver normally (61, 62), suggesting additional pathways to parturition exist (e.g. prostaglandin

signalling, increased coordination of excitability of myocyte through gap junction proteins) and may

be compensating for the functional loss of oxytocin. Interestingly, in the rat, giving low doses of

oxytocin actually produces a delay to subsequent fetal expulsion, and does so more effectively than

administration of high doses of an oxytocin antagonist (63). As noted by Russell & Leng (50) this may

be due to desensitisation of uterine oxytocin receptors, and reduced OTR gene expression (see (64)

and later).

Oxytocin Receptor expression

In the myometrial tissues of humans, OTR mRNA levels and OTR density increase at labour onset

(possible mechanisms for this are discussed later) which is thought to mediate an increase in

sensitivity of the myometrium to oxytocin at term (65-67). OTRs were shown to be low in expression

in early gestation but rise to twelve-fold by 37-41 weeks and to be maximally expressed after labour

onset. Furthermore, OTR numbers in the myometrium are increased in preterm as well as term

labour (65). In rats, OTR expression is significantly lower mid-gestation compared to non-pregnant

levels but increases significantly alongside other uterine stimulant systems at parturition (68).

Similarly to oxytocin production, expression of OTRs is not confined to the myometrium. Chorio-

decidual tissue OTR expression also increases during parturition (69) and spatial differences exist

between OTR expression within uterine tissues. For example in the myometrium, OTR expression

was found to be higher in the fundus compared to the lower segment (65, 70). It is suggested that

This article is protected by copyright. All rights reserved.


this spatial distribution would allow for an increase in contractile force arising from the fundal region
Accepted Article whilst the lower segment remains relaxed and therefore would aid fetal decent and passage during

labour. Thus, it is now thought that a change in the receptor expression (amount and location)

rather than concentration of the peptide, is important for labour onset and progression (59).

Post partum, OTR levels decline rapidly in both the chorio-deciual tissues and uterus in rats (71). In

contrast, OT binding sites in the mammary glands remain up-regulated postpartum during the

ensuing lactation period (72) It is suggested that this down regulation of receptors in the uterus may

be needed to prevent unwanted uterine contractile activity in response to oxytocin production

during lactation.

In addition, there is also a central role for oxytocin in the timing of parturition in rats. Neurons within

the CNS also express OTRs and there is dynamic change in OTR mRNA in the brain perinatally, with a

marked increase in specific brain regions such as the supra-optic nucleus, prior to labour (73).

Oxytocin is able to facilitate its own release and can act centrally as a neuromodulator or

neurotransmitter For example, it has recently been shown to cause a shift in neuronal GABA

signalling that might be relevant to the aetiology of autism (74). It can also control the pathways

mediating sensory input from the uterus e.g. cervical stretch, and uterine afferents that terminate

within the brainstem are thought to aid a positive feedback loop (also known as the Ferguson reflex)

to further stimulate oxytocin release (75). That OTRs are expressed in the brain also provides further

evidence for a central role of oxytocin in maternal behaviour such as mother-child bonding.

Regulation of receptor expression

Despite the marked increase in both OTR mRNA and protein with parturition being one of the most

consistent findings in all models examined, the mechanisms regulating myometrial OTR expression

in the human still remain largely undefined, but are likely to be multifactorial.

This article is protected by copyright. All rights reserved.


Steroids: It is suggested that the receptor mRNA concentration is hormonally regulated as in
Accepted Article rodents, there is a good correlation between circulating steroid concentrations and OTR expression.

The general consensus is that oestrogens increase the amount of uterine OTR mRNA and the

number of OTR binding sites whilst conversely, progesterone suppresses OTR expression: Use of the

oestrogen receptor antagonist tamoxifen, was shown to delay parturition by 24 hours and was

associated with a delay in the normal increase in OTR levels in rats (76), whilst administration of the

progesterone receptor antagonist RU486 caused a rapid and marked increase in OTR expression (77,

78). In humans and ruminants however, the mechanism of steroid regulation of OTR expression is

not fully understood and its importance is contested by some particularly because there is no

evidence to support a significant change in circulating oestrogen or progesterone concentration, or

a change in the oestrogen:progesterone ratio, such as seen in rodents (80). Instead, it is thought that

humans undergo a 'functional progesterone withdrawal' which is likely to result from local

progesterone metabolism (81) a shift in the ratio of the expression of progesterone receptor

isoforms (PR-A/PR-B) (82) and altered progesterone receptor co-factor expression (83).

Cholesterol is one of the most abundant lipids within the membrane and has a major role in

determining membrane fluidity and thus the function and organisation of membrane proteins,

including GPCRs (84). There is now a body of evidence to show that the plasma membrane

cholesterol content can regulate the ligand binding affinities and stability of the OTR and therefore

affect receptor signalling (85, 86). Oxytocin receptors preferably localise to cholesterol-rich domains

(also known as lipid rafts) which are often enriched with the protein caveolin, forming the plasma

membrane invaginations known as caveolae (87). It is when localised to within these cholesterol-

rich domains that OTRs show a higher affinity for oxytocin binding (88).

In the myometrium, modulation of membrane cholesterol which subsequently disrupts raft and

caveolae formation causes profound changes in myometrial contractility, including contractions

This article is protected by copyright. All rights reserved.


stimulated by oxytocin (89-91). That the oxytocin receptor is regulated by the biophysical properties
Accepted Article of the membrane therefore suggests that any perturbations to this may have important implications

for parturition, for example labours in obese women where serum cholesterol is predicted to be

high.

Interestingly, progesterone has been shown to inhibit cholesterol esterification and transport to and

from the plasma membrane, which would lead to a reduced cholesterol content within caveolae.

Thus a continuous presence of high progesterone concentration, such as during gestation, would

maintain the OTRs in a low affinity state. However, a subsequent progesterone withdrawal (such as

observed in rodents) would restore cholesterol transport and thereby also increase cholesterol

within caveolae. The responsiveness of the receptors to oxytocin would then increase as they would

switch to their high affinity state (8). This may aid the explanation of the increase in OTR

responsiveness towards labour onset in rodents. However, given that humans undergo a functional

progesterone withdrawal, progesterone-regulation of cholesterol and its relationship to activities of

the OTR in humans warrants further attention.

Stretch: As well as steroidal regulation of receptor expression, stretch of the myometrium is also

considered to be a major stimulus for increased OTR expression and is therefore thought to be a

further level in the control of uterine activity. Studies of animals with bicornuate uterus (e.g.

rodents) (78, 92) or double uterus (e.g. wallabies) (93) have shown that only in pregnant horns or

uterus (subjected to stretch from the growing fetus), was there an increase in the OTR. No increase

was observed in the non-pregnant horn demonstrating that endocrinological changes alone were

not sufficient to stimulate OTR expression and that mechanical stretch is needed. In studies of

primary cultures of human uterine myocyte cells obtained from non-labouring women at late

gestation, stretch also caused an increase in OTR expression. However this was not observed from

cells obtained during labour or from non-pregnant women (94) and may therefore indicate a

This article is protected by copyright. All rights reserved.


gestational dependent effect of stretch on induction of OTR expression or reflect the beginning of
Accepted Article down regulation of receptor expression that occurs post labour.

Increased or aberrant uterine distension may also help explain the high rates of preterm labour

often associated with cases of polyhydramnios (95) and multiple pregnancy (96-98), particularly, if

the uterus is subjected to higher degree of stretch at an earlier time point in gestation. Interestingly,

our recent in vitro studies of myometrial contraction from twin pregnancies showed an increased

response to oxytocin compared to singleton pregnancies (97) although oxytocin expression was not

examined. The stimulus for increased OT expression in found in preterm labours where they are not

confounded by a large fetus or multiple pregnancy (i.e. a relatively unstretched uterus) however is

not known.

Given the relationship between increased stretch and OTR expression, one may expect that

myometrium from prolonged pregnancies (given their extended gestation and associated higher

birth weight babies, hence greater stretch), would have a greater OTR expression and therefore

higher sensitivity to oxytocin. However, the expression of OTRs in cases of failed induction of labour

and postterm pregnancies (43-46 weeks) was shown to be significantly lower than in spontaneous

labour at term (65). In vitro, we also showed that the oxytocin response of the myometrium from

women with postdates pregnancy (<41weeks +3 days) in labour was significantly lower compared to

myometrium from women at term in labour (99) which may be as a consequence of reduced OTR

expression in these tissues.

Desensitisation

Repeated or prolonged stimulation of some GPCRs results in the loss of hormonal responsiveness,

known as desensitisation. The molecular mechanism responsible for desensitisation is mediated via

the recruitment of β-arrestin to the receptor following agonist stimulation. Following agonist

This article is protected by copyright. All rights reserved.


binding, GPCR kinases (or other kinases such as PKC) phosphorylate the C-terminal tail of the
Accepted Article receptor causing β-arrestin recruitment, which then sterically hinders any additional G protein

coupling and thus diminishes further second messenger signalling (100). Often, receptors then

become internalised via the classical clathrin-mediated pathway where they are then either targeted

for recycling to the cell membrane or degradation by lysosomes. Continuous and prolonged

exposure of myometrial cells to oxytocin in vitro leads to a reduced response (64, 101) which is

associated with a loss of OT binding sites, decreased receptor mRNA as well as receptor

internalisation (102). A loss of oxytocin receptors has also been demonstrated during oxytocin-

induced and oxytocin-augmented labours (103) suggesting that receptor downregulation also occurs

in vivo which may have important clinical consequences (discussed later).

