Skip to main content
Physiological Reviews logoLink to Physiological Reviews
. 2024 Feb 8;104(3):1121–1145. doi: 10.1152/physrev.00019.2023

The hormonal control of parturition

Emily Hamburg-Shields 1,2, Sam Mesiano 1,2,
PMCID: PMC11380996  PMID: 38329421

graphic file with name prv-00019-2023r01.jpg

Keywords: hormonal, human, parturition, pregnancy

Abstract

Parturition is a complex physiological process that must occur in a reliable manner and at an appropriate gestation stage to ensure a healthy newborn and mother. To this end, hormones that affect the function of the gravid uterus, especially progesterone (P4), 17β-estradiol (E2), oxytocin (OT), and prostaglandins (PGs), play pivotal roles. P4 via the nuclear P4 receptor (PR) promotes uterine quiescence and for most of pregnancy exerts a dominant block to labor. Loss of the P4 block to parturition in association with a gain in prolabor actions of E2 are key transitions in the hormonal cascade leading to parturition. P4 withdrawal can occur through various mechanisms depending on species and physiological context. Parturition in most species involves inflammation within the uterine tissues and especially at the maternal-fetal interface. Local PGs and other inflammatory mediators may initiate parturition by inducing P4 withdrawal. Withdrawal of the P4 block is coordinated with increased E2 actions to enhance uterotonic signals mediated by OT and PGs to promote uterine contractions, cervix softening, and membrane rupture, i.e., labor. This review examines recent advances in research to understand the hormonal control of parturition, with focus on the roles of P4, E2, PGs, OT, inflammatory cytokines, and placental peptide hormones together with evolutionary biology of and implications for clinical management of human parturition.


CLINICAL HIGHLIGHTS.

Parturition is controlled by hormones acting in an endocrine and/or paracrine manner to either maintain pregnancy (e.g., progesterone) or transform the uterine myometrium and cervix into the labor state (e.g., inflammatory and stress-associated signals). Understanding of the cell and molecular biology underlying the action and regulation of specific progestation and prolabor hormones involved in human parturition is revealing novel therapeutic strategies (e.g., boosting the progesterone block and/or preventing its withdrawal) to optimize the clinical control of birth timing (e.g., prevent pre- and postterm birth).

1. INTRODUCTION

Birth of a viable offspring through the process of parturition is a defining event for reproduction in viviparous species. Parturition is fundamentally a function of uterine activity. As the site for internal gestation, also a defining trait of viviparity, the uterus accommodates the conceptus (products of conception): the developing embryo/fetus with its placenta, extraembryonic membrane, and amniotic fluid. For most of pregnancy the muscular uterine wall, the myometrium, is composed of hypertrophied myometrial smooth muscle cells that are generally relaxed, producing minimal tone with only sporadic and weak asynchronous contractions. The myometrium is also compliant, distending to accommodate the increasing volume of the developing conceptus. At the same time, the uterine outlet, the cervix, is composed of stroma with a dense extracellular matrix (ECM) and secretory endocervical mucosa that forms a rigid and closed barrier to contain the conceptus. The inner endometrial lining of the uterus, referred to as the decidua, interfaces directly with trophoblast cells of the conceptus and establishes a local microenvironment that is immunologically quiescent to tolerate the allogeneic fetal cells.

At parturition, the myometrium transforms to become the engine for birth. Myometrial cells become electrically coupled and produce forceful, synchronized, and rhythmic contractions that force the conceptus against the cervical outlet. At the same time, collagen fibrils in the cervix ECM remodel to soften the stroma, allowing the uterine outlet to dilate in response to increased intrauterine pressure produced by myometrial contractions. In the decidua, immune/inflammatory quiescence transforms to tissue-level inflammation with the infiltration of activated maternal immune cells that produce factors, especially prostaglandins (PGs), that weaken the adjacent amnion membrane, leading to its rupture, and induce myometrium contraction and softening of cervical stroma. Myometrial contractions coupled with cervix dilation and membrane rupture eventually force the fetus, and then the placenta with the attached amnion-chorion membrane and decidua membrane, through the cervix, bony pelvic outlet, and vaginal canal. These uterine tissue-specific paracrine and endocrine events are reinforced by the neuroendocrine release of oxytocin (OT), which further propagates uterine contractions to facilitate labor and delivery (for review see Refs. 1, 2) (FIGURE 1).

FIGURE 1.

FIGURE 1.

