Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Jan 7;231(4):e13607. doi: 10.1111/apha.13607

Uterine contractions in rodent models and humans

Manasi Malik 1, Michelle Roh 1, Sarah K England 1,
PMCID: PMC8047897  PMID: 33337577

Abstract

Aberrant uterine contractions can lead to preterm birth and other labour complications and are a significant cause of maternal morbidity and mortality. To investigate the mechanisms underlying dysfunctional uterine contractions, researchers have used experimentally tractable small animal models. However, biological differences between humans and rodents change how researchers select their animal model and interpret their results. Here, we provide a general review of studies of uterine excitation and contractions in mice, rats, guinea pigs, and humans, in an effort to introduce new researchers to the field and help in the design and interpretation of experiments in rodent models.

Keywords: animal models, contraction, myometrium, parturition, pregnancy, species differences, uterus

1. INTRODUCTION

Problems in pregnancy, labour and delivery affect millions of people per year and contribute to significant mortality and morbidity worldwide. 1 , 2 , 3 Dysfunctional uterine contractions in labour can cause complications such as preterm birth, dystocia, uterine atony and post‐partum haemorrhage. Therefore, research in this field is of paramount importance to public health. Current understanding of uterine contractions relies on evidence from both human subjects and animal models. Because of practical and ethical concerns with research on pregnant women, uterine biologists have turned animal models, primarily using mice, rats, and guinea pigs. However, because of physiological differences between species, experiments on rodent models must be carefully designed and interpreted.

The basic architecture of pregnancy differs considerably between humans and rodents (Figure 1). Typically, humans have a 40‐week gestation and deliver one to two offspring, whereas rodent gestations are shorter in length with larger litter sizes. The shape and structure of the uterus is different between humans and rodents: humans have pyriform (lightbulb‐shaped) uteri, whereas mice, rats and guinea pigs have bicornuate (two‐horned) uteri with multiple implantation sites in each uterine horn. 4 In all species, the uterus consists of an inner mucosal layer called the endometrium or decidua, an intermediate muscular layer called the myometrium, and an outer serosal layer called the perimetrium. The myometrium consists of both longitudinal and circular muscle fibres. In rodents, the myometrium has a clearly defined outer longitudinal layer and inner circular layer. However, in humans, the longitudinal and circular fibres are not easily distinguishable. 5

FIGURE 1.

FIGURE 1

Uterine anatomy, histology, and pregnancy compared across species

The precipitating factors that trigger parturition also differ between humans, mice, rats and guinea pig. In mice and rats, labour is likely triggered by progesterone withdrawal. 6 During gestation, progesterone is produced by the corpus luteum. 6 At term pregnancy, prostaglandin production causes destruction of the corpus luteum (luteolysis) and consequently a decline in serum progesterone levels. 6 Early progesterone withdrawal, caused by treatment with a progesterone blocker such as RU486, causes premature labour, whereas progesterone supplementation causes prolongation of pregnancy. 7 , 8 These conditions differ in the human and the guinea pig. In both these species, the corpus luteum involutes early in pregnancy and progesterone is instead produced by the placenta. Progesterone levels remain high into late pregnancy in the human and guinea pig; further supplementation with progesterone does not prolong gestation in the guinea pig and has varying efficacy at preventing preterm birth in humans. 9 , 10 Thus, progesterone withdrawal alone is not the trigger for labour in guinea pigs or humans, although potential mechanisms for “functional” progesterone withdrawal are an area of active investigation. 11 Mitchell and Taggart 6 have hypothesized that labour in humans and guinea pigs is instead caused by multiple processes that occur in parallel to alter the balance between pro‐quiescent and pro‐contractile stimuli, as well as pro‐ and anti‐inflammatory stimuli. Still, further research is necessary to determine whether these processes are similar between humans and guinea pigs.

When choosing an experimental model, researchers should consider the practical and ethical constraints that inform the types of experiments that can be performed with each species. In the United States, federal regulations provide special protections for pregnant women in research because of the risks that experimental interventions may pose to foetuses. For example, a pregnant woman can only be included in a trial if the potential benefit to the mother or foetus significantly outweighs any risks, or if the risks are negligible. Thus, pregnancy‐related experimental studies in humans are rare, constituting only 0.27% of the drug trials registered on Clinicaltrials.gov. 12 To supplement these in vivo studies, researchers use tissue collected from pregnant participants. Intact uterine specimens can be used for ex vivo contractility experiments, or human myometrial smooth muscle cells can be isolated and studied in culture. Many of the experiments described in this review were performed on uterine biopsies taken at the time of Caesarean section (C‐section). C‐sections are routinely performed on labouring and non‐labouring patients at term and preterm. However, these biopsies are necessarily samples of convenience: samples from pre‐pregnancy, mid‐pregnancy, or the postpartum period are rare, or nearly impossible to obtain ethically. Moreover, tissue strips are usually resected from the lower uterine segment, so results may or may not be applicable to the uterine fundus, the contractile portion of the uterus. 6 Finally, environmental and genetic factors vary widely between the patients who provide biopsies, resulting in heterogeneous samples and sometimes inconsistent results.

In contrast, when working with rodent models, researchers can obtain tissue at any time point prior to and during pregnancy and from any location within the uterus. Heterogeneity is minimal because environmental influences are similar in animals housed together, and genetic differences are negligible in inbred mouse strains commonly used for research. However, this homogeneity can limit the relevance of animal studies to human pathophysiology. For example, mice and rats rarely experience spontaneous preterm labour, so researchers must inoculate animals with infectious agents or treat them with lipopolysaccharide or progesterone blockers to induce early delivery. 13 Additionally, studies in rats and mice cannot recapitulate the myriad causes of idiopathic preterm labour in humans. In contrast, guinea pigs experience preterm labour and uterine dystocia at similar rates to humans, so researchers interested in dysfunctional labour might prefer the guinea pig model. 6 For guinea pigs, as for humans, the causes of dysfunctional labour are unknown.

Other practical considerations may inform a researcher's choice of experimental animal model (Figure 1). One consideration may be gestation length: compared to guinea pigs, mice and rats have shorter gestations. Mice are the least expensive mammalian model to maintain, and inbred mouse strains are ideal targets for genetic manipulation. As reviewed by Ratajczak and Muglia, 14 phenotypes in knockout mouse models have provided gold‐standard evidence to support the importance of certain molecules, such as progesterone and prostaglandins, in parturition. However, with the advent of CRISPR‐Cas9 gene editing, genetic manipulation in rats and guinea pigs may soon be equally practical. Ultimately, selection of the most appropriate animal model must be based on the scientific question at hand. In the next section, we review the current literature on the physiology of uterine contractions, focusing on key similarities and differences in physiology between species, highlighted in Table 1.

TABLE 1.

Key similarities and differences in myometrial physiology between species

Mouse Rat Guinea pig Human
Mechanism of uterine contractions IP3 receptor induces calcium release from endoplasmic reticulum. Yes Yes Yes Yes
RyR modulates contractions. No No No No
SOCE components are present/active in myometrium. Yes Yes No Data Yes
Action potentials and ion channels Both simple and complex action potentials are present. Yes Yes Yes Yes
Kv4 channel function is downregulated by oestrogen. No Data Yes No Data Yes
SK channels promote myometrial quiescence. Yes Yes No Data Yes
KATP openers inhibit contractions at parturition. No Data No No Data Yes
BKCa is pro‐contractile in pro‐inflammatory conditions. No Data No Data No Data Yes
BKCa alternative splicing decreases channel calcium sensitivity at parturition. Yes Yes No Data Yes
LTCC expression (nifedipine binding sites) increases towards parturition. No Data Yes Yes No Data
TTCC inhibition decreases contraction frequency. No Data Yes No Data Yes
Timing and propagation of action potentials Multiple pacemakers are active in the uterus. No Data Yes Yes Yes
Uterine contractions are peristaltic. No Data Yes No No
ICC‐like cells have been identified in uterus. Yes Yes No Data Yes
Gap junctions coordinate contractions. Yes Yes Yes Yes
Uterotonics Oxytocin promotes contractions. Yes Yes Yes Yes
Oxytocin binding sites increase towards parturition. Yes Yes No Yes
Relaxant prostaglandin receptors are downregulated at parturition and pro‐contractile prostaglandin receptors are upregulated at parturition. No Data Yes No Yes
Prostaglandins promote contractions at parturition. Yes Yes Yes Yes

2. PHYSIOLOGY OF UTERINE CONTRACTIONS

2.1. Mechanism of uterine contractions

In all species, smooth muscle contractility is driven by a transient increase in intracellular calcium (Ca2+) (reviewed in 15). Ca2+ binds to calmodulin, and the Ca2+‐calmodulin complex activates myosin light chain kinase (MLCK) (Figure 2). MLCK phosphorylates the regulatory light chain of myosin, which then allows myosin to hydrolyse ATP and move along the actin filament. A decrease in intracellular Ca2+ concentration deactivates MLCK, allowing myosin to be dephosphorylated by myosin light chain phosphatase (MLCP), leading to muscle relaxation.

FIGURE 2.

FIGURE 2

Mechanisms of uterine contractility. Ca2+ entry into the cell can occur through voltage‐gated Ca2+ channels (LTCC) and STIM/Orai, members of the store‐operated calcium entry complex. Ca2+ also effluxes from the sarcoplasmic reticulum through the IP3R to increase intracellular Ca2+ concentration. Intracellular Ca2+ promotes myosin phosphorylation, which in turn enables uterine contraction. Hormones and tocolytic agents modulate myosin phosphorylation through G protein‐coupled receptor‐mediated pathways. cAMP, cyclic AMP; EP, prostaglandin E receptors 1‐4; FP, prostaglandin F receptor; IP3, inositol triphosphate; IP3R, IP3 receptor; LTCC, L‐type calcium channel; MLCK, myosin light chain kinase; MLCP, myosin light chain phosphatase; OXTR, oxytocin receptor; PG, prostaglandins; PIP2, phosphatidylinositol 4,5‐bisphosphate; PLC, phospholipase C; ROCK, Rho kinase; SERCA, sarcoplasmic/endoplasmic reticulum calcium ATPase; STIM, stromal interaction molecule; β2AR, β2 adrenergic receptor

Although these basic players are conserved across species and smooth muscle types, their activities can be differentially regulated. For example, the pro‐labour hormones oxytocin and prostaglandin F2 increase contractility towards the end of pregnancy by increasing the ratio of active MLCK to active MLCP. 16 , 17 These hormones first bind to their respective GPCRs, causing activation of the small G protein RhoA. 18 , 19 , 20 , 21 RhoA in turn activates Rho kinase (ROCK), which phosphorylates MLCP to inactivate it (Figure 2). 18 , 19 , 20 , 21

In contrast, GPCRs that signal through Gs and cyclic AMP (cAMP), such as the β2 adrenergic receptor (β2AR), decrease myometrial contractility (Figure 2). In this process, increases in cAMP and cGMP work through protein kinase A and protein kinase G to decrease the ratio of active MLCK to active MLCP and promote myosin dephosphorylation. 21 While the mechanism for this has yet to be defined in myometrial smooth muscle, experiments in other smooth muscle types suggest that protein kinase A phosphorylates MLCK to decrease its affinity for the Ca2+‐calmodulin complex. 21 Protein kinase A and protein kinase G can also phosphorylate the protein telokin, which activates MLCP. 21 Given that telokin expression in the uterus increases during pregnancy, this mechanism may contribute to the maintenance of uterine quiescence. 22 , 23 , 24

In myometrial cells, the increase in intracellular Ca2+ that drives contractions can occur in several ways. Like most smooth muscle cells, myometrial cells in all species have an extensive sarcoplasmic reticulum (SR) network that maintains a high luminal Ca2+ concentration. The SR takes up Ca2+ from the cytosol through the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA, Figure 2) and stores Ca2+ via Ca2+‐binding proteins. Thus, the SR acts as a Ca2+ buffer, providing Ca2+ to activate actin/myosin contractility and then quickly removing Ca2+ from the cytosol to allow relaxation following a contraction.