Cross- talk between oxytocin and arginine vasopressin receptors

Arginine vasopressin (AVP, also known as antidiuretic hormone and simply, vasopressin) is another

cyclic neurohypophysial peptide. As its name suggests, its major functions are to retain water and

constrict blood vessels. It is produced within magnocellular neurons of the hypothalamus which are

situated adjacent to the neurons that produce oxytocin. AVP is also released from the posterior

pituitary. In humans, the oxytocin and vasopressin peptides are structurally very similar, each

having nine amino acids and a disulfide bond, and differing by only two amino acids (in positions 3

and 8). It also has a short half life (~20 minutes) (104, 105) and is derived from a preprohormone

(106). Like oxytocin, AVP also signals through GPCRs (V1aR, V1bR and V2R) which have a relatively high

homology to OTRs. V1aRs couple to Gαq/11 proteins and mediate effects via activation of PLC-β whilst

V2Rs selectively couple to Gαs proteins which activate cyclic AMP (cAMP) signalling. V1bR (also known

as V3Rs) can activate several signalling pathways via different G-proteins, according to the level of

receptor expression and concentration of vasopressin e.g. PKC via Gαq/11 and cAMP via Gαs (107). The

structural similarity of the two peptides (~80% homology) as well as high sequence homology of the

This article is protected by copyright. All rights reserved.


receptors, specifically around the extracellular binding domain, cause some cross reactivity between
Accepted Article peptides and receptors. That is, studies of receptor binding show that AVP binds to its own receptor

and to the OTR, and in a similar fashion, oxytocin binds mainly to the OTR but can also, to some

extent activate vasopressin receptors (108, 109). Oxytocin and vasopressin receptors can also form

functional homo- or heterodimers (110) adding further complexity into this system.

That AVP has been shown to induce contraction of the uterus in a number of animals including

human (111-114), suggests a physiological role for this peptide in the myometrium exists; the nature

of which however is unclear. Human myometrium is more sensitive to AVP than to oxytocin, which

has been demonstrated in both in vitro (114) and in vivo studies (111, 115, 116). The effect is

thought to be mediated via the V1aR which, as well as the uterus, is also expressed in smooth muscle

cells of blood vessels, liver, adrenal cortex, brain and platelets. The V1bR is expressed within the

adrenal medulla, anterior pituitary and other parts of the brain whilst the V2R is mainly located to

the kidneys where it mediates AVP's antidiuretic effects. As with the OTR, there is some evidence to

suggest that the density of V1aR is moderately increased at latter weeks of gestation (117) and

expression of V1aR has also been reported in preterm labour tissues (114, 115, 118), however a

change in receptor expression has not been reported by all (119).

What is consistent between studies however is that the V1aR expression remains high in the

myometrium throughout gestation, which suggests that a biologically active role for AVP in the

pregnant myometrium does exist. This is further supported by the clinical observations that infusion

of AVP is able to induce labour whilst a 24 hour water fast was also able to increase delivery rates

(120). That both OTR and V1aRs are highly expressed in the myometrium however, has also posed

the question of which peptide and or receptor is (most) responsible for contraction and/or

parturition? (58).

This article is protected by copyright. All rights reserved.


Similarly to oxytocin, there appears to be no increase in AVP concentration in the maternal
Accepted Article circulation prior to labour onset. However this does not rule out a role for local peptide synthesis

and paracrine signalling, such as AVP arising from the fetus. Amniotic fluid AVP concentration has

been reported to increase which is likely to be from fetal origin. Moreover, fetal AVP is also known

to be produced during progression of established labour (121) and in response to fetal stress such as

fetal hypoxia (122). This increase in local formation however, occurs well after the onset of labour,

thus the role of AVP in initiation of parturition is still questioned.

The cross reactivity of receptors and peptides also has important consequences for pharmacologic

therapies employing OTR receptor antagonists (discussed later). That the uterus expresses both OTR

and V1a R and that they are functionally coupled to myometrial contraction, also poses the question

of whether antagonism of both receptors is required for effective tocolysis, such as for preterm

labour (discussed later). The relative importance of the two receptors for labour can probably not be

fully understood until more specific receptor-subtype ligands have been developed and tested in

humans.

Clinical uses of oxytocin receptor agonists and antagonists

Since oxytocin induces contraction in the myometrium, both the activation and the inhibition of its

receptor have long been targets in the management of dysfunctional and preterm labours

respectively. Clinically, synthetic oxytocins (Syntocin and Pitocin) have been developed for use in

labour induction and augmentation whilst a synthetic analogue of oxytocin (Carbetocin) is used to

prevent uterine atony and haemorrhage following caesarean section.

This article is protected by copyright. All rights reserved.


Oxytocin agonists, labour induction and augmentation
Accepted Article
The goal of labour induction is to achieve vaginal delivery by stimulating uterine contractions prior to

spontaneous labour onset and is indicated when the benefits of early delivery outweigh the harms

associated with continuation of pregnancy. The role of oxytocin as an inducing agent is not to be

confused with its role in the augmentation of labour (discussed later). It is estimated that 20% to

30% of women will receive labour induction. Of these, less than two thirds will give birth without

further intervention and 22% will require caesarean section delivery (123). Oxytocin has been used

alone, in combination with amniotomy (breaking of the amniotic membranes), or following cervical

ripening with other pharmacological (e.g., Prostaglandin treatment) or non-pharmacological

methods. However studies suggest that intravenous oxytocin alone should not be used for induction

of labour, since when compared with vaginal PGE2 as an inducing agent, oxytocin alone results in

fewer vaginal deliveries within 24 hours, a lower Bishop score (assessment of cervical favourability,

(124)) at 24 hours and more caesarean section deliveries (125). The current use of oxytocin infusion

is secondary to prostaglandin treatment, after sufficient effacement/ripening of the cervix has been

achieved (123).

Dystocia is a perturbation in the normal progression of labour and has complex causes but inefficient

uterine action is the end result. There is natural variation in the length of labour, thus there is no

‘normal length’ per se. First labours last, on average around 8 hours (unlikely to last >18hours) and

second labours last around 5 hours (unlikely to last >12 hours) (126). Assessment of labour

progression is multifactorial including degree of dilation, fetal rotation, head decent and strength,

duration and frequency of uterine contractions. It is suggested that up to one third of women can

experience delay in labour (127).

Poor progress in labour can be managed medically with uterotonic agents, assisted fetal extraction

or failing this, caesarean section. Currently oxytocin is the only pharmacologic treatment available

This article is protected by copyright. All rights reserved.


for dysfunctional labour and unfortunately this only works in around 50 % of cases (128). Moreover
Accepted Article the prediction of success is complex, requiring assessment of a number of fetal and maternal factors

including obstetric history, gestational age of fetus, fetal presentation and cervical status. Labour

augmentation may be necessary when there is a failure of cervical dilatation or fetal descent with

spontaneous uterine contractions. The goal of labour augmentation with oxytocin is therefore to

enhance inadequate uterine contractions to achieve vaginal delivery. The National Institute of

Clinical Excellence guidelines state that oxytocin should be delivered as a continuous infusion with

incremental doses (given at intervals no more frequent than 30mins) until adequate contractions are

achieved, typically 4-5 contractions in 10 minutes (126).

Although oxytocin is widely used in obstetric care, there is a lack of consensus with respect to its

optimal dosage, timing (early or delayed administration) safety and efficacy. Early

radioimmunoassay studies suggested the half life of oxytocin to be around 3-6 minutes (129) which

gave recommendations for increases of infusion rates to be every 15-20 minutes. More recent in

vivo studies however, found that steady-state plasma oxytocin concentration and maximal uterine

contractile response was achieved after 40 minutes infusion, suggesting that oxytocin's half-life is

more likely to be between 10 and 15minutes (130), and intervals of 30-40 minutes are more superior

in terms of efficacy and safety (131).

Howclinicians should administer oxytocin to women for labour induction and augmentation, and the

monitoring of its effects is therefore an important obstetrical issue (132). A number of studies have

shown that vaginal birth can be achieved with a cervical dilatation rate of 0.5-1cm per hour.

However, for some women, progress may be slower but still remain within normal limits. Some

therefore believe patience with ongoing fetal assessment may also be beneficial and would avoid

unnecessary augmentation for longer periods than necessary (133). As discussed above,

desensitisation of the OTR can arise after prolonged exposure to oxytocin. Clinical studies have

demonstrated a loss of uterine activity, measured via intrauterine pressure catheter recordings in

This article is protected by copyright. All rights reserved.


women undergoing labour augmentation (134), which may account for reduced efficacy of oxytocin.
Accepted Article That the OTRs become desensitised, such as following prolonged labour augmentation, also poses a

risk for uterine atony and haemorrhage in the peripartum period (135), discussed later.

Each contraction-relaxation cycle of the uterus causes an intermittent decrease or interruption in

blood flow which decreases oxygen exchange between the mother and the fetus (136). Under

normal physiological conditions, the feto-placental unit can tolerate these changes, and the greater

the interval between contractions, the greater the time to perfuse the placenta and deliver oxygen

to the fetus (137). Oxytocin infusion however can lead to uterine hyperstimulation. If uterine activity

is excessive and the effects exceed the ability of the feto-placental unit to compensate, blood

flowing to the placenta and oxygen delivery to the fetus is reduced which can lead to intrapartum

fetal hypoxemia or distress. Oxytocin is also capable of causing tetanic contractions, which can result

in fetal death (138-140).