Endocrine, paracrine, and neuroendocrine interactions controlling parturition. Multiple physiological factors (some examples shown) contribute signals impacting uterine tissues that are integrated into a putative inflammatory load. In a threshold-limited manner, inflammatory load induces progesterone (P4)/P4 receptor (PR) withdrawal leading to loss of the P4/PR block to labor, which includes increased local inflammation due to loss of P4/PR anti-inflammatory activity, leading to increased local production of inflammatory cytokines, some of which [e.g., prostaglandin (PG)F] are potent uterotonins. P4/PR withdrawal also increases expression of ESR1 [encodes estrogen receptor-α (ERα)], allowing circulating estrogens (mainly estradiol) to increase response of uterine tissues to uterotonins [especially PGF and oxytocin (OT)] and increase extracellular matrix (ECM) breakdown in the cervix and fetal membrane, leading to cervix softening and membrane weakening, respectively. A neuroendocrine positive feedback loop involving OT amplifies contractions at the time of active labor: phasic myometrium contractions, cervix softening and dilation, and membrane rupture.

Transformation of the gravid uterus from quiescence to active labor is controlled primarily by hormonal interactions involving two classes of hormones, uterotonins, typified by PGs and OT, that directly affect myometrium contraction and uterotropins, primarily the steroid hormones progesterone (P4) and estradiol (E2), that promote uterine growth and produce a phenotype conducive to pregnancy maintenance or parturition (FIGURE 2). Here we review recent advances in the understanding of how these hormones control the process and timing of parturition.

FIGURE 2.

FIGURE 2.

The majority of pregnancy is a quiescent state, with low myometrial contractile activity, maintained by progesterone (P4), which is the dominant hormonal effector of ongoing gestation. The progression from quiescent pregnancy to latent (early) labor is facilitated by a functional progesterone withdrawal and increased estradiol (E2) effect. Uterotonins [prostaglandins (PGs) and oxytocin (OT)] are the predominant effectors of active labor and postpartum uterine involution. Image is an adaptation of material from Ref. 3.

2. PROGESTERONE

The hormonal control of pregnancy and parturition is dominated by P4. In all species, P4 supplied by luteinized granulosa cells in the ovarian corpus luteum (CL) modifies the endometrium to create a site conducive to establishing a uterine pregnancy. In some species (e.g., mouse, goat), the maternal CL persists throughout pregnancy and is the sole source of P4 to maintain pregnancy. In other species (e.g., sheep, human), the placenta also produces P4 (4, 5). In women, the placenta is the principal source of P4 during the second and third trimesters (4, 6, 7).

In all species, actions of P4 to maintain pregnancy are mediated by the nuclear P4 receptor (PR) isoforms, PR-A and PR-B, that function primarily as ligand-activated transcription factors. PR-A and PR-B share amino acid sequence except that PR-B has an extra 164 NH2-terminal residues (8). Both isoforms are detectable by immunostaining in myometrial, cervical, and decidual cells in the gravid uterus of all species studied so far, and in each cell type the PRs localize exclusively to the nucleus, consistent with their function as ligand-activated transcription factors. The importance of P4/PR signaling for the maintenance of pregnancy is demonstrated by numerous clinical and animal studies showing that disruption of P4/PR signaling with PR antagonists, such as RU486 (aka mifepristone) and onapristone, induces labor and the full parturition cascade at all stages of pregnancy (9, 10).

In the 1950s Arpad Csapo (11) proposed the “progesterone block” hypothesis, which posits that for most of pregnancy P4 actively blocks labor. Consistent with the genomic mode of steroid hormone/nuclear receptor action, P4/PR is thought to maintain pregnancy and block parturition by affecting expression of specific genes in uterine cells during pregnancy (12). P4/PR is considered to affect expression of genes encoding factors collectively referred to as contraction-associated proteins (CAPs). Studies in vitro and in vivo (mainly in mice and rats) suggest that P4/PR inhibits expression of the genes that encode procontraction CAPs (e.g., GJA1 that encodes connexin-43, which electrically couples myometrial cells to synchronize contractions; PTGS2 that encodes the rate-limiting enzyme for the synthesis of bioactive PGs that promote myometrial cell contraction; OXTR that encodes the OT receptor to increase responsiveness to OT) and increases expression of genes encoding prorelaxation factors (e.g., adrenergic receptor subtypes that mediate relaxation in response to adrenaline) (1315). P4/PR also decreases responsiveness to E2 in uterine cells by inhibiting expression of ESR1 that encodes estrogen receptor-α (ERα) (16, 17). In myometrial cells E2/ERα increases expression of prolabor CAP genes and in cervix stromal cells expression of genes encoding metalloproteinases that break down collagen, leading to softening and dilation of the cervix (1822). These effects are consistent with the “seesaw hypothesis” proposed by Arpad Csapo in the 1970s, which posits that the labor state of the gravid uterus is controlled by the balance between the P4 block and prolabor stimuli, especially E2, OT, and PGs (23). For most of pregnancy the P4/PR block prevails.