Signalling from plasma membrane‐associated Gq‐coupled receptors, such as the oxytocin receptor (OXTR) and prostaglandin F2 receptor (FP), leads to production of inositol triphosphate (IP3), which activates the IP3 receptor on the sarcolemma (Figure 2). 25 The IP3 receptor is also a Ca2+ channel that allows Ca2+ to flow down its concentration gradient from the SR lumen into the cytosol. 26 In non‐myometrial cells, including cardiac, skeletal, and some smooth muscle cells, the SR can also release Ca2+ upon entry of Ca2+ through plasma membrane channels, a process called Ca2+‐induced Ca2+ release (CICR). 27 CICR occurs when entry of Ca2+ through L‐type Ca2+ channels (LTCCs) activates ryanodine receptors (RyRs) on the SR, causing the RyR channel domain to open transiently. This transient opening leads to calcium “sparks” that activate calcium‐gated potassium channels, contributing to repolarization. Researchers initially hypothesized that this could occur in myometrial cells, but studies in both pregnant and nonpregnant animals have shown that these calcium sparks do not occur in mouse or rat myometrium. 28 , 29 , 30 Furthermore, pregnant and nonpregnant mouse myometrium express a dominant negative splice variant of RyR3 that inhibits full length RyR3. 28 , 31 Accordingly, knocking out RyR3 in mice had no effect on pregnancy and labor. 28 While RyR3 is the only RyR isoform found in mice, RyR1 and RyR2 have been found in human pregnant myometrium. 32 , 33 Still, treatment with the RyR inhibitor ryanodine had little effect on mouse, rat or human myometrium. 28 , 34 , 35 Thus, RyRs are likely not important contributors to SR calcium release in mouse, rat, guinea pig or human. 36

Both intracellular and extracellular Ca2+ sources help regulate myometrial contractility via the process of store‐operated calcium entry (SOCE). When the SR releases Ca2+ through the IP3 receptor, Ca2+ concentration in the SR lumen drops, leading to a conformational change in the stromal interaction molecule (STIM) protein located on the SR membrane (Figure 2). 37 Activated STIM then binds to and activates Ca2+ release‐activated Ca2+ channel protein (CRAC, or Orai) and transient receptor potential cation (TRPC) Ca2+ channels located in the plasma membrane, allowing extracellular Ca2+ to enter the cell. 37 SOCE was originally described in non‐excitable cells and striated muscle, but several lines of evidence suggest that this process also occurs in myometrial smooth muscle cells. 38 First, STIM and Orai are expressed in both human and mouse myometrium. 38 , 39 , 40 Second, STIM1 knockout mice have thin uterine walls, and explanted uterine strips from these mice do not contract normally. 38 Finally, Noble et al showed in rat myometrium that depleting Ca2+ from the SR caused an increase in intracellular Ca2+ and phasic contractions that were inhibited by the SOCE blocker Lanthanum. 41

Recent studies have shown that mitochondria might also serve as a myometrial intracellular Ca2+ source. Much like the SR, mitochondria can “buffer” intracellular Ca2+, taking up and releasing Ca2+ to drive the rhythmic contraction and relaxation seen in smooth muscle tissues such as the gastrointestinal tract. In the “pacemaker” cells of the gastrointestinal tract and prostate gland, inhibition of mitochondrial Ca2+ transport dampens pacemaker activity. 42 , 43 Additionally, Gravina et al found that drugs that modulate mitochondrial activity altered the force and frequency of contractions in uterine tissue isolated from nonpregnant mice. 44 However, more work is necessary to describe the exact mechanism of action of these drugs on uterine contractility and to clarify the role of mitochondrial Ca2+ in the myometrium.

2.2. Action potentials and ion channels

2.2.1. Action potentials

Myometrial cells maintain a negative resting membrane potential, meaning that the inside of the plasma membrane is negative relative to the outside of the cell. In the basal state, voltage‐gated Ca2+ channels are closed and intracellular Ca2+ concentrations are low, promoting a non‐contractile state. Studies in isolated human myometrium have shown that as the third trimester progresses, the resting membrane potential becomes less negative, rising from −70 mV at week 29 to −55 mV at week 40. 45 Similarly, the membrane potential in the rat myometrium is the most negative during mid‐pregnancy (−60 mV to −68 mV during days 11 through 15) and becomes less negative (−54 mV) closer to term (rats generally deliver around day 21). 46 , 47 In contrast, the resting membrane potential in guinea pig uterine tissue remains at −58 mV throughout the second half of pregnancy. 48

Slow depolarization of the myometrial cell membrane to around −50 mV allows Ca2+ to enter the cell through nifedipine‐sensitive, voltage‐gated Ca2+ channels, otherwise known as LTCCs. 49 , 50 Ca2+ entry through LTCCs depolarizes the myometrial membrane and causes the upstroke of the uterine action potential. Voltage‐, Ca2+‐, and time‐dependent potassium (K+) channels are responsible for repolarizing the membrane potential. 51 , 52 Action potentials in the myometrium can take the form of a simple spike or a complex plateau (Figure 3). 53 Complex “plateau”‐type action potentials are formed by long depolarizations that often have several superimposed simple spikes, known as calcium bursts. In pregnant rats, simple spikes have been reported to be more common in the longitudinal myometrium, and complex action potentials more common in the circular muscle layer. 53 , 54 , 55 The opposite is true in guinea pigs, as complex action potentials predominate in the longitudinal myometrium, and simple spikes predominate in the circular myometrium. 53 Both types of action potentials have been described in human myometrium, which does not have clearly defined circular and longitudinal layers. 56 , 57 , 58 , 59 Hormones and drugs can modulate the shape of these action potentials in both humans and rats. 54 , 57 For example, oxytocin can extend the depolarization phase so that simple spikes become complex plateaus with calcium bursting activity, 57 which can increase both the strength and duration of a contraction (Figure 3). 30

FIGURE 3.

FIGURE 3

Examples of simple and complex action potentials (APs), Ca2+ bursts, and the resultant contractile force. Bursting activity can increase the duration of a contraction. RMP: resting membrane potential

2.2.2. Sodium channels

As reviewed recently by our group, studies have provided conflicting evidence as to the role of voltage‐gated sodium (Na+) channels in the myometrium in any species. 60 Voltage‐insensitive Na+ channels may slowly depolarize the myometrial cell membrane to the threshold required to activate LTCCs. 60 Reinl et al demonstrated that, in both humans and in mice, Na+ leaks into the myometrial cell through the voltage‐insensitive channel sodium leak current, nonselective (NALCN). 61 , 62 Smooth‐muscle specific deletion of NALCN in mice shortens myometrial action potential bursts and leads to delayed and dysfunctional labour. 62 Thus, NALCN was initially thought to increase myometrial excitability, at least in mice. However, a recent study showed that NALCN is upregulated by the pro‐quiescent hormone progesterone and downregulated by the pro‐contractile hormone oestrogen in human myometrial cells, consistent with a role for NALCN in maintaining myometrial quiescence. 63 This raises the possibility that NALCN functions differently in the mouse and human myometrium, although further studies are necessary to test this hypothesis.

2.2.3. Calcium channels

Ca2+ currents in human myometrial cells can be separated into two components: long‐lasting L‐type currents and transient T‐type currents (TTCCs). 50 Early studies showed that nifedipine‐sensitive LTCCs cause the rapid depolarization phase of the myometrial action potential. Expression of mRNAs encoding the ⍺1c pore‐forming subunit and β2 regulatory subunit of LTCCs increase before parturition in both guinea pigs and rats. 64 , 65 , 66 , 67 These mRNA changes seem to functionally increase both LTCC drug binding sites and LTCC function in both species, consistent with a conserved role for LTCCs in initiating myometrial contractions. 65 , 66

Researchers have used the LTCC inhibitor nifedipine and TTCC inhibitors nickel and mibefradil to dissect out the role of TTCCs in uterine contraction. Inhibiting TTCCs decreases the frequency of spontaneous contractions in both human and rat explanted uterine tissue, leading researchers to hypothesize that TTCCs play a role in initiating and pacing contractions. 68 , 69 , 70 This hypothesis is particularly attractive given findings that TTCCs contribute to pacemaker currents in sinoatrial nodal cells in the heart, 71 interstitial cells of Cajal in the intestine, 72 and lymphatic smooth muscle cells. 73

The nonselective cation channel TRPV4 may also allow Ca2+ to enter myometrial cells. Whereas LTCCs and TTCCs are activated by the less negative membrane potential at the end of pregnancy, TRPV4 is activated by stretch or heat. 74 TRPV4 expression and activity increases throughout gestation in rats, and TRPV4 expression is higher in myometrium from pregnant women than in myometrium from non‐pregnant women. 75 , 76 Silencing TRPV4 expression in human myometrial cells prevents them from contracting in a collagen gel matrix. 75 Similarly, a TRPV4 antagonist attenuates contractions in both rat and mouse myometrium. 75 , 76 , 77 Furthermore, treatment with TRPV4 antagonists delays parturition in two experimental models of preterm labour in mice. 75 These lines of evidence suggest that TRPV4 enables myometrial contractions in pregnant humans, mice and rats.

2.2.4. Potassium channels

Potassium channels underlie the repolarizing current to bring the myometrial cell back to basal states. The number of potassium channels that contribute to myometrial excitability is vast, as reviewed previously. 78 , 79 Here we will highlight only a few K+ channels know to contribute to uterine activity.