There is much variation in the responsiveness of women to oxytocin, as evident from studies

showing that one dose for one may not have the same outcomes for another. Kimura et al.,

demonstrated that the reaction to the uterus to oxytocin even within the same patient could vary

from one day to the next (141). Some have suggested that the uterus should perhaps be rested in

labour, rather than continuous infusion that results in little progress (142). Moreover, the nature of

oxytocin release from the pituitary is pulsatile and therefore, a more physiological approach to

oxytocin administration ( such as a pulsatile infusion) has also been suggested and trialled (143-145).

Postpartum haemorrhage

Post delivery of the fetus and placenta, forceful uterine contractions are required to clamp uterine

blood vessels and stem bleeding. Thus oxytocin’s endogenous role in labour is also extended to

This article is protected by copyright. All rights reserved.


reinforcing contractions after delivery. Postpartum haemorrhage (PPH) occurs in up to 15% of
Accepted Article vaginal deliveries (146) and represents the most important cause of maternal morbidity and

mortality worldwide. Excessive bleeding during or postpartum in most cases is a result of uterine

atony, therefore the prophylactic use of uterine contractants to enhance myometrial contraction

and uterine retraction have long been used in the prevention of PPH. Oxytocin has been most

widely used but it's relatively short half-life requires continual intravenous infusion, as is the case for

labour augmentation. Syntometrine is an oxytocin/ergot alkaloid combination drug which is

frequently used in vaginal deliveries. It has the rapid uterotonic activity of oxytocin combined with

the prolonged effects of ergometrine, however it is associated with more side effects due to the

activity of ergometrine on other smooth muscles (147).

More recently, Carbetocin, a synthetic analogue of oxytocin with a suggested half life approximately

4-10 times longer than that reported for oxytocin (148), has been developed for prevention of PPH.

It has been shown to have a greater safety and tolerability profile compared to oxytocin with the

added benefit that it can be administered as a single dose injection rather than long term infusion as

required for oxytocin. Today, carbetocin is often the primary treatment administered after delivery

to prevent PPH. It has been shown to be associated with less blood loss compared to syntometrine

in women who have vaginal deliveries, and has significantly fewer adverse effects (148, 149). It is

also routinely administered prophylactically following caesarean section delivery to prevent severe

blood loss and therefore reduce maternal morbidity and mortality.

Novel receptor agonists

In 1909, shortly after its discovery, oxytocin (pituary extract) was used for uteronic action in the

treatment of postpartum haemorrhage (150). Fifty years later it was then used for labour induction

(151). Since then however, little has changed, with only the synthetic oxytocins and oxytocin

This article is protected by copyright. All rights reserved.


analogues being developed and making it into clinical practice. This has prompted a shift in the
Accepted Article focus of uterine stimulants from other sources (152). Natural cyclotides, as well natural plant

extracts have shown some oxytocic activity and may therefore provide a novel source of uterine

stimulants (see (153, 154)). In a study of pomegranate seed extract, for example, a large stimulation

of rat uterine contractility was found (155). These authors went on to show that this effect was due

to β-sitosterol and inhibition of SERCA (and K channels) which is one of the potential mechanisms of

oxytocin’s action. In a review of uterotonic plant preparations in sub-Saharan Africa, Tripathi et al.,

(2013) identified 208 plant species (156). Thus the possibility of some of these being novel oxytocics

is strong. The development of OTR ligands that prevent receptor desensitisation may also be a novel

approach in the treatment of adverse clinical events such as post partum haemorrhage following

prolonged oxytocin therapy.

Oxytocin antagonists and preterm birth

Preterm birth (birth before 37 weeks gestation) is the most important cause of perinatal morbidity

and mortality. Early uterine contractions are one of the most recognised signs of spontaneous

preterm labour. The main method for delay of preterm birth therefore often involves

pharmacological inhibition of myometrial contractions (tocolysis). The aim of tocolytic treatment is

to delay labour sufficiently (24-48 hours) such that the mother can be safely transferred to a

specialist unit with appropriate neonatal care facilities and for the giving of antenatal corticosteroids

to aid fetal lung maturation and increase survival. There are several classes of tocolytic available.

Each has a different mechanism of action and have been reviewed elsewhere (157, 158).

There is some evidence for a premature activation of oxytocin secretion in preterm birth, suggesting

a pathogenic role for it in preterm labour. In this regard, OTR antagonists are one class of treatments

that have been developed as tocolytic agents for women in preterm labour. Oxytocin-receptor

This article is protected by copyright. All rights reserved.


antagonists are also useful for obtaining acute tocolysis in term labour, in situations when there is a
Accepted Article need to stop labour e.g. fetal distress (discussed elsewhere, (159)). Atosiban (Tractocile) is a mixed

oxytocin/vasopressin receptor competitive antagonist. That is, it has actions on both the OTR and

vasopressin (V1a) receptor. Its structure is based upon the amino acid structure of oxytocin except

with modifications at position 1, 2, 4 and 8 and it competes and thus, blocks oxytocin binding.

However, given its lack of receptor specificity, it is not known whether (or how much) the

antagonistic activity of atosiban on the V1aR also contributes to its tocolytic properties. In clinical

studies, atosiban has been shown to be as efficacious as other tocolytics in the treatment of

spontaneous preterm labour (160-162) and in the case of β2-adrenergic agonists, atosiban is thought

to have less unwanted side effects (160). Despite this, a systematic review of the evidence did not

find atosiban to be any more superior to other tocolytics towards neonatal outcomes (163, 164).

Furthermore, atosiban has limited bioavailability and requires parenteral administration and

hospitalisation. Its low affinity for the OTR and antagonistic activities at the V1aR, has ignited

interest in the development of other peptide and non-peptide antagonists with greater specificity

for the OTR.

One of the more OTR-selective competitive antagonists barusiban (Ferring), when tested, showed a

greater specificity for the human OTR than either the V1aR or V2R respectively (165) with greater

potency and longer duration of activity than atosiban. In in vitro experiments using isolated

myometrium from term and preterm women, barusiban was shown to be as effective as atosiban in

inhibiting oxytocin- induced myometrial contractions (166). Non-human primate studies also

showed that barusiban was successful in suppressing oxytocin-induced uterine activity (167, 168).

However, in a proof of concept clinical trial vs. placebo, barusiban was not found to be any more

effective in stopping preterm labour in pregnant women at late gestational age and no reduction in

uterine contractility was observed (169). At present barusiban is being trialled for reducing

implantation failure (due to uterine contraction) during embryo transfer for women undergoing

This article is protected by copyright. All rights reserved.


fertility treatment (In vitro fertilisation or intra-cytoplasmic sperm injection) (170). Results have not
Accepted Article yet been published.

Interestingly, whilst barusiban has not been proven to be effective in inhibiting labour at late

gestation, the more V1aR specific antagonist, relcovaptan has been reported to inhibit contractions in

women with preterm labour (171). However, women also received rescue tocolysis after 2 hours,

and delay to delivery was not measured in this group. This drug may have better clinical uses in the

treatment of dysmenorrhoea (172).

There is clearly need for more efficacious oxytocin (and perhaps vasopressin) receptor agonists with

more specific pharmacological profiles and greater bio availabilities. With this in mind

pharmaceutical companies and researchers have now expanded their searches towards

development of non-peptide oxytocin antagonists, having anticipated them having a greater bio

availability orally than former peptide forms (see Manning et al., 2012 (173)). The 4 most described

are GSK 221149A also known as Retosiban, L368899 (Merck), SSR126768 (Sanofi-Aventis) and

WAY1627720 (Pfizer). Disappointingly both L368899 and WAY162770 failed in clinical and pre-

clinical development for treatment of preterm labour respectively. L368899 showed poor oral

bioavailability and pharmacokinetic profile as well as unwanted behavioural side effects when tested

in rhesus monkeys (174). However, whilst WAY 1627720 failed in preclinical development, it may

have potential as a research tool, particularly in the study of oxytocin activity in the CNS, as

described for other OTR antagonists (173). SSR126768 showed uterine-relaxing properties in vitro

and in vivo in various rat and human models (175). As of January 2008, SSR126768 was still under

active development (phase 1 trials) (176).

The most promising non-peptide OT antagonist for use in treatment of preterm labour is Retosiban.

Preliminary studies in rats showed it had a higher affinity for OTR than V1aR or V2R and in in vivo

studies also in the rat, it was effective in reducing oxytocin-induced contractions and spontaneous

This article is protected by copyright. All rights reserved.


contractions when tested in late gestation (d19-d21) (177). More recently, the efficacy and safety of
Accepted Article retosiban has been tested in a multicentre, randomised control phase 2 clinical trial in women with

spontaneous preterm labour (between 30 and 36 weeks). Women received either IV infusion of

retosiban for 48 hours or placebo. Results indicated that retosiban was associated with a significant

increase in time to delivery (mean increase of 8.2 days) and decrease in preterm birth rate. The

incidence of preterm birth in the retosiban group being 18.7% compared to 47.2% in the placebo

group with an associated relative risk of 0.38 (95% CIs 0.15, 0.81) for preterm birth in the retosiban

group (178). Furthermore, the safety profile of retosiban was favourable and recommendation by

the researchers for progression into phase 3 studies was given. Thus it remains to be seen how

effective this compound will be and how valuable in clinical practice.