Studies in transgenic mice have revealed specific roles of PR-A and PR-B in murine pregnancy and parturition. Mice lacking both PRs (2427) develop normally to adulthood; however, female mice are infertile because of neuroendocrine disorders and multiple defects in the reproductive tissues including endometrial hyperplasia and inflammation, defective mammary gland development, and anovulation. Female mice lacking only PR-A (i.e., having only PR-B) are also infertile, with a reproductive phenotype similar to that of the mice lacking both PRs. However, PR-B mediates a normal response to P4 in the mammary epithelium in the PR-A-null mice, suggesting that PR-B is sufficient for mammary development and lactation whereas PR-A is critical for uterine function and neuroendocrine control of reproduction (28, 29). In contrast, mice lacking only PR-B (i.e., having only PR-A) have normal pregnancy and parturition but defective mammary gland morphogenesis and lactation (28). The data suggest that PR-A alone is sufficient to mediate P4 actions to establish and maintain murine pregnancy.

With regard to cell type-specific roles for P4/PR in pregnancy maintenance, recent studies suggest that P4/PR signaling in myometrial cells is not necessary for pregnancy and parturition (30). Mice lacking PR expression in smooth muscle cells, which included uterine myometrial cells, had markedly decreased fertility, primarily due to failure of embryos to transit from the oviducts to the uterine lumen. Even though myometrial cell PR deletion impaired adaptation of the myometrium to pregnancy, in the few pregnancies established by embryos that managed to enter the uterus, implantation, pregnancy, and parturition were normal. This suggests that, at least in mice, P4/PR signaling in myometrial cells is not necessary for pregnancy establishment and maintenance. Interestingly, in decidual stromal cells P4/PR signaling was not affected by myometrial cell PR deletion, suggesting that the decidua in mice is the key uterine tissue mediating the P4/PR actions necessary for pregnancy. Taken together, data from PR isoform ablation experiments in mice provide novel insights into the cell/tissue-specific roles of PR-A and PR-B in pregnancy and parturition, with PR-A appearing to play a major role. Whether this applies to pregnancy in other species, especially human, is unclear.

Siiteri and colleagues (31, 32) in the 1970s proposed that P4 maintains pregnancy by exerting anti-inflammatory actions. It is now generally accepted, on the basis of clinical and animal data, that parturition is an inflammatory event associated with increased expression in uterine tissue of genes encoding proinflammatory cytokines and activation and increased infiltration of neutrophils and macrophages into each of the uterine compartments (3339). Animal and clinical studies also show that parturition is induced by inflammatory stimuli such as intrauterine infection and chorioamnionitis (4045). Multiple lines of evidence suggest that P4/PR blocks labor, at least in part, by inhibiting the transcriptional response of uterine cells and tissues to prolabor/proinflammatory stimuli (27, 4653). Thus, data in the last 20–30 years justify updating Csapo’s seesaw model for the hormonal control of parturition to include inflammation and proinflammatory hormones as prolabor factors that are counterbalanced by anti-inflammatory actions of P4/PR.