The repolarization phase of the myometrial action potential is driven by the action of voltage‐ and Ca2+‐gated potassium (K+) channels. The voltage‐gated, rapidly‐inactivating K+ currents initially identified by Mirroneau 51 , 52 are predominantly carried by the Kv4 family of K+ channels, which are abundant in both human and rat myometrium. 79 In ovariectomized rats and isolated human cells, oestrogen decreases Kv4 expression and function; consistent with this, Kv4 expression decreases throughout gestation in rats. 80 , 81

The Ca2+‐activated large conductance K+ channel (BKCa) also contributes to this repolarizing current and has been extensively studied in the myometrium. In both rats and humans, activation of BKCa causes myometrial relaxation, whereas inhibition of BKCa causes myometrial contractions. 78 In guinea pigs and humans, chorionic factors (i.e. human chorionic gonadotropin) that promote myometrial relaxation do so by activating BKCa. 82 , 83 However, BKCa knockout mice do not deliver preterm, perhaps indicating that other K+ channels compensate for loss of BKCa. 84 Nonetheless, recent evidence suggests that BKCa increases myometrial contractions under certain inflammatory conditions. Specifically, Wakle‐Prabagaran et al found that the pro‐inflammatory cytokine alpha‐2‐macroglobulin interacted with BKCa to potentiate Ca2+ oscillations in oxytocin‐treated human myometrial cells. 85

BKCa activity can be regulated by alternative splicing, interaction with auxiliary subunits, post‐translational modifications, hormonal modulation and several other mechanisms. 78 Here, we will focus on alternative splicing, a mechanism that appears to be species‐specific. In all species in which BKCa isoforms have been studied (rats, mice and humans), splice forms expressed at term are less sensitive to Ca2+ than those expressed earlier in pregnancy. The splice forms expressed at term would allow intracellular Ca2+ levels to rise without increasing K+ efflux and repolarizing the membrane. For example, in human myometrium, alternative splicing produces the mK44 isoform, which has 44 extra amino acids and is less sensitive to Ca2+ than the canonical form. 86 The 44 amino acid insertion also causes BKCa to undergo proteolytic cleavage so that only the N‐terminus reaches the cell membrane. 87 Production of mK44 increases at labour, while overall BKCa production decreases, 88 thus reducing the overall repolarizing current. In rats and mice, alternative splicing produces a different BKCa isoform containing the stress axis regulated exon (STREX). The 59 amino acids in STREX make this isoform more sensitive to Ca2+ than canonical BKCa, thus promoting myometrial repolarization. 89 However, STREX regulation is slightly different in mice and rats. In mouse myometrium, STREX is expressed throughout pregnancy, whereas BKCa without the STREX insertion is upregulated at term. 90 In rat myometrium, STREX expression is reduced by oestrogen and increased by progesterone; accordingly, its expression diminishes at the end of gestation. 91

Another family of K+ channels, SK channels, may be important for hyperpolarization after an action potential and therefore also promote myometrial relaxation. 92 Pierce et al showed that mice overexpressing SK3 channels had weaker uterine contractions. 93 Conversely, inhibiting SK channels with the selective inhibitor apamin increased spontaneous contractions in myometrial tissue samples from pregnant rats 94 and prevented relaxation in myometrial tissue samples from pregnant and non‐pregnant women. 95 , 96 Moreover SK3 expression is downregulated at term in both human and mouse myometrium, 93 , 97 consistent with a conserved role for SK3 in promoting myometrial quiescence.

Functional studies suggest that ATP‐sensitive inward rectifying K+ channels (KATP) also play a role in promoting myometrial quiescence. In general, activation and overexpression of KATP channels leads to myometrial relaxation in both rat and human tissue. 79 Consistent with this, in humans, KATP channel expression decreases at parturition, and KATP function further decreases during active labour. 98 , 99 However, in rats, KATP channel activity is similar between mid‐pregnancy and parturition. 100 Additionally, KATP channel openers could inhibit agonist‐induced contractions in human myometrial tissue, 99 but not in rat myometrial tissue. 101 Thus, KATP channels may play different roles in the rat and human myometrium, although further studies directly comparing the two species are necessary to confirm this conclusion.

Finally, the inward rectifying K+ channel Kir7.1 may play a role in regulating the resting membrane potential in the myometrium. Activation of Kir7.1 hyperpolarizes the cell membrane, thereby decreasing myometrial excitability. 102 Consistent with this role, Kir7.1 expression is highest during mid‐pregnancy in mice and declines towards term. 102 In humans, Kir7.1 expression decreases as the uterus transitions from the non‐labouring to the labouring state. 102 Pharmacological inhibition of Kir7.1 induces contractions in both mouse and human myometrial tissue strips. 102 Taken together, these results suggest that Kir7.1 promotes myometrial quiescence in both mice and humans.

The examples highlighted above show that while ion channels often have similar roles in several species, more extensive study almost always reveals species‐specific differences in channel regulation. Researchers must keep this possibility in mind when designing ion channel studies in rodent models.

2.3. Timing and propagation of action potentials

2.3.1. The uterine pacemaker

When Alvarez and Caldeyro measured intrauterine pressure and uterine electrical activity for the first time in the 1950s, 103 they proposed that labour contractions originated from a single “pacemaker” region. 104 However, with more advanced electromyography techniques, later investigators observed that multiple possible pacemaker regions could be active during a contraction. 105 Further work with newer technologies have confirmed this idea of multiple pacemakers. 106 In particular, Eswaran et al and Ramon et al used a 151‐channel magnetomyographic array and showed that electrical waves appeared “suddenly in random locations that were not contiguous or anatomically related to each other”. 107 , 108 More recently, Wu et al developed electromyometrial imaging, a non‐invasive technology that combines electrical data with magnetic resonance imaging to map electrical activity onto the entire three‐dimensional uterus. 109 Data collected by using this technique support the existence of multiple pacemaker sites in sheep, although this technology has not yet been reported in humans.

These studies in humans and large animals at parturition have been complemented by ex vivo multi‐electrode array (MEA) studies on rodent myometrium. 110 Several studies from Lammers and collaborators, who pioneered this technique in the myometrium, suggested that the coordinated uterine contractions at parturition result from action potentials propagating from many possible initiating pacemaker regions. 110 , 111 , 112 In guinea pigs, contractions can originate at the cervical or the ovarian end of the uterine horn and propagate in either direction. 112 , 113 In contrast, in rats, most electrical activities start at the ovarian end of the uterine horn and proceed toward the cervical end in a “peristaltic” contraction pattern. 112 These differences may be due to the fact that rats can carry up to 10 pups, whereas guinea pigs carry fewer offspring. Investigators recently proposed that multiple pacemakers in rat uterus originate from structures located in each implantation site called “myometrial‐placental pacemaker zones”. 114 These zones have not (yet) been identified in guinea pigs or in mice, and it is unclear whether these findings translate to humans.

Most MEA studies show that, in all animals tested (humans, sheep, guinea pigs and rats), action potentials do not propagate in one direction; instead, they tend to travel in tortuous paths before terminating. Thus, at least in large animals such as humans, an action potential originating at one point could not travel throughout the whole uterus fast enough to coordinate a contraction. Given these MEA studies and magnetomyography data showing that multiple pacemaker regions contribute to each contraction in humans, Young proposed a dual model for contractions. 115 In this model, a burst of action potentials in one pacemaker region propagates to produce a “local” contraction, which causes an increase in wall stress that activates another mechanically sensitive pacemaker region. This process theoretically continues until enough pacemaker regions are recruited to produce a coordinated contraction. However, more research is necessary to support this theory.

Although researchers may never identify a definitive uterine pacemaker region, the search is still on for a pacemaker cell type. In the gut, slow electrical waves initiate in pacemaker cells called interstitial cells of Cajal (ICCs) and then propagate to the rest of the gut myocytes. Several groups have sought to find an “ICC‐like cell” in the uterus, searching for interstitial cells that produce similar slow waves or express similar membrane markers. So‐called ICC‐like cells have been found in the uterus of humans, rats, mice, and monkeys, 116 , 117 , 118 , 119 , 120 but thus far, no group has definitively shown that these cells are capable of producing slow waves in isolation. Furthermore, myometrial cells may not exhibit the slow waves seen in the gut. 113 Thus, further research is needed to determine whether these ICC‐like cells act as pacemakers in any species.

2.3.2. Gap junctions

Regardless of where action potentials originate, their coordination and propagation depend on uterine gap junctions. Gap junctions consist of connexin 43 proteins that form connexons, or pores, between neighbouring myometrial cells. 121 These low‐resistance pathways allow electrical signals to travel through the uterus. 122 Early studies by Garfield's group in rat, guinea pig and human myometrium showed that gap junctions are scarce or absent in non‐pregnant and mid‐pregnant myometrium but become abundant at term. 123 , 124 , 125 Induction of preterm labour with RU486 or prostaglandin E2 induces gap junction formation in rats and guinea pigs, respectively. 122 , 125 In participants undergoing C‐section at various points during labour, gap junction abundance correlated with cervical dilation and the frequency of uterine contractions. 124 Finally, conditional knockout of connexin 43 delays parturition in mice. 126 Together, these lines of evidence indicate that gap junctions play a conserved role in synchronizing uterine contractions at parturition.

2.4. Pro‐contractile agents

2.4.1. Oxytocin and the oxytocin receptor

With many uses in both clinical practice and basic science research, oxytocin is perhaps the best‐known modulator of uterine contractility. Oxytocin is a nonapeptide hormone produced and secreted in a pulsatile fashion by the hypothalamus, as well as by the placenta, decidua and myometrium during labour. 127 In the United States, clinicians prescribe exogenous oxytocin to about half of patients to induce or augment labour and recommend that all patients receive oxytocin to prevent postpartum haemorrhage. 128 , 129 Researchers use oxytocin to induce contractions and simulate a pro‐labour environment in ex vivo experiments on uterine tissue and cells isolated from humans and animal models. In humans, rats, and mice, oxytocin binding sites in the uterus increase by 6‐ to 12‐fold throughout pregnancy and reaches a peak at or just before labour. 130 , 131 , 132 , 133 However, in guinea pigs, oxytocin binding capacity does not change significantly between mid‐pregnancy and late gestation (40 and 69 days of gestation, respectively). 134

As mentioned above and reviewed in detail elsewhere, 127 OXTR works classically by coupling to Gq to induce the release of intracellular Ca2+ stores and allow for muscle contraction (Figure 2). OXTR may also promote an increase in intracellular Ca2+ concentration by increasing SOCE and preventing Ca2+ efflux through the Ca2+ ATPase. 127 Additionally, OXTR inhibits the K+ efflux channel Slo2.1, thereby depolarizing the cell membrane toward the threshold required to induce an action potential. 135 In the longer term, oxytocin can work in the myometrium and other tissues to increase NFκB‐dependent transcription and increase production of pro‐inflammatory cytokines, cyclooxygenase 2, and prostaglandins. 136 , 137 , 138 In fact, some researchers have hypothesized that the most important function of oxytocin is to increase production of prostaglandins, which are also potent inducers of uterine contractions. 139

Pharmacological inhibition of oxytocin impairs parturition to varying degrees in mice, 140 rats, 141 and guinea pigs, 142 but clinical trials have shown that the oxytocin antagonists atosiban and barusiban may not be effective tocolytics in humans. 143 Mice that lack oxytocin or OXTR deliver normally, 144 , 145 , 146 showing definitively that mice can compensate for lack of oxytocin signalling when initiating labour. The OXTR gene has not been deleted in guinea pigs or rats, so it is unclear whether this is true in other rodents. In humans, however, loss‐of‐function OXTR mutations have been linked to preterm labour rather than to delayed or absent contractions. 147 These lines of evidence have led researchers to conclude that signalling via OXTR is sufficient, but not necessary, to initiate uterine contractions in all species studied.