Future

It is suggested that up to one third of women will require augmentation of labour, which is likely to

take the form of an oxytocin agonist. However oxytocin is reported to only be effective in around

50% of cases (128). When compared to other medical situations such as cardiac disease where there

is a plethora of treatments available, one treatment which at best works 50 % of the time for women

in labour is lamentable. Likewise for preterm labour, atosiban is the only oxytocin receptor

antagonist available clinically as a tocolytic in Europe, but it is without Food and Drug agency

approval in the US due to its lack of efficacy over other treatments and no evidence of improvement

in neonatal outcomes. As a result, there is increasing interest in neurohypophysial peptide research,

particularly towards the design of more selective OTR and AVPR agonists and antagonists in both

peptide and nonpeptide forms (see Manning et al., 2012 (173)). In addition, oxytocin analogues with

greater bio-availabilities and stability ex vivo also offer the potential for improved treatment

strategies in lower resource settings. However, questions over the role and contribution of the

peptides oxytocin and vasopressin and their receptors in preterm and term labour still remain.

This article is protected by copyright. All rights reserved.


Answers require pharmacological input coupled with basic science research to unravel the complex
Accepted Article roles of these neurohypophysial peptides in parturition.

Figure 1. Oxytocin receptor signalling in the myometrium leading to contraction. Binding of

oxytocin to its receptor activates Gαq/11 which activates PLC-β, which in turn hydrolyses PIP2 to IP3

and DAG. IP3 causes release of Ca from the sarcoplasmic reticulum (SR) and DAG activates PKC.

Activation of Gαq/11 is also suggested to cause the opening of voltage-operated Ca channels and Ca2+

entry, the mechanism of which is not clear and may be as a result of direct activation or indirect

activation of channel opening. Inhibition of the Ca-ATPase pump inhibits Ca exit from the cell, thus

promoting elevated [Ca]i. The reduction in lumenal SR [Ca] is thought to trigger capacitative or store-

operated Ca entry. The combined elevation in [Ca]i leads to formation of the Ca-calmodulin complex

which then activates MLC kinase, resulting in acto-myosin crossbridge cycling and myometrial

contraction.

In addition, DAG activation of PKC activates the MAPK cascade resulting in increased PLA2 activity

and prostaglandin production, which also contributes to contraction (mechanism not shown). DAG-

activated PKC also signals for phosphorylation of CPI-17 whilst oxytocin binding to OTR also activates

Rho-A which in turn activates ROCK. Both phosphorylated CPI-17 and ROCK inhibit MLCP leading to

increased MLC phosphorylation and is the proposed mechanism of Ca sensitisation in the

myometrium. Oxytocin receptor signalling in other uterine tissues (e.g. decidua and amnion) also

signals for prostaglandin production which may mediate local paracrine signalling with the

myometrium.

OTR: oxytocin receptor, ROC: Receptor operated channel, VOCC: voltage operated Ca channel, LTCC:

L-type Ca channel, TRP: Transient receptor potential channel: PLC-β: phospholipase C-β, PIP2:

phosphatidylinositol 4,5-bisphosphate, IP3: inositol 1,4,5- triphosphate, DAG: diacylglycerol, PKC:

This article is protected by copyright. All rights reserved.


protein kinases type C, CPI-17: C-kinase-activated protein phosphatase-1 (PP1) inhibitor 17kDa, Ca-
Accepted Article CAM: Ca-calmodulin complex, MLCK: myosin light-chain kinase, MLCP: myosin light chain

phosphatase, MAPK: mitogen-activated protein kinase, ERK: extracellular signal-regulated protein

kinase, PLA2: phospholipase A2, ROCK: RhoA-associated protein kinase, SOCE: store-operated Ca

entry.

Red pathways indicate signalling pathways with direct influences on [Ca]i whilst purple and turquoise

lines indicate Ca-independent pathways to contraction including Ca sensitisation (purple lines) and

production of prostaglandins (turquoise pathways). Dotted lines indicate where mechanisms are not

yet fully elucidated.

References

1. Dale HH. The Action of Extracts of the Pituitary Body. The Biochemical journal. 1909; 4(9):
427-47.
2. Du Vigneaud V, Ressler C, Trippett S. The sequence of amino acids in oxytocin, with a
proposal for the structure of oxytocin. The Journal of biological chemistry. 1953; 205(2): 949-57.
3. Bethlehem RA, van Honk J, Auyeung B, Baron-Cohen S. Oxytocin, brain physiology, and
functional connectivity: a review of intranasal oxytocin fMRI studies. Psychoneuroendocrinology.
2013; 38(7): 962-74.
4. Brownstein MJ, Russell JT, Gainer H. Synthesis, transport, and release of posterior pituitary
hormones. Science (New York, NY). 1980; 207(4429): 373-8.
5. Landgraf R, Neumann ID. Vasopressin and oxytocin release within the brain: a dynamic
concept of multiple and variable modes of neuropeptide communication. Frontiers in
neuroendocrinology. 2004; 25(3-4): 150-76.
6. Renaud LP, Bourque CW. Neurophysiology and neuropharmacology of hypothalamic
magnocellular neurons secreting vasopressin and oxytocin. Progress in neurobiology. 1991; 36(2):
131-69.
7. Wray S. Insights into the uterus. Experimental physiology. 2007; 92(4): 621-31.
8. Gimpl G, Fahrenholz F. The oxytocin receptor system: structure, function, and regulation.
Physiological reviews. 2001; 81(2): 629-83.
9. Shmygol A, Gullam J, Blanks A, Thornton S. Multiple mechanisms involved in oxytocin-
induced modulation of myometrial contractility. Acta pharmacologica Sinica. 2006; 27(7): 827-32.
10. Luckas MJ, Taggart MJ, Wray S. Intracellular calcium stores and agonist-induced contractions
in isolated human myometrium. American journal of obstetrics and gynecology. 1999; 181(2): 468-
76.
11. Wray S, Kupittayanant S, Shmygol A, Smith RD, Burdyga T. The physiological basis of uterine
contractility: a short review. Experimental physiology. 2001; 86(2): 239-46.

This article is protected by copyright. All rights reserved.


12. Mironneau J. Effects of oxytocin on ionic currents underlying rhythmic activity and
contraction in uterine smooth muscle. Pflugers Archiv : European journal of physiology. 1976; 363(2):
Accepted Article 113-8.
13. Arnaudeau S, Lepretre N, Mironneau J. Oxytocin mobilizes calcium from a unique heparin-
sensitive and thapsigargin-sensitive store in single myometrial cells from pregnant rats. Pflugers
Archiv : European journal of physiology. 1994; 428(1): 51-9.
14. Inoue Y, Shimamura K, Sperelakis N. Oxytocin actions on voltage-dependent ionic channels
in pregnant rat uterine smooth muscle cells. Canadian journal of physiology and pharmacology.
1992; 70(12): 1597-603.
15. Monga M, Campbell DF, Sanborn BM. Oxytocin-stimulated capacitative calcium entry in
human myometrial cells. American journal of obstetrics and gynecology. 1999; 181(2): 424-9.
16. Sanborn BM. Hormones and calcium: mechanisms controlling uterine smooth muscle
contractile activity. The Litchfield Lecture. Experimental physiology. 2001; 86(2): 223-37.
17. Floyd R, Wray S. Calcium transporters and signalling in smooth muscles. Cell calcium. 2007;
42(4-5): 467-76.
18. Shlykov SG, Sanborn BM. Stimulation of intracellular Ca2+ oscillations by diacylglycerol in
human myometrial cells. Cell calcium. 2004; 36(2): 157-64.
19. Waldron RT, Short AD, Gill DL. Store-operated Ca2+ entry and coupling to Ca2+ pool
depletion in thapsigargin-resistant cells. The Journal of biological chemistry. 1997; 272(10): 6440-7.
20. Putney JW. Physiological mechanisms of TRPC activation. Pflugers Archiv : European journal
of physiology. 2005; 451(1): 29-34.
21. Minke B. TRP channels and Ca2+ signaling. Cell calcium. 2006; 40(3): 261-75.
22. Yang M, Gupta A, Shlykov SG, Corrigan R, Tsujimoto S, Sanborn BM. Multiple Trp isoforms
implicated in capacitative calcium entry are expressed in human pregnant myometrium and
myometrial cells. Biology of reproduction. 2002; 67(3): 988-94.
23. Ku CY, Babich L, Word RA, Zhong M, Ulloa A, Monga M, Sanborn BM. Expression of transient
receptor channel proteins in human fundal myometrium in pregnancy. Journal of the Society for
Gynecologic Investigation. 2006; 13(3): 217-25.
24. Wray S, Burdyga T. Sarcoplasmic reticulum function in smooth muscle. Physiological reviews.
2010; 90(1): 113-78.
25. Shmigol AV, Eisner DA, Wray S. Simultaneous measurements of changes in sarcoplasmic
reticulum and cytosolic. The Journal of physiology. 2001; 531(Pt 3): 707-13.
26. Shmygol A, Wray S. Modulation of agonist-induced Ca2+ release by SR Ca2+ load: direct SR
and cytosolic Ca2+ measurements in rat uterine myocytes. Cell calcium. 2005; 37(3): 215-23.
27. Shmigol A, Eisner DA, Wray S. Carboxyeosin decreases the rate of decay of the [Ca2+]i
transient in uterine smooth muscle cells isolated from pregnant rats. Pflugers Archiv : European
journal of physiology. 1998; 437(1): 158-60.
28. Popescu LM, Nutu O, Panoiu C. Oxytocin contracts the human uterus at term by inhibiting
the myometrial Ca2+-extrusion pump. Bioscience reports. 1985; 5(1): 21-8.
29. Soloff MS, Sweet P. Oxytocin inhibition of (Ca2+ + Mg2+)-ATPase activity in rat myometrial
plasma membranes. The Journal of biological chemistry. 1982; 257(18): 10687-93.
30. Kupittayanant S, Luckas MJ, Wray S. Effect of inhibiting the sarcoplasmic reticulum on
spontaneous and oxytocin-induced contractions of human myometrium. BJOG : an international
journal of obstetrics and gynaecology. 2002; 109(3): 289-96.
31. Aguilar HN, Tracey CN, Tsang SC, McGinnis JM, Mitchell BF. Phos-tag-based analysis of
myosin regulatory light chain phosphorylation in human uterine myocytes. PloS one. 2011; 6(6):
e20903.
32. Hudson CA, Heesom KJ, Lopez Bernal A. Phasic contractions of isolated human myometrium
are associated with Rho-kinase (ROCK)-dependent phosphorylation of myosin phosphatase-targeting
subunit (MYPT1). Molecular human reproduction. 2012; 18(5): 265-79.