The NF-κB transcription factor complex is a key mediator of inflammatory stimuli and is activated in the uterine tissues in association with the onset of labor (5456). Several lines of evidence in a variety of cell types indicate that P4/PR exerts anti-inflammatory activity by inhibiting NF-κB. In kidney fibroblast cells P4/PR inhibits NF-κB activity by physically interacting (albeit weakly) with the P65 subunit of NF-κB (57). In myometrial cells, P4/PR increases expression and decreases the degradation of IκBα that inhibits NF-κB by binding to and sequestering the NF-κB complex to the cytoplasm (47, 58) and by inducing expression of dual-specificity phosphatase 1, which decreases NF-κB translocation to the nucleus (57, 58). Some studies suggest that P4/PR indirectly decreases NF-κB activity by affecting other transcription factors and/or coregulators (5961). Studies of DNA promoter occupancy showed that in human myometrial cells P4/PR-B decreases interleukin (IL)-1β-induced interaction of NF-κB with cognate response elements in the PTGS2 promoter (47). Thus, the anti-inflammatory actions of P4/PR appear to be promoter specific. Recent transcriptome analyses in myometrial cells suggest that P4/PR inhibition of NF-κB involves the activator protein-1 (AP-1) transcription factor complex. In a human myometrial cell line and in human term myometrium, P4/PR anti-inflammatory activity does not affect all IL-1β/NF-κB-responsive genes, but instead promoter motif analysis shows that genes whose promoters contain response elements to both NF-κB and AP-1 were enriched in the cohort of IL-1β-responsive genes inhibited by P4/PR (51). Studies in other cell types show that AP-1 and NF-κB function in synergy to mediate gene transcription in response to proinflammatory stimuli (62, 63). In myometrial cells, AP-1 is thought to be a master transcription factor for the expression of genes encoding key CAPs, especially GJAI (64, 65). Importantly, PR-A and PR-B each physically interact with AP-1 subunits in myometrial cells in an isoform- and ligand-dependent manner (61). PR-A interacts with the FOS subunits, whereas PR-B has preference for JUN homodimers. PR-A-FOS in a ligand-independent manner induces expression of GJA1, whereas P4/PR-B-JUN inhibits GJA1 expression. According to this model, for most of pregnancy P4/PR-B via interaction with JUN blocks parturition by inhibiting CAP gene expression. Whether this applies to the expression of NF-κB-dependent inflammatory genes is not known.

P4/PR may promote uterine quiescence indirectly by modulating expression of specific transcription factors that affect the expression of CAP genes. This mechanism is exemplified by the observation that in myometrial cells P4/PR increases expression of the zinc finger E-box-binding transcriptional repressor ZEB1 that decreases expression of key contractile CAP genes (66). In addition, P4/PR may increase expression, either directly or via ZEB1, of specific miRNAs, especially miR-199a-3p and miR-214, that repress CAP and inflammatory gene expression (67). At parturition, P4/PR withdrawal (discussed below) would decrease ZEB1, leading to increased miR-200 that further decreases ZEB1 and ZEB2 to remove ZEB inhibition of CAP genes. Loss of P4/PR would also decrease levels of the anti-inflammatory miR-199a-3p and miR-214. This regulatory network may be critical in establishing the labor state of the myometrium.

Regulation of the inflammatory state in the gravid uterus by P4/PR is not limited to myometrium. In decidual stromal cells P4/PR induces expression of IL15, which is thought to promote the differentiation of resident uterine natural killer cells to produce an immune quiescent cellular microenvironment that is tolerant of the allogeneic conceptus (6870). In decidual stromal cells IL-1β inhibits expression of IL-15, and this is prevented by P4/PR (70).

Thus, taken together current data demonstrate that P4/PR blocks labor via multiple pathways leading to the inhibition of CAP gene expression and the prevention of tissue-level inflammation (FIGURE 3).

FIGURE 3.

FIGURE 3.

Signaling pathways by which progesterone (P4)/P4 receptor (PR) blocks labor. P4/PR via decidual cell expression of IL-15 promotes immune quiescence at the maternal-fetal interface. P4/PR directly inhibits expression of ESR1, which decreases levels of estrogen receptor-α (ERα), leading to refractoriness to estradiol (E2)/ERα-induced expression of contraction-associated protein (CAP) genes. P4/PR increases expression of ZEB1, whose downstream effects inhibit CAP and inflammatory cytokine gene expression. P4/PR interacts physically with the activator protein-1 (AP-1) transcription factor complex to inhibit activity of AP-1 and NF-κB, leading to a broad anti-inflammatory effect and inhibition of AP-1-driven CAP genes. For most of pregnancy P4/PR effects (green) dominate. STAT5b, signal transducer and activator of transcription 5b; 20αHSD, 20α-hydroxysteroid dehydrogenase.

3. PROGESTERONE WITHDRAWAL

Just as the P4 block to labor dominates the hormonal control of pregnancy maintenance, P4 withdrawal is a major trigger for parturition. Loss of the P4/PR block alone is sufficient to induce the full parturition cascade; and this appears to be a conserved trait among viviparous species as evidenced by the induction of labor by RU486 in essentially all viviparous studied to date. This implies that the uterus would be poised for parturition at all stages of pregnancy were it not for the P4/PR block. In this system the mechanism for P4/PR withdrawal and how it is induced is central to the hormonal regulation of parturition.