2.4.2. Prostaglandins and prostaglandin receptors

Like oxytocin, synthetic prostaglandins are used clinically to induce and augment labour and to prevent postpartum haemorrhage. In vivo, prostaglandins are produced from arachidonic acid by the enzyme cyclooxygenase 2 (COX2). COX2 is upregulated in the uterus as pregnancy progresses towards labour, in part due to increased oxytocin signalling, causing an increase in local prostaglandin concentration. 136 , 148 , 149 The synthetic prostaglandins misoprostol (prostaglandin E1), dinoprostone (prostaglandin E2) and carboprost (prostaglandin F2) act on several GPCRs: the prostaglandin F2 receptor (FP) and four prostaglandin E receptors (EP1‐4). The pro‐contractile prostaglandin receptors—FP, EP1 and EP3 – work similarly to OXTR. FP and EP1 couple to Gq, whereas EP3 couples to Gi (Figure 2). 150 The relaxant receptors EP2 and EP4 couple to Gs to produce cyclic AMP and activate protein kinase A (Figure 2). 150 During parturition in human and rat myometrium, expression of the relaxant receptors EP2 and E4 is downregulated, while expression of the pro‐contractile prostaglandin receptors FP, EP1 and EP3 is upregulated. 151 , 152 Therefore, increased expression of prostaglandin receptors and induction of prostaglandin production by COX2 at parturition appear to stimulate uterine contraction in the rat and human uterus. However, in the guinea pig, EP2, EP3 and EP4 mRNA expression does not change at parturition. 153

Evidence from knockout mice highlights the necessity of prostaglandins for labour. Sugimoto et al found that mice that lacked the prostaglandin F receptor had normal fertility, but never delivered their pups. 154 However, when these knockout mice were ovariectomized at 19 days of pregnancy, the mice delivered normally. These results suggest that in mice, prostaglandin F is necessary for luteolysis and progesterone withdrawal but not for causing uterine contractions.

Clinicians and researchers hypothesized that COX2 inhibitors that prevent prostaglandin production would be effective tocolytics (see “Tocolytics” section below). Treatment with COX2 inhibitors delayed parturition in rats 155 , 156 and prevented lipopolysaccharide‐induced preterm labour in mice. 157 Of note, both rats and mice require luteolysis and progesterone withdrawal for parturition, unlike humans and guinea pigs. 6 The COX2 inhibitor indomethacin has been shown to inhibit uterine contractions in explanted tissue strips from mice, rats, guinea pigs, and humans. 164, 172, 173, 174 . Indomethacin is used clinically as a tocolytic, although results from clinical trials have been mixed. 158 Thus, although prostaglandins are clearly essential for parturition in rats and mice, prostaglandin signalling may be dispensable for labour induction in humans. However, more systematic study is necessary to support this conclusion.

2.5. Tocolytics

The agents most commonly used by obstetricians to inhibit contractions in preterm labour take advantage of the molecular mechanisms of uterine contractility described above. Their broad classes include Ca2+ channel blockers, which target LTCCs; COX2 inhibitors, which target prostaglandins; β2 adrenergic receptor agonists, which increase cAMP to cause Ca2+ desensitization; and the oxytocin receptor antagonists atosiban and barusiban. As shown in Table 2, all of these tocolytics were tested for efficacy in mice, rats and/or guinea pigs before being approved for human use. However, tocolytics that effectively prevent parturition in rodent models have varying success in actually preventing preterm labour in humans. 159 Although these failures could reflect physiological differences between animal models and humans, it is important to note that no experimental model can capture the range of environmental and genetic factors that contribute to human preterm birth. Thus, it is impossible to compare the results of a randomized controlled trial to an animal study.

TABLE 2.

Common tocolytics

Class Calcium channel blockers COX inhibitors β2AR agonists OXTR antagonists
Preferred agent Nifedipine Indomethacin Terbutaline Atosiban
Other drugs in class

Nicardipine

Diltiazem (not used)

Ibuprofen

Sulindac

Nimesulide

COX‐2 specific inhibitors (‐coxibs)

Salbutamol

Hexoprenaline

Ritodrine

Barusiban
Mechanism of action Block calcium entry from voltage‐gated calcium channels Block prostaglandin synthesis Activate Gs signalling to inhibit MLCK Inhibits oxytocin signalling
Clinical use 160 , 161 Nifedipine is first‐line therapy (32‐34 weeks); second‐line therapy (24‐32 weeks) Indomethacin is first‐line therapy (24‐32 weeks) Terbutaline is second‐line therapy (32‐34 weeks) Not available in the United States
Species considerations Efficacy originally demonstrated in rat. 162 , 163 Nifedipine also inhibits oxytocin‐induced contractions in explanted uterus from mice. 92 Indomethacin initially tested in rats, 156 successfully inhibits LPS‐induced preterm birth in mice. 157 Indomethacin inhibits spontaneous contractions in explanted guinea pig uterus. 164 Direct comparison of effect of ritodrine on isolated myometrial strips from rat and human myometrium showed identical results. 165 Efficacy originally demonstrated in rat and guinea pig. 166 , 167 Efficacy in humans in is contested. 143

Nevertheless, experiments comparing the effects of tocolytic drugs on explanted uterine tissue may shed some light on how tocolytics affect uterine contractions in different species. In Table 3, we compiled IC50 values of several tocolytics in ex vivo uterine tension recording experiments. These results are easiest to interpret when researchers use the same experimental conditions to assay contractions in multiple species, as in one study by Kantas. 165 Comparing results from separate studies is more difficult, especially when considering systematic differences between experimental setups. For example, researchers may use nonpregnant or pregnant animals and may measure spontaneous or induced contractions, as indicated in Table 3. Other methodological differences—such as different tension recording setups, dissection methods, or salt concentrations in buffer solutions—are even more difficult to account for.

TABLE 3.

IC50 values for tocolytic drugs

Mouse Rat Guinea pig Human
Nifedipine (calcium channel blocker) «1 × 10−6 mol/L a (nonpregnant, spontaneous contractions) 92 ~1 × 10−7 mol/L (gestation day 22, contractions induced by 25 mmol/L KCl) 168 ~1 × 10−6 mol/L (gestation day 62‐66, PGF‐induced contractions) 169

5.5 × 10−8 mol/L (gestation weeks 38‐40, spontaneous contractions) 170

~1 × 10−7 mol/L (gestation weeks 36‐40, spontaneous contractions) 171

Indomethacin (COX inhibitor)

3 × 10−5 mol/L

(gestation day 19, spontaneous contractions) 172

1.8 × 10−5 mol/L (gestation day 18, spontaneous contractions) 173 «1 × 10−5 mol/L (gestation day 7‐20, spontaneous contractions) 164 ~1 × 10−4 mol/L (term pregnancy, spontaneous contractions) 174
Ritodrine (β2AR agonist) «1 × 10−10 mol/L a (nonpregnant, spontaneous contractions) 175

~1 × 10−6 mol/L (gestation day 19‐21, spontaneous contractions) 165

3.1 × 10−7 mol/L (gestation day 18, spontaneous contractions) 173

Not available

~1 × 10−6 mol/L (term pregnancy, spontaneous contractions) 165

~1 × 10−6 mol/L

(gestation weeks 38‐40, oxytocin‐induced contractions 176

Atosiban (OXTR antagonist) 3 × 10−7 mol/L (gestation day 19, spontaneous contractions) 172 1 × 10−7 mol/L (gestation day 18, spontaneous contractions) 173 ~4 × 10−7 mol/L (gestation day 60‐65, oxytocin‐induced contractions) 177 ~6 × 10−8 mol/L (gestation weeks 38‐40, oxytocin‐induced contractions) 176
a

Indicates dose used to achieve maximal inhibition (no dose‐response curve shown in study).

3. CONCLUSIONS AND OPPORTUNITIES FOR FURTHER RESEARCH

As mentioned in the introduction of this review, each animal model has experimental benefits and limitations that make it difficult to determine whether certain features of pregnancy and parturition are conserved between species. Knockout studies and randomized controlled trials are considered the gold standards of evidence in animal and human research, but there is no human equivalent of a knock‐out mouse and no rodent equivalent of a randomized controlled trial. Thus, results that appear divergent may reflect unavoidable experimental differences rather than physiological differences.

Here, we attempted to draw conclusions about species‐level differences by comparing results from different studies, often conducted by different groups, that use similar experimental approaches (Table 1). However, even this imperfect strategy reveals significant gaps in the literature. In particular, relatively few molecular studies have been performed in guinea pig myometrium to characterize ion channels and receptors that have been extensively studied in other animals. Because both humans and guinea pigs maintain high progesterone levels even in late pregnancy, 6 molecular studies may provide insight into hormonal regulation of contractile‐associated proteins that may be more translatable to human physiology than similar studies in the mouse or rat.

Our review of the literature for this article revealed a lack of true comparative studies, in which researchers use identical techniques to analyse uterine tissue or cells from multiple species. One rare example was performed by Kantas et al, who used tension recording to compare ex vivo contractility between rat and human myometrial strips. 165 Other techniques such as patch clamping, MEA, and gene and protein expression analyses are equally suited for species comparison.

Furthermore, the advent of unbiased, high‐information approaches such as RNA sequencing now allows researchers to conduct systematic studies of pregnancy and uterine contractions across species. These studies would identify species differences in the expression of important pro‐contractile and pro‐quiescent genes throughout pregnancy, which would help to confirm the fragmented conclusions drawn from disparate studies in Table 1. Similarly, proteomic and metabolomic studies may provide insight into how hormones and inflammatory markers vary between species. These systematic studies would help investigators determine the most appropriate animal model for their questions, potentially improving the translatability of their study. In the absence of such a systematic comparison, researchers must rely on the existing literature to determine the most appropriate animal model for their study that takes into account the study question and needs for experimental tractability and translatability.

CONFLICT OF INTEREST

The original draft for this review was adapted from a white paper commissioned by Corteva Agriscience on species differences in uterine contractions. This draft was rewritten and expanded to be more useful to a general audience.

ACKNOWLEDGEMENTS

The writing of the original draft of this review was funded by Corteva Agriscience, which commissioned a white paper on species differences in uterine contractions. Manasi Malik is funded by the National Institute of Child Health and Human Development (F30 HD097925). Sarah K. England is funded by the National Institute of Child Health and Human Development (R01HD088097, R01HD096737, and R01HD037831) and the March of Dimes (#6‐FY18‐664). The authors thank Dr Deborah Frank for her invaluable assistance in editing this manuscript. Figures were designed with BioRender.com.