This article is protected by copyright. All rights reserved.


33. Lartey J, Lopez Bernal A. RHO protein regulation of contraction in the human uterus.
Reproduction (Cambridge, England). 2009; 138(3): 407-24.
Accepted Article 34. Somlyo AP, Wu X, Walker LA, Somlyo AV. Pharmacomechanical coupling: the role of calcium,
G-proteins, kinases and phosphatases. Reviews of physiology, biochemistry and pharmacology. 1999;
134201-34.
35. Shabir S, Borisova L, Wray S, Burdyga T. Rho-kinase inhibition and electromechanical
coupling in rat and guinea-pig ureter smooth muscle: Ca2+-dependent and -independent
mechanisms. The Journal of physiology. 2004; 560(Pt 3): 839-55.
36. McKillen K, Thornton S, Taylor CW. Oxytocin increases the [Ca2+]i sensitivity of human
myometrium during the falling phase of phasic contractions. The American journal of physiology.
1999; 276(2 Pt 1): E345-51.
37. Woodcock NA, Taylor CW, Thornton S. Effect of an oxytocin receptor antagonist and rho
kinase inhibitor on the [Ca++]i sensitivity of human myometrium. American journal of obstetrics and
gynecology. 2004; 190(1): 222-8.
38. Kupittayanant S, Burdyga T, Wray S. The effects of inhibiting Rho-associated kinase with Y-
27632 on force and intracellular calcium in human myometrium. Pflugers Archiv : European journal
of physiology. 2001; 443(1): 112-4.
39. Khan-Dawood FS, Yang J, Dawood MY. Hormonal regulation of connexin-43 in baboon
corpora lutea. The Journal of endocrinology. 1998; 157(3): 405-14.
40. Garfield RE, Ali M, Yallampalli C, Izumi H. Role of gap junctions and nitric oxide in control of
myometrial contractility. Seminars in perinatology. 1995; 19(1): 41-51.
41. Pont JN, McArdle CA, Lopez Bernal A. Oxytocin-stimulated NFAT transcriptional activation in
human myometrial cells. Molecular endocrinology (Baltimore, Md). 2012; 26(10): 1743-56.
42. Hinko A, Soloff MS. Characterization of oxytocin receptors in rabbit amnion involved in the
production of prostaglandin E2. Endocrinology. 1992; 130(6): 3547-53.
43. Benedetto MT, De Cicco F, Rossiello F, Nicosia AL, Lupi G, Dell'Acqua S. Oxytocin receptor in
human fetal membranes at term and during labor. Journal of steroid biochemistry. 1990; 35(2): 205-
8.
44. Terzidou V, Blanks AM, Kim SH, Thornton S, Bennett PR. Labor and inflammation increase
the expression of oxytocin receptor in human amnion. Biology of reproduction. 2011; 84(3): 546-52.
45. Chibbar R, Miller FD, Mitchell BF. Synthesis of oxytocin in amnion, chorion, and decidua may
influence the timing of human parturition. The Journal of clinical investigation. 1993; 91(1): 185-92.
46. Wilson T, Liggins GC, Whittaker DJ. Oxytocin stimulates the release of arachidonic acid and
prostaglandin F2 alpha from human decidual cells. Prostaglandins. 1988; 35(5): 771-80.
47. Pasetto N, Piccione E, Zicari A, Fontana L, De Carolis C, Perricone R, Pontieri G, Ticconi C.
Short report: cytokine production by human fetal membranes and uterine decidua at term gestation
in relation to labour. Placenta. 1993; 14(3): 361-4.
48. Burns PD, Mendes JO, Jr., Yemm RS, Clay CM, Nelson SE, Hayes SH, Silvia WJ. Cellular
mechanisms by which oxytocin mediates ovine endometrial prostaglandin F2alpha synthesis: role of
G(i) proteins and mitogen-activated protein kinases. Biology of reproduction. 2001; 65(4): 1150-5.
49. Blanks AM, Thornton S. The role of oxytocin in parturition. BJOG : an international journal of
obstetrics and gynaecology. 2003; 110 Suppl 2046-51.
50. Russell JA, Leng G. Sex, parturition and motherhood without oxytocin? The Journal of
endocrinology. 1998; 157(3): 343-59.
51. Leake RD, Weitzman RE, Glatz TH, Fisher DA. Plasma oxytocin concentrations in men,
nonpregnant women, and pregnant women before and during spontaneous labor. The Journal of
clinical endocrinology and metabolism. 1981; 53(4): 730-3.
52. Fuchs AR, Goeschen K, Husslein P, Rasmussen AB, Fuchs F. Oxytocin and initiation of human
parturition. III. Plasma concentrations of oxytocin and 13,14-dihydro-15-keto-prostaglandin F2 alpha
in spontaneous and oxytocin-induced labor at term. American journal of obstetrics and gynecology.
1983; 147(5): 497-502.

This article is protected by copyright. All rights reserved.


53. Dawood MY, Ylikorkala O, Trivedi D, Fuchs F. Oxytocin in maternal circulation and amniotic
fluid during pregnancy. The Journal of clinical endocrinology and metabolism. 1979; 49(3): 429-34.
Accepted Article 54. Thornton S, Davison JM, Baylis PH. Plasma oxytocin during the first and second stages of
spontaneous human labour. Acta endocrinologica. 1992; 126(5): 425-9.
55. Burd JM, Davison J, Weightman DR, Baylis PH. Evaluation of enzyme inhibitors of pregnancy
associated oxytocinase: application to the measurement of plasma immunoreactive oxytocin during
human labour. Acta endocrinologica. 1987; 114(3): 458-64.
56. Fuchs AR, Dawood MY. Oxytocin release and uterine activation during parturition iin rabbits.
Endocrinology. 1980; 107(4): 1117-26.
57. Higuchi T, Tadokoro Y, Honda K, Negoro H. Detailed analysis of blood oxytocin levels during
suckling and parturition in the rat. The Journal of endocrinology. 1986; 110(2): 251-6.
58. Thornton S, Vatish M, Slater D. Oxytocin antagonists: clinical and scientific considerations.
Experimental physiology. 2001; 86(2): 297-302.
59. Mitchell BF, Schmid B. Oxytocin and its receptor in the process of parturition. Journal of the
Society for Gynecologic Investigation. 2001; 8(3): 122-33.
60. Otsuki Y, Tanizawa O, Yamaji K, Fujita M, Kurachi K. Feto-maternal plasma oxytocin levels in
normal and anencephalic pregnancies. Acta obstetricia et gynecologica Scandinavica. 1983; 62(3):
235-7.
61. Nishimori K, Young LJ, Guo Q, Wang Z, Insel TR, Matzuk MM. Oxytocin is required for nursing
but is not essential for parturition or reproductive behavior. Proceedings of the National Academy of
Sciences of the United States of America. 1996; 93(21): 11699-704.
62. Young WS, 3rd, Shepard E, Amico J, Hennighausen L, Wagner KU, LaMarca ME, McKinney C,
Ginns EI. Deficiency in mouse oxytocin prevents milk ejection, but not fertility or parturition. Journal
of neuroendocrinology. 1996; 8(11): 847-53.
63. Antonijevic IA, Leng G, Luckman SM, Douglas AJ, Bicknell RJ, Russell JA. Induction of uterine
activity with oxytocin in late pregnant rats replicates the expression of c-fos in neuroendocrine and
brain stem neurons as seen during parturition. Endocrinology. 1995; 136(1): 154-63.
64. Phaneuf S, Asboth G, Carrasco MP, Europe-Finner GN, Saji F, Kimura T, Harris A, Lopez Bernal
A. The desensitization of oxytocin receptors in human myometrial cells is accompanied by down-
regulation of oxytocin receptor messenger RNA. The Journal of endocrinology. 1997; 154(1): 7-18.
65. Fuchs AR, Fuchs F, Husslein P, Soloff MS. Oxytocin receptors in the human uterus during
pregnancy and parturition. American journal of obstetrics and gynecology. 1984; 150(6): 734-41.
66. Kimura T, Takemura M, Nomura S, Nobunaga T, Kubota Y, Inoue T, Hashimoto K, Kumazawa
I, Ito Y, Ohashi K, Koyama M, Azuma C, Kitamura Y, Saji F. Expression of oxytocin receptor in human
pregnant myometrium. Endocrinology. 1996; 137(2): 780-5.
67. Fuchs AR, Fields MJ, Freidman S, Shemesh M, Ivell R. Oxytocin and the timing of parturition.
Influence of oxytocin receptor gene expression, oxytocin secretion, and oxytocin-induced
prostaglandin F2 alpha and E2 release. Advances in experimental medicine and biology. 1995;
395405-20.
68. Arthur P, Taggart MJ, Zielnik B, Wong S, Mitchell BF. Relationship between gene expression
and function of uterotonic systems in the rat during gestation, uterine activation and both term and
preterm labour. The Journal of physiology. 2008; 586(Pt 24): 6063-76.
69. Takemura M, Kimura T, Nomura S, Makino Y, Inoue T, Kikuchi T, Kubota Y, Tokugawa Y,
Nobunaga T, Kamiura S, et al. Expression and localization of human oxytocin receptor mRNA and its
protein in chorion and decidua during parturition. The Journal of clinical investigation. 1994; 93(6):
2319-23.
70. Blanks AM, Vatish M, Allen MJ, Ladds G, de Wit NC, Slater DM, Thornton S. Paracrine
oxytocin and estradiol demonstrate a spatial increase in human intrauterine tissues with labor. The
Journal of clinical endocrinology and metabolism. 2003; 88(7): 3392-400.