P4 withdrawal can occur systemically, locally, or functionally (FIGURE 4). Although the mechanism of P4 withdrawal that triggers term parturition in a particular species is considered that species’ primary mechanism for the hormonal control of parturition, it is now clear that the various P4 withdrawal modalities may coexist and function synergistically or in response to specific physiological states. For example, although systemic P4 withdrawal triggers parturition in mice, a local and/or functional P4 withdrawal appears to operate in response to parturition induced by inflammation (71).

FIGURE 4.

FIGURE 4.

Modes of progesterone (P4) withdrawal. Systemic P4 withdrawal occurs by P4 metabolism in the source tissue [e.g., corpus luteum (CL) via 20α-hydroxysteroid dehydrogenase (20αHSD) or placenta via P450c17]. Similarly, local withdrawal occurs by P4 metabolism by 20αHSD in uterine target cells to prevent P4 from interacting with P4 receptors (PRs). Functional P4 withdrawal occurs by changes in PR transcriptional activity caused by interaction with specific repressors, transrepression by phosphorylated PR-A, and/or direct activation of contraction-associated protein (CAP) gene expression by liganded PR-A. A4, androstenedione; ERα, estrogen receptor-α; 20αOHP, 20α-hydroxyprogesterone.

3.1. Systemic P4 Withdrawal

Systemic P4 withdrawal is caused by decreased production of P4 from the CL or placenta. In both tissues reduced P4 release is caused by increased P4 metabolism. In mice and rats parturition is preceded by increased activity in CL cells of the 20α-hydroxysteroid dehydrogenase (20αHSD) enzyme, a member of the aldoketo reductase family that converts P4 to 20α-hydroxyprogesterone (20αOHP) which lacks progestin activity (7274). Loss of P4 removes negative feedback on the hypothalamus-pituitary, which increases follicle-stimulating hormone that increases E2 production by developing follicles, which leads to the postpartum estrus typical of rodents. In sheep increased expression in placental trophoblast cells of Cyp17a1 that encodes the 17α-hydroxylase, 17/20 lyase (P450c17) enzyme, decreases P4 release by converting it to androstenedione (A4) (75). A4 is then converted to E2 in placental cells, leading to a coordinated increase in maternal E2 levels. Thus, induction of 20αHSD in the CL (mice) and P450c17 in the placenta (sheep) each cause systemic P4 withdrawal coupled with an increase in E2. The net effect of P4 withdrawal and increased E2 drives the uterine tissues to the labor state.

In human pregnancy, P4 is supplied initially by the CL and subsequently (during the later two-thirds of pregnancy) by the placenta. Unlike the sheep, human placental cells do not express CYP17A1 and continue to produce P4 before and during parturition, with maternal P4 levels decreasing only after delivery of the placenta. Thus, human parturition occurs without systemic P4 withdrawal.

3.2. Local P4 Withdrawal

Although parturition in most species is preceded by a systemic P4 withdrawal, the level of circulating total P4 is not completely abolished, and in some species such as mice the levels are within the range that establishes pregnancy (76). This suggests the existence of other P4 withdrawal mechanisms. One of those mechanisms is local P4 withdrawal mediated by P4 metabolism in PR-expressing target cells.

The core principle of local P4 withdrawal is that P4 entering target cells is prevented from interacting with PRs by conversion to a PR-inactive form. This may be mediated by the 20αHSD enzyme that also functions in CL cells to decrease the release of P4 (61, 71, 77). In mice, systemic P4 withdrawal is caused by increased 20αHSD in CL cells, and this is induced by PGF (78, 79). Although parturition in mice lacking 20αHSD is generally delayed, because of persistent CL P4 production, in some 20αHSD-deficient mice parturition was prolonged even though circulating P4 levels declined, suggesting that systemic P4 withdrawal is not sufficient to precipitate parturition (80, 81). Local P4 withdrawal via 20αHSD-mediated P4 metabolism in the uterine tissues may be a mechanism to compensate for insufficient systemic P4 withdrawal. Thus, 20αHSD acts as a gatekeeper to metabolize P4 to 20αOHP before it accesses the PRs. The 5α-reductase enzymes also may contribute to P4 metabolism in cervical stromal cells (82). Recent studies show that expression of AKR1C1, the gene that encodes 20αHSD, increases in human term myometrium in association with the onset of labor (83). This finding is consistent with earlier studies showing that in term myometrium the level of 20αOHP relative to P4 increases with labor (84, 85). These data support the hypothesis that 20αHSD-mediated local P4 withdrawal occurs in the human parturition process. Thus, local P4 metabolism in conjunction with a decline in circulating P4 levels and possibly altered PR activity (see below) represents redundancy to ensure a P4/PR withdrawal that is sufficient to initiate parturition.