Malik M, Roh M, England SK. Uterine contractions in rodent models and humans. Acta Physiol.2021;231:e13607. 10.1111/apha.13607

REFERENCES

  • 1. Creanga AA, Syverson C, Seed K, Callaghan WM. Pregnancy‐related mortality in the United States, 2011–2013. Obstet Gynecol. 2017;130(2):366‐373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Say L, Chou D, Gemmill A, et al. Global causes of maternal death: a WHO systematic analysis. Lancet Glob Health. 2014;2(6):e323‐e333. [DOI] [PubMed] [Google Scholar]
  • 3. Liu L, Oza S, Hogan D, et al. Global, regional, and national causes of under‐5 mortality in 2000–15: an updated systematic analysis with implications for the Sustainable Development Goals. Lancet. 2016;388(10063):3027‐3035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Ratajczak CK, Fay JC, Muglia LJ. Preventing preterm birth: the past limitations and new potential of animal models. Dis Model Mech. 2010;3(7–8):407‐414. [DOI] [PubMed] [Google Scholar]
  • 5. Arrowsmith S, Keov P, Muttenthaler M, Gruber CW. Contractility measurements of human uterine smooth muscle to aid drug development. J Vis Exp. 2018;131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Mitchell BF, Taggart MJ. Are animal models relevant to key aspects of human parturition? Am J Physiol Regul Integr Comp Physiol. 2009;297(3):R525‐545. [DOI] [PubMed] [Google Scholar]
  • 7. Dudley DJ, Branch DW, Edwin SS, Mitchell MD. Induction of preterm birth in mice by RU486. Biol Reprod. 1996;55(5):992‐995. [DOI] [PubMed] [Google Scholar]
  • 8. Skarnes RC, Harper MJ. Relationship between endotoxin‐induced abortion and the synthesis of prostaglandin F. Prostaglandins. 1972;1(3):191‐203. [DOI] [PubMed] [Google Scholar]
  • 9. Zarrow MX, Anderson NC, Callantine MR. Failure of progestogens to prolong pregnancy in the guinea pig. Nature. 1963;198:690‐692. [DOI] [PubMed] [Google Scholar]
  • 10. Dodd JM, Jones L, Flenady V, Cincotta R, Crowther CA. Prenatal administration of progesterone for preventing preterm birth in women considered to be at risk of preterm birth. Cochrane Database Syst Rev. 2013;7:CD004947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Nadeem L, Shynlova O, Matysiak‐Zablocki E, Mesiano S, Dong X, Lye S. Molecular evidence of functional progesterone withdrawal in human myometrium. Nat Commun. 2016;7:11565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Scaffidi J, Mol BW, Keelan JA. The pregnant women as a drug orphan: a global survey of registered clinical trials of pharmacological interventions in pregnancy. BJOG. 2017;124(1):132‐140. [DOI] [PubMed] [Google Scholar]
  • 13. McCarthy R, Martin‐Fairey C, Sojka DK, et al. Mouse models of preterm birth: suggested assessment and reporting guidelines. Biol Reprod. 2018;99(5):922‐937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ratajczak CK, Muglia LJ. Insights into parturition biology from genetically altered mice. Pediatr Res. 2008;64(6):581‐589. [DOI] [PubMed] [Google Scholar]
  • 15. Somlyo AP, Somlyo AV. Signal transduction and regulation in smooth muscle. Nature. 1994;372(6503):231‐236. [DOI] [PubMed] [Google Scholar]
  • 16. Aguilar HN, Mitchell BF. Physiological pathways and molecular mechanisms regulating uterine contractility. Hum Reprod Update. 2010;16(6):725‐744. [DOI] [PubMed] [Google Scholar]
  • 17. Lartey J, López BA. RHO protein regulation of contraction in the human uterus. Reproduction. 2009;138(3):407‐424. [DOI] [PubMed] [Google Scholar]
  • 18. Woodcock NA, Taylor CW, Thornton S. Prostaglandin F2alpha increases the sensitivity of the contractile proteins to Ca2+ in human myometrium. Am J Obstet Gynecol. 2006;195(5):1404‐1406. [DOI] [PubMed] [Google Scholar]
  • 19. Tahara M, Morishige K, Sawada K, et al. RhoA/Rho‐kinase cascade is involved in oxytocin‐induced rat uterine contraction. Endocrinology. 2002;143(3):920‐929. [DOI] [PubMed] [Google Scholar]
  • 20. Woodcock NA, Taylor CW, Thornton S. Effect of an oxytocin receptor antagonist and rho kinase inhibitor on the [Ca++]i sensitivity of human myometrium. Am J Obstet Gynecol. 2004;190(1):222‐228. [DOI] [PubMed] [Google Scholar]
  • 21. Somlyo AP, Somlyo AV. From pharmacomechanical coupling to G‐proteins and myosin phosphatase. Acta Physiol Scand. 1998;164(4):437‐448. [DOI] [PubMed] [Google Scholar]
  • 22. Moore F, Bernal AL. Myosin light chain kinase and the onset of labour in humans. Exp Physiol. 2001;86(2):313‐318. [DOI] [PubMed] [Google Scholar]
  • 23. Smith AF, Bigsby RM, Word RA, Herring BP. A 310‐bp minimal promoter mediates smooth muscle cell‐specific expression of telokin. Am J Physiol. 1998;274(5):C1188‐C1195. discussion C1187. [DOI] [PubMed] [Google Scholar]
  • 24. Herring BP, El‐Mounayri O, Gallagher PJ, Yin F, Zhou J. Regulation of myosin light chain kinase and telokin expression in smooth muscle tissues. Am J Physiol Cell Physiol. 2006;291(5):C817‐C827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Shmygol A, Gullam J, Blanks A, Thornton S. Multiple mechanisms involved in oxytocin‐induced modulation of myometrial contractility. Acta Pharmacol Sin. 2006;27(7):827‐832. [DOI] [PubMed] [Google Scholar]
  • 26. Mikoshiba K. Role of IP3 receptor signaling in cell functions and diseases. Adv Biol Regul. 2015;57:217‐227. [DOI] [PubMed] [Google Scholar]
  • 27. Noble K, Matthew A, Burdyga T, Wray S. A review of recent insights into the role of the sarcoplasmic reticulum and Ca entry in uterine smooth muscle. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S11‐S19. [DOI] [PubMed] [Google Scholar]
  • 28. Matsuki K, Takemoto M, Suzuki Y, et al. Ryanodine receptor type 3 does not contribute to contractions in the mouse myometrium regardless of pregnancy. Pflugers Arch. 2017;469(2):313‐326. [DOI] [PubMed] [Google Scholar]
  • 29. Burdyga T, Wray S, Noble K. In situ calcium signaling: no calcium sparks detected in rat myometrium. Ann N Y Acad Sci. 2007;1101:85‐96. [DOI] [PubMed] [Google Scholar]
  • 30. Burdyga T, Borisova L, Burdyga AT, Wray S. Temporal and spatial variations in spontaneous Ca events and mechanical activity in pregnant rat myometrium. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S25‐S32. [DOI] [PubMed] [Google Scholar]
  • 31. Dabertrand F, Fritz N, Mironneau J, Macrez N, Morel JL. Role of RYR3 splice variants in calcium signaling in mouse nonpregnant and pregnant myometrium. Am J Physiol Cell Physiol. 2007;293(3):C848‐C854. [DOI] [PubMed] [Google Scholar]
  • 32. Awad SS, Lamb HK, Morgan JM, Dunlop W, Gillespie JI. Differential expression of ryanodine receptor RyR2 mRNA in the non‐pregnant and pregnant human myometrium. Biochem J. 1997;322(Pt 3):777‐783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Martin C, Chapman KE, Thornton S, Ashley RH. Changes in the expression of myometrial ryanodine receptor mRNAs during human pregnancy. Biochim Biophys Acta. 1999;1451(2–3):343‐352. [DOI] [PubMed] [Google Scholar]
  • 34. Taggart MJ, Wray S. Contribution of sarcoplasmic reticular calcium to smooth muscle contractile activation: gestational dependence in isolated rat uterus. J Physiol. 1998;511(Pt 1):133‐144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Kupittayanant S, Luckas MJ, Wray S. Effect of inhibiting the sarcoplasmic reticulum on spontaneous and oxytocin‐induced contractions of human myometrium. BJOG. 2002;109(3):289‐296. [DOI] [PubMed] [Google Scholar]
  • 36. Wray S, Prendergast C. The myometrium: from excitation to contractions and labour. Adv Exp Med Biol. 2019;1124:233‐263. [DOI] [PubMed] [Google Scholar]
  • 37. Putney JW. Forms and functions of store‐operated calcium entry mediators. STIM and Orai. Adv Biol Regul. 2018;68:88‐96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Feldman CH, Grotegut CA, Rosenberg PB. The role of STIM1 and SOCE in smooth muscle contractility. Cell Calcium. 2017;63:60‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Chin‐Smith EC, Slater DM, Johnson MR, Tribe RM. STIM and Orai isoform expression in pregnant human myometrium: a potential role in calcium signaling during pregnancy. Front Physiol. 2014;5:169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Murtazina DA, Chung D, Ulloa A, Bryan E, Galan HL, Sanborn BM. TRPC1, STIM1, and ORAI influence signal‐regulated intracellular and endoplasmic reticulum calcium dynamics in human myometrial cells. Biol Reprod. 2011;85(2):315‐326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Noble D, Borysova L, Wray S, Burdyga T. Store‐operated Ca2⁺ entry and depolarization explain the anomalous behaviour of myometrial SR: effects of SERCA inhibition on electrical activity, Ca2⁺ and force. Cell Calcium. 2014;56(3):188‐194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Ward SM, Ordog T, Koh SD, et al. Pacemaking in interstitial cells of Cajal depends upon calcium handling by endoplasmic reticulum and mitochondria. J Physiol. 2000;525(Pt 2):355‐361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Exintaris B, Nguyen DT, Lam M, Lang RJ. Inositol trisphosphate‐dependent Ca stores and mitochondria modulate slow wave activity arising from the smooth muscle cells of the guinea pig prostate gland. Br J Pharmacol. 2009;156(7):1098‐1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Gravina FS, Parkington HC, Kerr KP, et al. Role of mitochondria in contraction and pacemaking in the mouse uterus. Br J Pharmacol. 2010;161(6):1375‐1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Parkington HC, Tonta MA, Brennecke SP, Coleman HA. Contractile activity, membrane potential, and cytoplasmic calcium in human uterine smooth muscle in the third trimester of pregnancy and during labor. Am J Obstet Gynecol. 1999;181(6):1445‐1451. [DOI] [PubMed] [Google Scholar]