This article is protected by copyright. All rights reserved.


71. Zingg HH, Rozen F, Breton C, Larcher A, Neculcea J, Chu K, Russo C, Arslan A. Gonadal steroid
regulation of oxytocin and oxytocin receptor gene expression. Advances in experimental medicine
Accepted Article and biology. 1995; 395395-404.
72. Breton C, Di Scala-Guenot D, Zingg HH. Oxytocin receptor gene expression in rat mammary
gland: structural characterization and regulation. Journal of molecular endocrinology. 2001; 27(2):
175-89.
73. Meddle SL, Bishop VR, Gkoumassi E, van Leeuwen FW, Douglas AJ. Dynamic changes in
oxytocin receptor expression and activation at parturition in the rat brain. Endocrinology. 2007;
148(10): 5095-104.
74. Tyzio R, Nardou R, Ferrari DC, Tsintsadze T, Shahrokhi A, Eftekhari S, Khalilov I, Tsintsadze V,
Brouchoud C, Chazal G, Lemonnier E, Lozovaya N, Burnashev N, Ben-Ari Y. Oxytocin-mediated GABA
inhibition during delivery attenuates autism pathogenesis in rodent offspring. Science (New York,
NY). 2014; 343(6171): 675-9.
75. Ferguson J. A study of the motility of the intact uterus at term. Surgery, gynecology and
Obstetrics. 1941; 73359-66.
76. Fang X, Wong S, Mitchell BF. Relationships among sex steroids, oxytocin, and their receptors
in the rat uterus during late gestation and at parturition. Endocrinology. 1996; 137(8): 3213-9.
77. Fang X, Wong S, Mitchell BF. Effects of RU486 on estrogen, progesterone, oxytocin, and their
receptors in the rat uterus during late gestation. Endocrinology. 1997; 138(7): 2763-8.
78. Ou CW, Chen ZQ, Qi S, Lye SJ. Increased expression of the rat myometrial oxytocin receptor
messenger ribonucleic acid during labor requires both mechanical and hormonal signals. Biology of
reproduction. 1998; 59(5): 1055-61.
79. Siebel AL, Gehring HM, Parry LJ. Steroid-independent regulation of uterine oxytocin
receptors. Journal of neuroendocrinology. 2004; 16(4): 398-402.
80. Murata T, Murata E, Liu CX, Narita K, Honda K, Higuchi T. Oxytocin receptor gene expression
in rat uterus: regulation by ovarian steroids. The Journal of endocrinology. 2000; 166(1): 45-52.
81. Mitchell BF, Wong S. Changes in 17 beta,20 alpha-hydroxysteroid dehydrogenase activity
supporting an increase in the estrogen/progesterone ratio of human fetal membranes at parturition.
American journal of obstetrics and gynecology. 1993; 168(5): 1377-85.
82. Mesiano S, Chan EC, Fitter JT, Kwek K, Yeo G, Smith R. Progesterone withdrawal and
estrogen activation in human parturition are coordinated by progesterone receptor A expression in
the myometrium. The Journal of clinical endocrinology and metabolism. 2002; 87(6): 2924-30.
83. Condon JC, Jeyasuria P, Faust JM, Wilson JW, Mendelson CR. A decline in the levels of
progesterone receptor coactivators in the pregnant uterus at term may antagonize progesterone
receptor function and contribute to the initiation of parturition. Proceedings of the National
Academy of Sciences of the United States of America. 2003; 100(16): 9518-23.
84. Draeger A, Wray S, Babiychuk EB. Domain architecture of the smooth-muscle plasma
membrane: regulation by annexins. The Biochemical journal. 2005; 387(Pt 2): 309-14.
85. Gimpl G, Fahrenholz F. Human oxytocin receptors in cholesterol-rich vs. cholesterol-poor
microdomains of the plasma membrane. European journal of biochemistry / FEBS. 2000; 267(9):
2483-97.
86. Gimpl G, Fahrenholz F. Cholesterol as stabilizer of the oxytocin receptor. Biochimica et
biophysica acta. 2002; 1564(2): 384-92.
87. Noble K, Zhang J, Wray S. Lipid rafts, the sarcoplasmic reticulum and uterine calcium
signalling: an integrated approach. The Journal of physiology. 2006; 570(Pt 1): 29-35.
88. Fahrenholz F, Klein U, Gimpl G. Conversion of the myometrial oxytocin receptor from low to
high affinity state by cholesterol. Advances in experimental medicine and biology. 1995; 395311-9.
89. Buxton IL, Vittori JC. Cholesterol depletion enhances both spontaneous and agonist-evoked
uterine smooth muscle contractions in a reversible manner. Proceedings of the Western
Pharmacology Society. 2005; 48126-8.

This article is protected by copyright. All rights reserved.


90. Smith RD, Babiychuk EB, Noble K, Draeger A, Wray S. Increased cholesterol decreases
uterine activity: functional effects of cholesterol alteration in pregnant rat myometrium. American
Accepted Article journal of physiology Cell physiology. 2005; 288(5): C982-8.
91. Zhang J, Kendrick A, Quenby S, Wray S. Contractility and calcium signaling of human
myometrium are profoundly affected by cholesterol manipulation: implications for labor?
Reproductive sciences (Thousand Oaks, Calif). 2007; 14(5): 456-66.
92. Wu WX, Ma XH, Yoshizato T, Shinozuka N, Nathanielsz PW. Differential expression of
myometrial oxytocin receptor and prostaglandin H synthase 2, but not estrogen receptor alpha and
heat shock protein 90 messenger ribonucleic acid in the gravid horn and nongravid horn in sheep
during betamethasone-induced labor. Endocrinology. 1999; 140(12): 5712-8.
93. Parry LJ, Bathgate RA, Shaw G, Renfree MB, Ivell R. Evidence for a local fetal influence on
myometrial oxytocin receptors during pregnancy in the tammar wallaby (Macropus eugenii). Biology
of reproduction. 1997; 56(1): 200-7.
94. Terzidou V, Sooranna SR, Kim LU, Thornton S, Bennett PR, Johnson MR. Mechanical stretch
up-regulates the human oxytocin receptor in primary human uterine myocytes. The Journal of
clinical endocrinology and metabolism. 2005; 90(1): 237-46.
95. Many A, Hill LM, Lazebnik N, Martin JG. The association between polyhydramnios and
preterm delivery. Obstetrics and gynecology. 1995; 86(3): 389-91.
96. Stock S, Norman J. Preterm and term labour in multiple pregnancies. Seminars in fetal &
neonatal medicine. 2010; 15(6): 336-41.
97. Turton P, Arrowsmith S, Prescott J, Ballard C, Bricker L, Neilson J, Wray S. A comparison of
the contractile properties of myometrium from singleton and twin pregnancies. PloS one. 2013; 8(5):
e63800.
98. Turton P, Neilson JP, Quenby S, Burdyga T, Wray S. A short review of twin pregnancy and
how oxytocin receptor expression may differ in multiple pregnancy. European journal of obstetrics,
gynecology, and reproductive biology. 2009; 144 Suppl 1S40-4.
99. Arrowsmith S, Quenby S, Weeks A, Burdyga T, Wray S. Poor spontaneous and oxytocin-
stimulated contractility in human myometrium from postdates pregnancies. PloS one. 2012; 7(5):
e36787.
100. Luttrell LM, Lefkowitz RJ. The role of beta-arrestins in the termination and transduction of G-
protein-coupled receptor signals. Journal of cell science. 2002; 115(Pt 3): 455-65.
101. Phaneuf S, Asboth G, Carrasco MP, Linares BR, Kimura T, Harris A, Bernal AL. Desensitization
of oxytocin receptors in human myometrium. Human reproduction update. 1998; 4(5): 625-33.
102. Smith MP, Ayad VJ, Mundell SJ, McArdle CA, Kelly E, Lopez Bernal A. Internalization and
desensitization of the oxytocin receptor is inhibited by Dynamin and clathrin mutants in human
embryonic kidney 293 cells. Molecular endocrinology (Baltimore, Md). 2006; 20(2): 379-88.
103. Phaneuf S, Rodriguez Linares B, TambyRaja RL, MacKenzie IZ, Lopez Bernal A. Loss of
myometrial oxytocin receptors during oxytocin-induced and oxytocin-augmented labour. Journal of
reproduction and fertility. 2000; 120(1): 91-7.
104. Baumann G, Dingman JF. Distribution, blood transport, and degradation of antidiuretic
hormone in man. The Journal of clinical investigation. 1976; 57(5): 1109-16.
105. Czaczkes JW, Kleeman CR, Koenig M. PHYSIOLOGIC STUDIES OF ANTIDIURETIC HORMONE BY
ITS DIRECT MEASUREMENT IN HUMAN PLASMA. The Journal of clinical investigation. 1964; 431625-
40.
106. Stoop R. Neuromodulation by oxytocin and vasopressin. Neuron. 2012; 76(1): 142-59.
107. Holmes CL, Landry DW, Granton JT. Science review: Vasopressin and the cardiovascular
system part 1--receptor physiology. Critical care (London, England). 2003; 7(6): 427-34.
108. Akerlund M, Bossmar T, Brouard R, Kostrzewska A, Laudanski T, Lemancewicz A, Serradeil-Le
Gal C, Steinwall M. Receptor binding of oxytocin and vasopressin antagonists and inhibitory effects
on isolated myometrium from preterm and term pregnant women. British journal of obstetrics and
gynaecology. 1999; 106(10): 1047-53.