The regulation of AKR1C1 expression leading to altered 20αHSD activity is a critical element in the hormonal control of parturition. Interestingly, this is linked to the P4/PR-ZEB1 signaling network. Expression of AKR1C1 is inhibited by the signal transducer and activator of transcription (STAT)5b transcription factor (81, 86). STAT5b-deficient mice have increased CL 20αHSD activity leading to lower than normal circulating P4 levels and increased incidence of midgestation pregnancy loss (81). Interestingly, the STAT5B promoter is a target for repression by miR-200 (77), whose levels are inhibited by P4/PR via ZEB1 (see above). Thus, a plausible model is that for most of pregnancy the P4/PR-ZEB1 interaction suppresses miR-200, allowing expression of STAT5B, leading to production of STAT5b that then suppresses AKR1C1, leading to low levels of 20αHSD allowing P4 to interact with PRs in target cells, leading to sustained suppression of AKR1C1. At parturition, loss of P4/PR removes the ZEB1-mediated inhibition of miR-200, leading to miR-200-mediated inhibition of STAT5B and loss of STAT5b repression of AKR1C1, leading to increased 20αHSD that prevents P4 from accessing PRs. This regulatory network contains positive feedback loops such that a small decrease in P4/PR activity is amplified to cause a significant local loss of P4 via 20αHSD metabolism. This local reduction in P4 increases miR-200-mediated repression of ZEB1 and ZEB2 that releases ZEB-mediated inhibition of CAP gene expression and response to inflammatory stimuli to transform the uterus to the labor state.

3.3. Functional P4 Withdrawal

Functional P4 withdrawal may occur by several mechanisms, each involving modulation of PR transcriptional activity. In response to P4 binding PR-A and PR-B each affect the expression of specific and overlapping gene sets in a cell type-specific manner, with the full-length PR-B having more robust P4-induced transcriptional activity than PR-A. In most cell types examined so far, including human myometrial cells, PR-A decreases P4/PR-B transcriptional activity in a concentration (based on immunoblot assay)-dependent manner (59, 87, 88). This transrepressive action of PR-A is thought to be a mechanism to regulate response to P4 especially when PR-A exceeds PR-B. This effect led to the PR-A/PR-B hypothesis for functional P4 withdrawal, which posits that an increase in the PR-A-to-PR-B ratio in uterine cells causes functional loss of the P4/PR block to labor. The hypothesis is supported by studies in myometrial cell lines, in term myometrium tissue, and in mice genetically modified to alter the PR-A-to-PR-B ratio in uterine cells (49, 50, 52, 8993). Taken together the data suggest that the P4/PR block to labor is mediated by PR-B and that this is antagonized by increased abundance and transrepressive activity of PR-A. Some studies suggest that functional P4/PR withdrawal is also mediated by inhibition of PR-B transcriptional activity via decreased levels and activity of critical PR transcriptional coactivators (94, 95) and by reduced interaction of PR-B with DNA elements in uterine cells (96).

A core tenet of the PR-A/PR-B hypothesis is increased abundance and transrepressive activity of PR-A in uterine cells. Recent studies of epigenetic marks in the distal PGR promoter (leads to full-length mRNA encoding PR-B) and the proximal PGR promoter (leads to a truncated mRNA encoding PR-A) show that term human myometrium levels of histone modifications are higher in the proximal PR-A promoter compared with the distal PR-B promoter (89, 97, 98). This may explain why PR-A levels increase in myometrium with advancing gestation and in association with term labor whereas PR-B levels remain relatively constant (90).

PR-A abundance and activity may also be affected by posttranslational modifications. The PR isoforms can be phosphorylated at multiple serine residues depending on cell type and physiological context that affect PR stability and transcriptional activity (99). Studies in endometrial and mammary cancer cells show that that IL-1β via the p38 MAPK-induced pSer increases the stability of PR-A (100). Likewise, in human myometrial cells IL-1β increases the transrepressive activity and stability of PR-A (49, 50). These findings suggest that inflammatory stimuli induce PR-A-mediated functional P4/PR withdrawal in myometrial cells. This concept was expanded by the finding that phosphorylation of PR-A at the serine-344/345 locus (pSer344/345-PRA) is induced by IL-1β, is necessary for PR-A transrepression of PR-B-mediated anti-inflammatory actions, and occurs in human term myometrium in association with the onset of labor (46, 49, 50).