  • 46. Casteels R, Kuriyama H. Membrane potential and ionic content in pregnant and non‐pregnant rat myometrium. J Physiol. 1965;177:263‐287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Kuriyama H, Suzuki H. Changes in electrical properties of rat myometrium during gestation and following hormonal treatments. J Physiol. 1976;260(2):315‐333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Parkington HC, Coleman HA. The role of membrane potential in the control of uterine motility. In CM E, MJ D, eds. Uterine function: molecular and cellular aspects (pp. 195‐248). Boston, MA: Springer; 1990: [Google Scholar]
  • 49. Shmigol AV, Eisner DA, Wray S. Properties of voltage‐activated [Ca2+]i transients in single smooth muscle cells isolated from pregnant rat uterus. J Physiol. 1998;511(Pt 3):803‐811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Young RC, Smith LH, McLaren MD. T‐type and L‐type calcium currents in freshly dispersed human uterine smooth muscle cells. Am J Obstet Gynecol. 1993;169(4):785‐792. [DOI] [PubMed] [Google Scholar]
  • 51. Mironneau J, Savineau JP. Effects of calcium ions on outward membrane currents in rat uterine smooth muscle. J Physiol. 1980;302:411‐425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Mironneau J, Savineau JP, Mironneau C. Fast outward current controlling electrical activity in rat uterine smooth muscle during gestation. J Physiol (Paris). 1981;77(8):851‐859. [PubMed] [Google Scholar]
  • 53. Parkington HC, Coleman HA. Ionic mechanisms underlying action potentials in myometrium. Clin Exp Pharmacol Physiol. 1988;15(9):657‐665. [DOI] [PubMed] [Google Scholar]
  • 54. Izumi H, Kishikawa T. Effects of ritodrine, a beta 2‐adrenoceptor agonist, on smooth muscle cells of the myometrium of pregnant rats. Br J Pharmacol. 1982;76(3):463‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Osa T, Fujino T. Electrophysiological comparison between the longitudinal and circular muscles of the rat uterus during the estrous cycle and pregnancy. Jpn J Physiol. 1978;28(2):197‐209. [DOI] [PubMed] [Google Scholar]
  • 56. Inoue Y, Nakao K, Okabe K, et al. Some electrical properties of human pregnant myometrium. Am J Obstet Gynecol. 1990;162(4):1090‐1098. [DOI] [PubMed] [Google Scholar]
  • 57. Nakao K, Inoue Y, Okabe K, Kawarabayashi T, Kitamura K. Oxytocin enhances action potentials in pregnant human myometrium–a study with microelectrodes. Am J Obstet Gynecol. 1997;177(1):222‐228. [DOI] [PubMed] [Google Scholar]
  • 58. Kawarabayashi T, Sugimori H. Effects of oxytocin and prostaglandin F2 alpha on pregnant human myometrium recorded by the single sucrose‐gap method–comparison of an in vitro experiment and an in vivo trial. Asia Oceania J Obstet Gynaecol. 1985;11(2):247‐253. [DOI] [PubMed] [Google Scholar]
  • 59. Kawarabayashi T, Kishikawa T, Sugimori H. Effect of oxytocin on spontaneous electrical and mechanical activities in pregnant human myometrium. Am J Obstet Gynecol. 1986;155(3):671‐676. [DOI] [PubMed] [Google Scholar]
  • 60. Amazu C, Ferreira JJ, Santi CM, England SK. Sodium channels and transporters in the myometrium. Curr Opin Physiol. 2020;13:141‐144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Reinl EL, Cabeza R, Gregory IA, Cahill AG, England SK. Sodium leak channel, non‐selective contributes to the leak current in human myometrial smooth muscle cells from pregnant women. Mol Hum Reprod. 2015;21(10):816‐824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Reinl EL, Zhao P, Wu W, et al. Na+‐Leak channel, non‐selective (NALCN) regulates myometrial excitability and facilitates successful parturition. Cell Physiol Biochem. 2018;48(2):503‐515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Amazu C, Ma X, Henkes C, Ferreira JJ, Santi CM, England SK. Progesterone and estrogen regulate NALCN expression in human myometrial smooth muscle cells. Am J Physiol Endocrinol Metab. 2020;318(4):E441‐E452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Tezuka N, Ali M, Chwalisz K, Garfield RE. Changes in transcripts encoding calcium channel subunits of rat myometrium during pregnancy. Am J Physiol. 1995;269(4 Pt 1):C1008‐C1017. [DOI] [PubMed] [Google Scholar]
  • 65. Collins PL, Moore JJ, Lundgren DW, Choobineh E, Chang SM, Chang AS. Gestational changes in uterine L‐type calcium channel function and expression in guinea pig. Biol Reprod. 2000;63(5):1262‐1270. [DOI] [PubMed] [Google Scholar]
  • 66. Mershon JL, Mikala G, Schwartz A. Changes in the expression of the L‐type voltage‐dependent calcium channel during pregnancy and parturition in the rat. Biol Reprod. 1994;51(5):993‐999. [DOI] [PubMed] [Google Scholar]
  • 67. Ohkubo T, Kawarabayashi T, Inoue Y, Kitamura K. Differential expression of L‐ and T‐type calcium channels between longitudinal and circular muscles of the rat myometrium during pregnancy. Gynecol Obstet Invest. 2005;59(2):80‐85. [DOI] [PubMed] [Google Scholar]
  • 68. Blanks AM, Zhao ZH, Shmygol A, Bru‐Mercier G, Astle S, Thornton S. Characterization of the molecular and electrophysiological properties of the T‐type calcium channel in human myometrium. J Physiol. 2007;581(Pt 3):915‐926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Young RC, Zhang P. Inhibition of in vitro contractions of human myometrium by mibefradil, a T‐type calcium channel blocker: support for a model using excitation‐contraction coupling, and autocrine and paracrine signaling mechanisms. J Soc Gynecol Investig. 2005;12(4):e7‐e12. [DOI] [PubMed] [Google Scholar]
  • 70. Lee SE, Ahn DS, Lee YH. Role of T‐type Ca channels in the spontaneous phasic contraction of pregnant rat uterine smooth muscle. Korean J Physiol Pharmacol. 2009;13(3):241‐249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Hagiwara N, Irisawa H, Kameyama M. Contribution of two types of calcium currents to the pacemaker potentials of rabbit sino‐atrial node cells. J Physiol. 1988;395:233‐253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Zheng H, Park KS, Koh SD, Sanders KM. Expression and function of a T‐type Ca2+ conductance in interstitial cells of Cajal of the murine small intestine. Am J Physiol Cell Physiol. 2014;306(7):C705‐C713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. To KHT, Gui P, Li M, Zawieja SD, Castorena‐Gonzalez JA, Davis MJ. T‐type, but not L‐type, voltage‐gated calcium channels are dispensable for lymphatic pacemaking and spontaneous contractions. Sci Rep. 2020;10(1):70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Shibasaki K. TRPV4 ion channel as important cell sensors. J Anesth. 2016;30(6):1014‐1019. [DOI] [PubMed] [Google Scholar]
  • 75. Ying L, Becard M, Lyell D, et al. The transient receptor potential vanilloid 4 channel modulates uterine tone during pregnancy. Sci Transl Med. 2015;7(319):319ra204. [DOI] [PubMed] [Google Scholar]
  • 76. Ducza E, Csányi A, Szőke É, et al. Significance of transient receptor potential vanilloid 4 and aquaporin 5 co‐expression in the rat uterus at term. Heliyon. 2019;5(10):e02697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Singh V, Ram M, Kandasamy K, et al. Molecular and functional characterization of TRPV4 channels in pregnant and nonpregnant mouse uterus. Life Sci. 2015;122:51‐58. [DOI] [PubMed] [Google Scholar]
  • 78. Lorca RA, Prabagaran M, England SK. Functional insights into modulation of BKCa channel activity to alter myometrial contractility. Front Physiol. 2014;5:289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Brainard AM, Korovkina VP, England SK. Potassium channels and uterine function. Semin Cell Dev Biol. 2007;18(3):332‐339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Suzuki T, Takimoto K. Differential expression of Kv4 pore‐forming and KChIP auxiliary subunits in rat uterus during pregnancy. Am J Physiol Endocrinol Metab. 2005;288(2):E335‐E341. [DOI] [PubMed] [Google Scholar]
  • 81. Song M, Helguera G, Eghbali M, et al. Remodeling of Kv4.3 potassium channel gene expression under the control of sex hormones. J Biol Chem. 2001;276(34):31883‐31890. [DOI] [PubMed] [Google Scholar]
  • 82. Doheny HC, Houlihan DD, Ravikumar N, Smith TJ, Morrison JJ. Human chorionic gonadotrophin relaxation of human pregnant myometrium and activation of the BKCa channel. J Clin Endocrinol Metab. 2003;88(9):4310‐4315. [DOI] [PubMed] [Google Scholar]
  • 83. Carvajal JA, Thompson LP, Weiner CP. Chorion‐induced myometrial relaxation is mediated by large‐conductance Ca2+‐activated K+ channel opening in the guinea pig. Am J Obstet Gynecol. 2003;188(1):84‐91. [DOI] [PubMed] [Google Scholar]
  • 84. Meredith AL, Thorneloe KS, Werner ME, Nelson MT, Aldrich RW. Overactive bladder and incontinence in the absence of the BK large conductance Ca2+‐activated K+ channel. J Biol Chem. 2004;279(35):36746‐36752. [DOI] [PubMed] [Google Scholar]
  • 85. Wakle‐Prabagaran M, Lorca RA, Ma X, et al. BKCa channel regulates calcium oscillations induced by alpha‐2‐macroglobulin in human myometrial smooth muscle cells. Proc Natl Acad Sci USA. 2016;113(16):E2335‐E2344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Korovkina VP, Fergus DJ, Holdiman AJ, England SK. Characterization of a novel 132‐bp exon of the human maxi‐K channel. Am J Physiol Cell Physiol. 2001;281(1):C361‐C367. [DOI] [PubMed] [Google Scholar]
  • 87. Korovkina VP, Brainard AM, England SK. Translocation of an endoproteolytically cleaved maxi‐K channel isoform: mechanisms to induce human myometrial cell repolarization. J Physiol. 2006;573(Pt 2):329‐341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Curley M, Morrison JJ, Smith TJ. Analysis of Maxi‐K alpha subunit splice variants in human myometrium. Reprod Biol Endocrinol. 2004;2:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Saito M, Nelson C, Salkoff L, Lingle CJ. A cysteine‐rich domain defined by a novel exon in a slo variant in rat adrenal chromaffin cells and PC12 cells. J Biol Chem. 1997;272(18):11710‐11717. [DOI] [PubMed] [Google Scholar]