This article is protected by copyright. All rights reserved.


109. Peter J, Burbach H, Adan RA, Lolait SJ, van Leeuwen FW, Mezey E, Palkovits M, Barberis C.
Molecular neurobiology and pharmacology of the vasopressin/oxytocin receptor family. Cellular and
Accepted Article molecular neurobiology. 1995; 15(5): 573-95.
110. Terrillon S, Durroux T, Mouillac B, Breit A, Ayoub MA, Taulan M, Jockers R, Barberis C,
Bouvier M. Oxytocin and vasopressin V1a and V2 receptors form constitutive homo- and
heterodimers during biosynthesis. Molecular endocrinology (Baltimore, Md). 2003; 17(4): 677-91.
111. Bossmar T, Akerlund M, Szamatowicz J, Laudanski T, Fantoni G, Maggi M. Receptor-
mediated uterine effects of vasopressin and oxytocin in nonpregnant women. British journal of
obstetrics and gynaecology. 1995; 102(11): 907-12.
112. Chan WY, Wo NC, Manning M. The role of oxytocin receptors and vasopressin V1a receptors
in uterine contractions in rats: implications for tocolytic therapy with oxytocin antagonists. American
journal of obstetrics and gynecology. 1996; 175(5): 1331-5.
113. Mackler AM, Ducsay CA, Veldhuis JD, Yellon SM. Maturation of spontaneous and agonist-
induced uterine contractions in the peripartum mouse uterus. Biology of reproduction. 1999; 61(4):
873-8.
114. Maggi M, Del Carlo P, Fantoni G, Giannini S, Torrisi C, Casparis D, Massi G, Serio M. Human
myometrium during pregnancy contains and responds to V1 vasopressin receptors as well as
oxytocin receptors. The Journal of clinical endocrinology and metabolism. 1990; 70(4): 1142-54.
115. Bossmar T, Akerlund M, Fantoni G, Szamatowicz J, Melin P, Maggi M. Receptors for and
myometrial responses to oxytocin and vasopressin in preterm and term human pregnancy: effects of
the oxytocin antagonist atosiban. American journal of obstetrics and gynecology. 1994; 171(6): 1634-
42.
116. Bossmar T, Rasmussen T, Akerlund M. Effect of the non-peptide, vasopressin V1a receptor
antagonist, SR 49059 and its enantiomer, SR 49770, on isolated human myometrium. Acta
obstetricia et gynecologica Scandinavica. 1996; 75(6): 516-9.
117. Fuchs AR, Behrens O, Maschek H, Kupsch E, Einspanier A. Oxytocin and vasopressin
receptors in human and uterine myomas during menstrual cycle and early pregnancy. Human
reproduction update. 1998; 4(5): 594-604.
118. Akerlund M. Targeting the oxytocin receptor to relax the myometrium. Expert opinion on
therapeutic targets. 2006; 10(3): 423-7.
119. Helmer H, Hackl T, Schneeberger C, Knofler M, Behrens O, Kaider A, Husslein P. Oxytocin and
vasopressin 1a receptor gene expression in the cycling or pregnant human uterus. American journal
of obstetrics and gynecology. 1998; 179(6 Pt 1): 1572-8.
120. Kaplan M, Eidelman AI, Aboulafia Y. Fasting and the precipitation of labor. The Yom Kippur
effect. JAMA : the journal of the American Medical Association. 1983; 250(10): 1317-8.
121. Hadeed AJ, Leake RD, Weitzman RE, Fisher DA. Possible mechanisms of high blood levels of
vasopressin during the neonatal period. The Journal of pediatrics. 1979; 94(5): 805-8.
122. Parboosingh J, Lederis K, Ko D, Singh N. Vasopressin concentration in cord blood: correlation
with method of delivery and cord pH. Obstetrics and gynecology. 1982; 60(2): 179-83.
123. NICE. National Institute for Health and Care Excellence Clinical Guildlines: Induction of
Labour (CG70). 2008.
124. Bishop EH. PELVIC SCORING FOR ELECTIVE INDUCTION. Obstetrics and gynecology. 1964;
24266-8.
125. Kelly AJ, Tan B. Intravenous oxytocin alone for cervical ripening and induction of labour. The
Cochrane database of systematic reviews. 2001(3): CD003246.
126. NICE. National Institute for Health and Care Excellence Clinical Guidelines: Intrapartum Care
(CG55). 2007.
127. Williams FL, Florey CV, Ogston SA, Patel NB, Howie PW, Tindall VR. UK study of intrapartum
care for low risk primigravidas: a survey of interventions. Journal of epidemiology and community
health. 1998; 52(8): 494-500.

This article is protected by copyright. All rights reserved.


128. Blanch G, Lavender T, Walkinshaw S, Alfirevic Z. Dysfunctional labour: a randomised trial.
British journal of obstetrics and gynaecology. 1998; 105(1): 117-20.
Accepted Article 129. Ryden G, Sjoholm I. Half-life of oxytocin in blood of pregnant and non-pregnant women.
Acta endocrinologica. 1969; 61(3): 425-31.
130. Gonser M. Labor induction and augmentation with oxytocin: pharmacokinetic
considerations. Archives of gynecology and obstetrics. 1995; 256(2): 63-6.
131. Amico JA, Seitchik J, Robinson AG. Studies of oxytocin in plasma of women during
hypocontractile labor. The Journal of clinical endocrinology and metabolism. 1984; 58(2): 274-9.
132. Clark SL, Simpson KR, Knox GE, Garite TJ. Oxytocin: new perspectives on an old drug.
American journal of obstetrics and gynecology. 2009; 200(1): 35 e1-6.
133. Bugg GJ, Siddiqui F, Thornton JG. Oxytocin versus no treatment or delayed treatment for
slow progress in the first stage of spontaneous labour. The Cochrane database of systematic reviews.
2013; 6CD007123.
134. Crall HD, Mattison DR. Oxytocin pharmacodynamics: effect of long infusions on uterine
activity. Gynecologic and obstetric investigation. 1991; 31(1): 17-22.
135. Belghiti J, Kayem G, Dupont C, Rudigoz RC, Bouvier-Colle MH, Deneux-Tharaux C. Oxytocin
during labour and risk of severe postpartum haemorrhage: a population-based, cohort-nested case-
control study. BMJ open. 2011; 1(2): e000514.
136. Harrison N, Larcombe-McDouall JB, Earley L, Wray S. An in vivo study of the effects of
ischaemia on uterine contraction, intracellular pH and metabolites in the rat. The Journal of
physiology. 1994; 476(2): 349-54.
137. Peebles DM, Spencer JA, Edwards AD, Wyatt JS, Reynolds EO, Cope M, Delpy DT. Relation
between frequency of uterine contractions and human fetal cerebral oxygen saturation studied
during labour by near infrared spectroscopy. British journal of obstetrics and gynaecology. 1994;
101(1): 44-8.
138. Bremme K, Bygdeman M. A comparative study of uterine activity and fetal heart rate pattern
in labor induced with oral prostaglandin E2 or oxytocin. Acta obstetricia et gynecologica
Scandinavica Supplement. 1980; 9223-9.
139. Chia YT, Arulkumaran S, Soon SB, Norshida S, Ratnam SS. Induction of labour: does internal
tocography result in better obstetric outcome than external tocography. The Australian & New
Zealand journal of obstetrics & gynaecology. 1993; 33(2): 159-61.
140. Liston R, Crane J, Hughes O, Kuling S, MacKinnon C, Milne K, Richardson B, Trepanier MJ,
Fetal Health Surveillance Working G. Fetal health surveillance in labour. Journal of obstetrics and
gynaecology Canada : JOGC = Journal d'obstetrique et gynecologie du Canada : JOGC. 2002; 24(4):
342-55.
141. Kimura T. Investigation of the oxytocin receptor at the molecular level. Advances in
experimental medicine and biology. 1995; 395259-68.
142. Quenby S, Pierce SJ, Brigham S, Wray S. Dysfunctional labor and myometrial lactic acidosis.
Obstetrics and gynecology. 2004; 103(4): 718-23.
143. Salamalekis E, Vitoratos N, Kassanos D, Loghis C, Panayotopoulos N, Sykiotis C. A randomized
trial of pulsatile vs continuous oxytocin infusion for labor induction. Clinical and experimental
obstetrics & gynecology. 2000; 27(1): 21-3.
144. Tribe RM, Crawshaw SE, Seed P, Shennan AH, Baker PN. Pulsatile versus continuous
administration of oxytocin for induction and augmentation of labor: two randomized controlled
trials. American journal of obstetrics and gynecology. 2012; 206(3): 230.e1-8.
145. Reid GJ, Helewa ME. A trial of pulsatile versus continuous oxytocin administration for the
induction of labor. Journal of perinatology : official journal of the California Perinatal Association.
1995; 15(5): 364-6; quiz 7-8.
146. Prendiville W, Elbourne D, Chalmers I. The effects of routine oxytocic administration in the
management of the third stage of labour: an overview of the evidence from controlled trials. British
journal of obstetrics and gynaecology. 1988; 95(1): 3-16.