PR-A may also promote parturition by directly increasing expression of CAP genes in myometrial cells. Studies of local P4 withdrawal suggest that P4 withdrawal via local 20αHSD-mediated metabolism produces unliganded PR-A that directly induces expression of genes encoding stimulatory CAPs (101). Loss of P4 leading to unliganded PR-A shifts the balance from P4/PR-B/JUN to PR-A/FOS activity, leading to increased CAP gene expression and unfettered response to proinflammatory stimuli producing a prolabor transcriptome (61, 65). Thus, as with local P4 withdrawal, functional P4/PR withdrawal appears to be redundant, involving various mechanisms including site-specific serine phosphorylation of PR-A, ligand-independent transcriptional activity of PR-A, and/or reduced direct transcriptional activity of PR-B.

3.4. Hormonal Control of Progesterone Withdrawal

A key implication of the P4/PR block hypothesis is that withdrawal of the block is a central event in the hormonal control of parturition and that this is induced by the convergence of endocrine and/or paracrine signals depending on physiological context (FIGURE 5). This concept is supported by studies in mice and sheep, in which the hormonal control of parturition is relatively well characterized.

FIGURE 5.

FIGURE 5.

Comparison of endocrine and paracrine pathways to progesterone (P4)/P4 receptor (PR) withdrawal in human (solid line), ovine (dashed line), and murine (dotted line) pregnancy. A comparative approach highlights the unique roles of the murine corpus luteum and ovine placental P4 metabolism in P4/PR withdrawal versus the suspected mechanism of functional P4 withdrawal in human myometrium/decidua. CRH, corticotropin-releasing hormone; DHEA, dehydroepiandrosterone; E2, estradiol; PGF, prostaglandin F. Image is an adaptation of material from Ref. 2.

In mice, PGF produced by epithelial cells of the maternal endometrium increases 20αHSD in CL cells, leading to a decrease in P4 release and a decline in systemic P4 (79, 102104). Loss of P4 removes negative feedback to the maternal hypothalamus-pituitary, leading to increased production of follicle-stimulating hormone and increased E2, thus synchronizing P4 withdrawal and estrogen activation. The control of PGF production by endometrial cells in mouse pregnancy is not clearly defined but is thought to involve some form of gestation clock mechanism (105).

In sheep, P4 withdrawal leading to term parturition is triggered by increased activity of the fetal hypothalamic-pituitary-adrenal (HPA) axis leading to a surge in cortisol level. Cortisol increases expression in ovine trophoblast cells of Cyp17a1 that encodes P450c17, which converts P4 to A4. The diversion of P4 from release to metabolism decreases systemic P4 levels in the ewe, leading to parturition (75). Ovine parturition does not occur if the fetal adrenal cortisol surge is prevented (e.g., by fetal adrenalectomy or fetal hypophysectomy) and can be induced prematurely by administration of glucocorticoid to the fetus (106). Importantly, A4 produced from P450c17-mediated metabolism of P4 is converted by sheep placental cells to E2, leading to a coordinated increase of E2 that acts on the uterine cells to promote labor. Thus, as with the mouse, in sheep parturition the systemic P4 withdrawal induced by specific enzymatic activity is coordinated with systemic estrogen activation.

Examination of the mechanism that increases activity of the fetal sheep HPA axis has shown that this is due to increased PGE2 released by the placenta that sensitizes the fetal adrenal cortex to adrenocorticotropin (ACTH) (107). Thus, the ovine placenta, and specifically its production of PGE2, is a trigger for parturition that via fetal adrenal cortisol induces systemic P4 withdrawal.

In contrast to the sheep, human parturition is not controlled by activity of the fetal HPA axis. Even in pregnancies in which the fetal HPA axis fails to produce cortisol (i.e., congenital adrenal hyperplasia and anencephaly), parturition occurs normally at term (108, 109). Moreover, administration of synthetic glucocorticoid to women presenting with threatened preterm labor does not appear to augment parturition or increase the incidence of preterm birth. Nonetheless, fetal organ system maturation is promoted by glucocorticoid. Also in contrast to the sheep, the human placenta lacks CYP17A1 and as such P4 production does not decline at any stage of pregnancy. Only after delivery of the placenta does systemic maternal P4 decline (110) (FIGURE 6).

FIGURE 6.

FIGURE 6.

Comparison of relative serum levels of progesterone (P4) and estradiol (E2) throughout gestation in human, sheep, and mouse. The level of P4 drops precipitously immediately before parturition in sheep and mouse but in human remains persistently elevated until after parturition. A functional withdrawal of P4/P4 receptor (PR), therefore, is not reflected in serum hormone levels in the context of human pregnancy. Image is an adaptation of material from Ref. 2.