  • 90. Benkusky NA, Fergus DJ, Zucchero TM, England SK. Regulation of the Ca2+‐sensitive domains of the maxi‐K channel in the mouse myometrium during gestation. J Biol Chem. 2000;275(36):27712‐27719. [DOI] [PubMed] [Google Scholar]
  • 91. Zhu N, Eghbali M, Helguera G, Song M, Stefani E, Toro L. Alternative splicing of Slo channel gene programmed by estrogen, progesterone and pregnancy. FEBS Lett. 2005;579(21):4856‐4860. [DOI] [PubMed] [Google Scholar]
  • 92. Brown A, Cornwell T, Korniyenko I, et al. Myometrial expression of small conductance Ca2+‐activated K+ channels depresses phasic uterine contraction. Am J Physiol Cell Physiol. 2007;292(2):C832‐C840. [DOI] [PubMed] [Google Scholar]
  • 93. Pierce SL, Kresowik JD, Lamping KG, England SK. Overexpression of SK3 channels dampens uterine contractility to prevent preterm labor in mice. Biol Reprod. 2008;78(6):1058‐1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Noble K, Floyd R, Shmygol A, Mobasheri A, Wray S. Distribution, expression and functional effects of small conductance Ca‐activated potassium (SK) channels in rat myometrium. Cell Calcium. 2010;47(1):47‐54. [DOI] [PubMed] [Google Scholar]
  • 95. Modzelewska B, Kostrzewska A, Sipowicz M, Kleszczewski T, Batra S. Apamin inhibits NO‐induced relaxation of the spontaneous contractile activity of the myometrium from non‐pregnant women. Reprod Biol Endocrinol. 2003;1:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Modzelewska B, Kleszczewski T, Kostrzewska A. The effect of a selective inhibition of potassium channels on the relaxation induced by nitric oxide in the human pregnant myometrium. Cell Mol Biol Lett. 2003;8(1):69‐75. [PubMed] [Google Scholar]
  • 97. Mazzone JN, Kaiser RA, Buxton IL. Calcium‐activated potassium channel expression in human myometrium: effect of pregnancy. Proc West Pharmacol Soc. 2002;45:184‐186. [PubMed] [Google Scholar]
  • 98. Curley M, Cairns MT, Friel AM, McMeel OM, Morrison JJ, Smith TJ. Expression of mRNA transcripts for ATP‐sensitive potassium channels in human myometrium. Mol Hum Reprod. 2002;8(10):941‐945. [DOI] [PubMed] [Google Scholar]
  • 99. Longo M, Jain V, Vedernikov YP, Hankins GD, Garfield RE, Saade GR. Effects of L‐type Ca(2+)‐channel blockade, K(+)(ATP)‐channel opening and nitric oxide on human uterine contractility in relation to gestational age and labour. Mol Hum Reprod. 2003;9(3):159‐164. [DOI] [PubMed] [Google Scholar]
  • 100. Okawa T, Longo M, Vedernikov YP, Chwalisz K, Saade GR, Garfield RE. Role of nucleotide cyclases in the inhibition of pregnant rat uterine contractions by the openers of potassium channels. Am J Obstet Gynecol. 2000;182(4):913‐918. [DOI] [PubMed] [Google Scholar]
  • 101. Bailie CA, Vedernikov YP, Saade GR, Garfield RE. Prostaglandin‐induced activation of uterine contractility in pregnant rats does not involve potassium channels. Am J Obstet Gynecol. 2002;186(3):453‐457. [DOI] [PubMed] [Google Scholar]
  • 102. McCloskey C, Rada C, Bailey E, et al. The inwardly rectifying K+ channel KIR7.1 controls uterine excitability throughout pregnancy. EMBO Mol Med. 2014;6(9):1161‐1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Alvarez H, Caldeyro R. Contractility of the human uterus recorded by new methods. Surg Gynecol Obstet. 1950;91(1):1‐13. [PubMed] [Google Scholar]
  • 104. Csapo A. The diagnostic significance of the intrauterine pressure. II. Clinical considerations and trials. Obstet Gynecol Surv. 1970;25(6):515‐543. [DOI] [PubMed] [Google Scholar]
  • 105. Wolfs GM, van Leeuwen M. Electromyographic observations on the human uterus during labour. Acta Obstet Gynecol Scand Suppl. 1979;90:1‐61. [DOI] [PubMed] [Google Scholar]
  • 106. Young RC. The uterine pacemaker of labor. Best Pract Res Clin Obstet Gynaecol. 2018;52:68‐87. [DOI] [PubMed] [Google Scholar]
  • 107. Eswaran H, Preissl H, Wilson JD, Murphy P, Robinson SE, Lowery CL. First magnetomyographic recordings of uterine activity with spatial‐temporal information with a 151‐channel sensor array. Am J Obstet Gynecol. 2002;187(1):145‐151. [DOI] [PubMed] [Google Scholar]
  • 108. Ramon C, Preissl H, Murphy P, Wilson JD, Lowery C, Eswaran H. Synchronization analysis of the uterine magnetic activity during contractions. Biomed Eng Online. 2005;4:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Wu W, Wang H, Zhao P, et al. Noninvasive high‐resolution electromyometrial imaging of uterine contractions in a translational sheep model. Sci Transl Med. 2019;11(483):eaau1428. [DOI] [PubMed] [Google Scholar]
  • 110. Lammers WJ, Arafat K, el‐Kays A, el‐Sharkawy TY. Spatial and temporal variations in local spike propagation in the myometrium of the 17‐day pregnant rat. Am J Physiol. 1994;267(5 Pt 1):C1210‐C1223. [DOI] [PubMed] [Google Scholar]
  • 111. Lammers WJ, Mirghani H, Stephen B, et al. Patterns of electrical propagation in the intact pregnant guinea pig uterus. Am J Physiol Regul Integr Comp Physiol. 2008;294(3):R919‐R928. [DOI] [PubMed] [Google Scholar]
  • 112. Lammers WJ, Stephen B, Al‐Sultan MA, Subramanya SB, Blanks AM. The location of pacemakers in the uteri of pregnant guinea pigs and rats. Am J Physiol Regul Integr Comp Physiol. 2015;309(11):R1439‐R1446. [DOI] [PubMed] [Google Scholar]
  • 113. Lammers WJ. The electrical activities of the uterus during pregnancy. Reprod Sci. 2013;20(2):182‐189. [DOI] [PubMed] [Google Scholar]
  • 114. Lutton EJ, Lammers WJEP, James S, van den Berg HA, Blanks AM. Identification of uterine pacemaker regions at the myometrial‐placental interface in the rat. J Physiol. 2018;596(14):2841‐2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Young RC. Mechanotransduction mechanisms for coordinating uterine contractions in human labor. Reproduction. 2016;152(2):R51‐R61. [DOI] [PubMed] [Google Scholar]
  • 116. Ciontea SM, Radu E, Regalia T, et al. C‐kit immunopositive interstitial cells (Cajal‐type) in human myometrium. J Cell Mol Med. 2005;9(2):407‐420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Cretoiu SM, Cretoiu D, Marin A, Radu BM, Popescu LM. Telocytes: ultrastructural, immunohistochemical and electrophysiological characteristics in human myometrium. Reproduction. 2013;145(4):357‐370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Cretoiu SM, Radu BM, Banciu A, et al. Isolated human uterine telocytes: immunocytochemistry and electrophysiology of T‐type calcium channels. Histochem Cell Biol. 2015;143(1):83‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Peri LE, Koh BH, Ward GK, et al. A novel class of interstitial cells in the mouse and monkey female reproductive tracts. Biol Reprod. 2015;92(4):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Duquette RA, Shmygol A, Vaillant C, et al. Vimentin‐positive, c‐kit‐negative interstitial cells in human and rat uterus: a role in pacemaking? Biol Reprod. 2005;72(2):276‐283. [DOI] [PubMed] [Google Scholar]
  • 121. Tabb T, Thilander G, Grover A, Hertzberg E, Garfield R. An immunochemical and immunocytologic study of the increase in myometrial gap junctions (and connexin 43) in rats and humans during pregnancy. Am J Obstet Gynecol. 1992;167(2):559‐567. [DOI] [PubMed] [Google Scholar]
  • 122. Miyoshi H, Boyle MB, MacKay LB, Garfield RE. Voltage‐clamp studies of gap junctions between uterine muscle cells during term and preterm labor. Biophys J. 1996;71(3):1324‐1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Garfield RE, Sims S, Daniel EE. Gap junctions: their presence and necessity in myometrium during parturition. Science. 1977;198(4320):958‐960. [DOI] [PubMed] [Google Scholar]
  • 124. Garfield RE, Hayashi RH. Appearance of gap junctions in the myometrium of women during labor. Am J Obstet Gynecol. 1981;140(3):254‐260. [DOI] [PubMed] [Google Scholar]
  • 125. Garfield RE, Daniel EE, Dukes M, Fitzgerald JD. Changes of gap junctions in myometrium of guinea pig at parturition and abortion. Can J Physiol Pharmacol. 1982;60(3):335‐341. [DOI] [PubMed] [Google Scholar]
  • 126. Döring B, Shynlova O, Tsui P, et al. Ablation of connexin43 in uterine smooth muscle cells of the mouse causes delayed parturition. J Cell Sci. 2006;119(Pt 9):1715‐1722. [DOI] [PubMed] [Google Scholar]
  • 127. Arrowsmith S, Wray S. Oxytocin: its mechanism of action and receptor signalling in the myometrium. J Neuroendocrinol. 2014;26(6):356‐369. [DOI] [PubMed] [Google Scholar]
  • 128. Obstetrics ACoPB . Obstetrics ACoPB – ACOG practice bulletin No. 107: induction of labor. Obstet Gynecol. 2009;114(2 Pt 1):386‐397. [DOI] [PubMed] [Google Scholar]
  • 129. Bulletins‐Obstetrics CoP . Practice Bulletin No. 183: postpartum Hemorrhage. Obstet Gynecol. 2017;130(4):e168–e186. [DOI] [PubMed] [Google Scholar]
  • 130. Fuchs AR, Fuchs F, Husslein P, Soloff MS, Fernström MJ. Oxytocin receptors and human parturition: a dual role for oxytocin in the initiation of labor. Science. 1982;215(4538):1396‐1398. [DOI] [PubMed] [Google Scholar]
  • 131. Fuchs AR, Fuchs F, Husslein P, Soloff MS. Oxytocin receptors in the human uterus during pregnancy and parturition. Am J Obstet Gynecol. 1984;150(6):734‐741. [DOI] [PubMed] [Google Scholar]
  • 132. Kubota Y, Kimura T, Hashimoto K, et al. Structure and expression of the mouse oxytocin receptor gene. Mol Cell Endocrinol. 1996;124(1–2):25‐32. [DOI] [PubMed] [Google Scholar]
  • 133. Alexandrova M, Soloff MS. Oxytocin receptors and parturition. I. Control of oxytocin receptor concentration in the rat myometrium at term. Endocrinology. 1980;106(3):730‐735. [DOI] [PubMed] [Google Scholar]
  • 134. Schellenberg JC, Pliska V, Lutz RA. Oxytocin receptors in guinea pig myometrium near term and during labor. Biol Reprod. 2000;62(2):427‐431. [DOI] [PubMed] [Google Scholar]