This article is protected by copyright. All rights reserved.


147. McDonald S, Abbott JM, Higgins SP. Prophylactic ergometrine-oxytocin versus oxytocin for
the third stage of labour. The Cochrane database of systematic reviews. 2004(1): CD000201.
Accepted Article 148. Rath W. Prevention of postpartum haemorrhage with the oxytocin analogue carbetocin.
European journal of obstetrics, gynecology, and reproductive biology. 2009; 147(1): 15-20.
149. Su LL, Chong YS, Samuel M. Oxytocin agonists for preventing postpartum haemorrhage. The
Cochrane database of systematic reviews. 2007(3): CD005457.
150. Bell WB. THE PITUITARY BODY AND THE THERAPEUTIC VALUE OF THE INFUNDIBULAR
EXTRACT IN SHOCK, UTERINE ATONY, AND INTESTINAL PARESIS. British medical journal. 1909;
2(2553): 1609-13.
151. Theobald GW, Robards MF, Suter PE. Changes in myometrial sensitivity to oxytocin in man
during the last six weeks of pregnancy. The Journal of obstetrics and gynaecology of the British
Commonwealth. 1969; 76(5): 385-93.
152. Kupittayanant S, Munglue P, Lijuan W, Promprom W, Budhaklala N, Wray S. Finding New
Agents in Medicinal Plants to Act on the Myometrium. Experimental physiology. 2014.
153. Gruber CW, O'Brien M. Uterotonic plants and their bioactive constituents. Planta medica.
2011; 77(3): 207-20.
154. Koehbach J, O'Brien M, Muttenthaler M, Miazzo M, Akcan M, Elliott AG, Daly NL, Harvey PJ,
Arrowsmith S, Gunasekera S, Smith TJ, Wray S, Goransson U, Dawson PE, Craik DJ, Freissmuth M,
Gruber CW. Oxytocic plant cyclotides as templates for peptide G protein-coupled receptor ligand
design. Proceedings of the National Academy of Sciences of the United States of America. 2013;
110(52): 21183-8.
155. Promprom W, Kupittayanant P, Indrapichate K, Wray S, Kupittayanant S. The effects of
pomegranate seed extract and beta-sitosterol on rat uterine contractions. Reproductive sciences
(Thousand Oaks, Calif). 2010; 17(3): 288-96.
156. Tripathi V, Stanton C, Anderson FW. Traditional preparations used as uterotonics in Sub-
Saharan Africa and their pharmacologic effects. International journal of gynaecology and obstetrics:
the official organ of the International Federation of Gynaecology and Obstetrics. 2013; 120(1): 16-22.
157. Arrowsmith S, Kendrick A, Wray S. Drugs acting on the pregnant uterus. Obstetrics,
Gynaecology and Reproductive Medicine. 2010; 20(8): 241-7.
158. Hubinont C, Debieve F. Prevention of preterm labour: 2011 update on tocolysis. Journal of
pregnancy. 2011; 2011941057.
159. de Heus R, Mulder EJ, Derks JB, Visser GH. Acute tocolysis for uterine activity reduction in
term labor: a review. Obstetrical & gynecological survey. 2008; 63(6): 383-8; quiz 405.
160. Coomarasamy A, Knox EM, Gee H, Khan KS. Oxytocin antagonists for tocolysis in preterm
labour -- a systematic review. Medical science monitor : international medical journal of
experimental and clinical research. 2002; 8(11): Ra268-73.
161. Kashanian M, Akbarian AR, Soltanzadeh M. Atosiban and nifedipin for the treatment of
preterm labor. International journal of gynaecology and obstetrics: the official organ of the
International Federation of Gynaecology and Obstetrics. 2005; 91(1): 10-4.
162. Al-Omari WR, Al-Shammaa HB, Al-Tikriti EM, Ahmed KW. Atosiban and nifedipine in acute
tocolysis: a comparative study. European journal of obstetrics, gynecology, and reproductive biology.
2006; 128(1-2): 129-34.
163. Papatsonis D, Flenady V, Cole S, Liley H. Oxytocin receptor antagonists for inhibiting preterm
labour. The Cochrane database of systematic reviews. 2005(3): CD004452.
164. Haas DM, Caldwell DM, Kirkpatrick P, McIntosh JJ, Welton NJ. Tocolytic therapy for preterm
delivery: systematic review and network meta-analysis. BMJ (Clinical research ed). 2012; 345e6226.
165. Nilsson L, Reinheimer T, Steinwall M, Akerlund M. FE 200 440: a selective oxytocin
antagonist on the term-pregnant human uterus. BJOG : an international journal of obstetrics and
gynaecology. 2003; 110(11): 1025-8.

This article is protected by copyright. All rights reserved.


166. Pierzynski P, Lemancewicz A, Reinheimer T, Akerlund M, Laudanski T. Inhibitory effect of
barusiban and atosiban on oxytocin-induced contractions of myometrium from preterm and term
Accepted Article pregnant women. Journal of the Society for Gynecologic Investigation. 2004; 11(6): 384-7.
167. Reinheimer TM, Bee WH, Resendez JC, Meyer JK, Haluska GJ, Chellman GJ. Barusiban, a new
highly potent and long-acting oxytocin antagonist: pharmacokinetic and pharmacodynamic
comparison with atosiban in a cynomolgus monkey model of preterm labor. The Journal of clinical
endocrinology and metabolism. 2005; 90(4): 2275-81.
168. Reinheimer TM, Chellman GJ, Resendez JC, Meyer JK, Bee WH. Barusiban, an effective long-
term treatment of oxytocin-induced preterm labor in nonhuman primates. Biology of reproduction.
2006; 75(5): 809-14.
169. Thornton S, Goodwin TM, Greisen G, Hedegaard M, Arce JC. The effect of barusiban, a
selective oxytocin antagonist, in threatened preterm labor at late gestational age: a randomized,
double-blind, placebo-controlled trial. American journal of obstetrics and gynecology. 2009; 200(6):
627 e1-10.
170. ClinicalTrials.gov. A Randomised, Placebo-controlled, Double-blind, Parallel Groups,
Multinational, Multicentre Trial Assessing the Effect of Barusiban Administered Subcutaneously on
the Day of Transfer on Implantation and Pregnancy Rates in IVF/ICSI Patients. 2012.
171. Steinwall M, Bossmar T, Brouard R, Laudanski T, Olofsson P, Urban R, Wolff K, Le-Fur G,
Akerlund M. The effect of relcovaptan (SR 49059), an orally active vasopressin V1a receptor
antagonist, on uterine contractions in preterm labor. Gynecological endocrinology : the official
journal of the International Society of Gynecological Endocrinology. 2005; 20(2): 104-9.
172. Brouard R, Bossmar T, Fournie-Lloret D, Chassard D, Akerlund M. Effect of SR49059, an orally
active V1a vasopressin receptor antagonist, in the prevention of dysmenorrhoea. BJOG : an
international journal of obstetrics and gynaecology. 2000; 107(5): 614-9.
173. Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini B, Durroux T, Mouillac B, Corbani
M, Guillon G. Oxytocin and vasopressin agonists and antagonists as research tools and potential
therapeutics. Journal of neuroendocrinology. 2012; 24(4): 609-28.
174. Boccia ML, Goursaud AP, Bachevalier J, Anderson KD, Pedersen CA. Peripherally
administered non-peptide oxytocin antagonist, L368,899, accumulates in limbic brain areas: a new
pharmacological tool for the study of social motivation in non-human primates. Hormones and
behavior. 2007; 52(3): 344-51.
175. Serradeil-Le Gal C, Valette G, Foulon L, Germain G, Advenier C, Naline E, Bardou M,
Martinolle JP, Pouzet B, Raufaste D, Garcia C, Double-Cazanave E, Pauly M, Pascal M, Barbier A,
Scatton B, Maffrand JP, Le Fur G. SSR126768A (4-chloro-3-[(3R)-(+)-5-chloro-1-(2,4-
dimethoxybenzyl)-3-methyl-2-oxo-2,3-dihydro -1H-indol-3-yl]-N-ethyl-N-(3-pyridylmethyl)-
benzamide, hydrochloride): a new selective and orally active oxytocin receptor antagonist for the
prevention of preterm labor. The Journal of pharmacology and experimental therapeutics. 2004;
309(1): 414-24.
176. Fisk NM, Atun R. Market failure and the poverty of new drugs in maternal health. PLoS
medicine. 2008; 5(1): e22.
177. McCafferty GP, Pullen MA, Wu C, Edwards RM, Allen MJ, Woollard PM, Borthwick AD, Liddle
J, Hickey DM, Brooks DP, Westfall TD. Use of a novel and highly selective oxytocin receptor
antagonist to characterize uterine contractions in the rat. American journal of physiology Regulatory,
integrative and comparative physiology. 2007; 293(1): R299-305.
178. Snidow J, Miller H, Valenzuela G, Thornton S, Stier B, Clayton L, Fossler M, Montague T,
Beach K, Williams P. A multicenter, randomized, double-blind, placebo controlled phase 2 trial of
retosiban, a selective oxytocin receptor antagonist, for the management of preterm labor. American
journal of obstetrics and gynecology. 2013(Suppl Jan 2013): S155.

This article is protected by copyright. All rights reserved.


Accepted Article

Vous aimerez peut-être aussi