  • 135. Ferreira JJ, Butler A, Stewart R, et al. Oxytocin can regulate myometrial smooth muscle excitability by inhibiting the Na. J Physiol. 2019;597(1):137‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Kim SH, MacIntyre DA, Firmino Da Silva M, et al. Oxytocin activates NF‐kappaB‐mediated inflammatory pathways in human gestational tissues. Mol Cell Endocrinol. 2015;403:64‐77. [DOI] [PubMed] [Google Scholar]
  • 137. Kim SH, Pohl O, Chollet A, et al. Differential effects of oxytocin receptor antagonists, atosiban and nolasiban, on oxytocin receptor‐mediated signaling in human amnion and myometrium. Mol Pharmacol. 2017;91(4):403‐415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Husslein P, Fuchs AR, Fuchs F. Oxytocin and the initiation of human parturition. I. Prostaglandin release during induction of labor by oxytocin. Am J Obstet Gynecol. 1981;141(6):688‐693. [DOI] [PubMed] [Google Scholar]
  • 139. Jenkin G, Young IR. Mechanisms responsible for parturition; the use of experimental models. Anim Reprod Sci. 2004;82–83:567‐581. [DOI] [PubMed] [Google Scholar]
  • 140. Douglas AJ, Leng G, Russell JA. The importance of oxytocin mechanisms in the control of mouse parturition. Reproduction. 2002;123(4):543‐552. [DOI] [PubMed] [Google Scholar]
  • 141. McCafferty GP, Pullen MA, Wu C, et al. Use of a novel and highly selective oxytocin receptor antagonist to characterize uterine contractions in the rat. Am J Physiol Regul Integr Comp Physiol. 2007;293(1):R299‐R305. [DOI] [PubMed] [Google Scholar]
  • 142. Schellenberg JC. The effect of oxytocin receptor blockade on parturition in guinea pigs. J Clin Invest. 1995;95(1):13‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Flenady V, Reinebrant HE, Liley HG, Tambimuttu EG, Papatsonis DN. Oxytocin receptor antagonists for inhibiting preterm labour. Cochrane Database Syst Rev. 2014;6:CD004452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Takayanagi Y, Yoshida M, Bielsky IF, et al. Pervasive social deficits, but normal parturition, in oxytocin receptor‐deficient mice. Proc Natl Acad Sci USA. 2005;102(44):16096‐16101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. 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. Proc Natl Acad Sci USA. 1996;93(21):11699‐11704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Young WS, Shepard E, Amico J, et al. Deficiency in mouse oxytocin prevents milk ejection, but not fertility or parturition. J Neuroendocrinol. 1996;8(11):847‐853. [DOI] [PubMed] [Google Scholar]
  • 147. Kim J, Stirling KJ, Cooper ME, et al. Sequence variants in oxytocin pathway genes and preterm birth: a candidate gene association study. BMC Med Genet. 2013;14:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Slater DM, Dennes WJ, Campa JS, Poston L, Bennett PR. Expression of cyclo‐oxygenase types‐1 and ‐2 in human myometrium throughout pregnancy. Mol Hum Reprod. 1999;5(9):880‐884. [DOI] [PubMed] [Google Scholar]
  • 149. Hirst JJ, Mijovic JE, Zakar T, Olson DM. Prostaglandin endoperoxide H synthase‐1 and ‐2 mRNA levels and enzyme activity in human decidua at term labor. J Soc Gynecol Investig. 1998;5(1):13‐20. [DOI] [PubMed] [Google Scholar]
  • 150. Olson DM. The role of prostaglandins in the initiation of parturition. Best Pract Res Clin Obstet Gynaecol. 2003;17(5):717‐730. [DOI] [PubMed] [Google Scholar]
  • 151. Brodt‐Eppley J, Myatt L. Changes in expression of contractile FP and relaxatory EP2 receptors in pregnant rat myometrium during late gestation, at labor, and postpartum. Biol Reprod. 1998;59(4):878‐883. [DOI] [PubMed] [Google Scholar]
  • 152. Brodt‐Eppley J, Myatt L. Prostaglandin receptors in lower segment myometrium during gestation and labor. Obstet Gynecol. 1999;93(1):89‐93. [DOI] [PubMed] [Google Scholar]
  • 153. Terry KK, Lebel WS, Riccardi KA, Grasser WA, Thompson DD, Paralkar VM. Effects of gestational age on prostaglandin EP receptor expression and functional involvement during in vitro contraction of the guinea pig uterus. Prostaglandins Leukot Essent Fatty Acids. 2008;78(1):3‐10. [DOI] [PubMed] [Google Scholar]
  • 154. Sugimoto Y, Yamasaki A, Segi E, et al. Failure of parturition in mice lacking the prostaglandin F receptor. Science. 1997;277(5326):681‐683. [DOI] [PubMed] [Google Scholar]
  • 155. Waltman R, Tricomi V, Shabanah EH, Arenas R. The effect of anti‐inflammatory drugs on parturition parameters in the rat. Prostaglandins. 1973;4(1):93‐106. [DOI] [PubMed] [Google Scholar]
  • 156. Chester R, Dukes M, Slater SR, Walpole AL. Delay of parturition in the rat by anti‐inflammatory agents which inhibit the biosynthesis of prostaglandins. Nature. 1972;240(5375):37‐38. [DOI] [PubMed] [Google Scholar]
  • 157. Lee PR, Kim SR, Jung BK, et al. Therapeutic effect of cyclo‐oxygenase inhibitors with different isoform selectivity in lipopolysaccharide‐induced preterm birth in mice. Am J Obstet Gynecol. 2003;189(1):261‐266. [DOI] [PubMed] [Google Scholar]
  • 158. Reinebrant HE, Pileggi‐Castro C, Romero CL, et al. Cyclo‐oxygenase (COX) inhibitors for treating preterm labour. Cochrane Database Syst Rev. 2015;6:CD001992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Haas DM, Imperiale TF, Kirkpatrick PR, Klein RW, Zollinger TW, Golichowski AM. Tocolytic therapy: a meta‐analysis and decision analysis. Obstet Gynecol. 2009;113(3):585‐594. [DOI] [PubMed] [Google Scholar]
  • 160. Haas DM, Caldwell DM, Kirkpatrick P, McIntosh JJ, Welton NJ. Tocolytic therapy for preterm delivery: systematic review and network meta‐analysis. BMJ. 2012;345:e6226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Simhan HN, Caritis S. Inhibition of acute preterm labor. In: Post TW ed. UpToDate. Waltham, MA: UpToDate; 2020. [Google Scholar]
  • 162. Hahn DW, McGuire JL, Vanderhoof M, Ericson E, Pasquale SA. Evaluation of drugs for arrest of premature labor in a new animal model. Am J Obstet Gynecol. 1984;148(6):775‐778. [DOI] [PubMed] [Google Scholar]
  • 163. Abel MH, Hollingsworth M. Comparison of nifedipine and diltiazem with salbutamol for prevention of preterm delivery in the ovariectomized, oestrogen‐treated late pregnant rat. J Reprod Fertil. 1986;77(2):559‐568. [DOI] [PubMed] [Google Scholar]
  • 164. Coleman HA, Parkington HC. Induction of prolonged excitability in myometrium of pregnant guinea‐pigs by prostaglandin F2 alpha. J Physiol. 1988;399:33‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Kantas E, Cetin A, Kaya T, Cetin M. Effect of magnesium sulfate, isradipine, and ritodrine on contractions of myometrium: pregnant human and rat. Acta Obstet Gynecol Scand. 2002;81(9):825‐830. [DOI] [PubMed] [Google Scholar]
  • 166. Melin P, Trojnar J, Johansson B, Vilhardt H, Akerlund M. Synthetic antagonists of the myometrial response to vasopressin and oxytocin. J Endocrinol. 1986;111(1):125‐131. [DOI] [PubMed] [Google Scholar]
  • 167. Hahn DW, Demarest KT, Ericson E, Homm RE, Capetola RJ, McGuire JL. Evaluation of 1‐deamino‐[D‐Tyr(Oethyl)2, Thr4, Orn8] vasotocin, an oxytocin antagonist, in animal models of uterine contractility and preterm labor: a new tocolytic agent. Am J Obstet Gynecol. 1987;157(4 Pt 1):977‐982. [DOI] [PubMed] [Google Scholar]
  • 168. Hajagos‐Tóth J, Falkay G, Gáspár R. Modification of the effect of nifedipine in the pregnant rat myometrium: the influence of progesterone and terbutaline. Life Sci. 2009;85(15–16):568‐572. [DOI] [PubMed] [Google Scholar]
  • 169. Coleman HA, Hart JD, Tonta MA, Parkington HC. Changes in the mechanisms involved in uterine contractions during pregnancy in guinea‐pigs. J Physiol. 2000;523(Pt 3):785‐798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Moynihan AT, Smith TJ, Morrison JJ. The relaxant effect of nifedipine in human uterine smooth muscle and the BK(Ca) channel. Am J Obstet Gynecol. 2008;198(2):237.e1–237.e8. [DOI] [PubMed] [Google Scholar]
  • 171. Ballejo G, Calixto JB, Medeiros YS. In vitro effects of calcium entry blockers, chlorpromazine and fenoterol upon human pregnant myometrium contractility. Br J Pharmacol. 1986;89(3):515‐523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Osaghae B, Arrowsmith S, Wray S.The effect of dual combination of Magnesium sulphate with atosiban and indomethacin on myometrial contractions of pregnant mouse. Physiology; 2016; Dublin, Ireland. [Google Scholar]
  • 173. Doret M, Mellier G, Benchaib M, Piacenza JM, Gharib C, Pasquier JC. In vitro study of tocolytic effect of rofecoxib, a specific cyclo‐oxygenase 2 inhibitor. comparison and combination with other tocolytic agents. BJOG. 2002;109(9):983‐988. [DOI] [PubMed] [Google Scholar]
  • 174. Sawdy R, Knock GA, Bennett PR, Poston L, Aaronson PI. Effect of nimesulide and indomethacin on contractility and the Ca2+ channel current in myometrial smooth muscle from pregnant women. Br J Pharmacol. 1998;125(6):1212‐1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Bafor EE, Rowan EG, Edrada‐Ebel R. Toward understanding myometrial regulation: metabolomic investigation reveals new pathways of oxytocin and ritodrine activity on the myometrium. Reprod Sci. 2017;24(5):691‐705. [DOI] [PubMed] [Google Scholar]
  • 176. Carvajal JA, Zambrano MJ, Theodor NM, et al. The synergic in vitro tocolytic effect of nifedipine plus ritodrine on human myometrial contractility. Reprod Sci. 2017;24(4):635‐640. [DOI] [PubMed] [Google Scholar]
  • 177. Demarest KT, Hahn DW, Ericson E, Capetola RJ, Fuchs AR, McGuire JL. Profile of an oxytocin antagonist, RWJ 22164 for treatment of preterm labor in laboratory models of uterine contractility. Am J Perinatol. 1989;6(2):200‐204. [DOI] [PubMed] [Google Scholar]

Articles from Acta Physiologica (Oxford, England) are provided here courtesy of Wiley

RESOURCES