Functional and/or local P4 withdrawal to initiate human parturition may be triggered by multiple redundant mechanisms, including a response to inflammatory stress. Studies in murine and nonhuman primate models show that parturition is induced by proinflammatory stimuli administered to the mother or fetus (38, 4143, 111, 112). In humans, PG production by the fetal membranes and PG levels in amniotic fluid increase before active labor (113117) and administration of PGs at any time in pregnancy can initiate parturition (118, 119). Taken together, the data suggest that inflammatory cytokines including locally produced uterotonic PGs, especially PGF, are important hormonal triggers of parturition. In human myometrial cell lines, PGF increased mRNA encoding PR-A (120) and IL-1β increased PR-A stability and transrepressive activity (50). This suggests, as described above, that inflammation induces PR-A-mediated functional P4 withdrawal (121).

4. INFLAMMATION

Clinical studies show that human parturition is affected by intrinsic physiological and extrinsic pathological inflammatory stressors. Normal term parturition is associated with tissue-level inflammation in the myometrium and chorion-decidua interface (37, 39), and risk for preterm labor leading to preterm birth is elevated in pregnancy affected by intrauterine infection, clinically silent upper genital tract infection, and bacterial vaginosis (122125). Contributors to inflammatory triggers of parturition could include intrinsic physiology related to inflammatory signals from the fetus related to normal fetal development, distension of the uterus as the conceptus grows (126130), and senescence of placental trophoblast cells (105, 131136). Extrinsic factors could include maternal signals derived from environmental and psychosocial stressors (137) mediated by placental corticotropin-releasing hormone (CRH; discussed below) (138), intrauterine infection (139), and the maternal microbiome (140, 141). Recent studies in mice suggest that parturition is triggered by uterus-intrinsic pathways mediated by IL-33 produced by uterine interstitial fibroblasts and resident innate lymphoid cells, and that this mechanism for parturition initiation occurs without systemic P4 withdrawal, demonstrating a role for the immune system in the control of parturition (142).

In mice, fetal lung maturation is coordinated with parturition via pulmonary surfactant protein-A (SP-A), a major component of lung function necessary for air breathing. Intra-amniotic administration of SP-A to mice at midgestation induces labor via the infiltration of fetal macrophages into the maternal myometrium and activation of myometrial cell IL-1β and NF-κB. In contrast, inhibition of SP-A by administration of a blocking antibody delays normal parturition (143). Whether this system links fetal lung maturation with inflammation in the myometrium or intrauterine tissues in other species is not known. In humans, fetal macrophages have not been detected in the myometrium, and fetal SP-A levels are elevated several weeks before the onset of term labor and do not correlate in time with parturition (144).

Recent studies of the microbiome colonizing the upper vaginal tract suggest that signals from specific microbiota affect parturition (145, 146). Detailed multiomic analyses of the vaginal microbiota showed that risk for preterm birth was inversely related to the levels of Lactobacillus crispatus and was associated with taxa whose presence is correlated with elevated levels of proinflammatory cytokines in cervicovaginal fluid (140, 147149). Although the mechanistic link between microbiome and parturition remains elusive, a longitudinal study integrating vaginal microbiota and cervicovaginal fluid immunophenotype in a cohort of women at high risk for preterm birth indicates involvement of maternal immune function, especially the complement system (150). Future studies may reveal microbiome-based biomarkers and therapeutics for preterm birth risk detection and prevention, respectively. Ongoing analyses linking vaginal microbiome community state and maternal immune function with risk for preterm birth support the concept that external factors contribute to the inflammatory load of pregnancy, affecting the trajectory toward the threshold for inflammation-induced P4/PR withdrawal.

As described above, proinflammatory stimuli may trigger parturition by inducing P4 withdrawal via systemic (e.g., PGF-induced AKR1C1/20αHSD in CL cells), local (e.g., inflammatory cytokine induction of AKR1C1/20αHSD in uterine target cells), and/or functional (e.g., inflammatory cytokine modulation of PR-A transcriptional activity) mechanisms. It is possible, therefore, that the hormonal control of parturition is governed by the net inflammatory load on the uterine tissues derived from the maternal and/or fetal compartments converging to induce P4/PR withdrawal. It is also possible that the various modes for P4/PR withdrawal have specific sensitivity to inflammatory stimuli and that this is species dependent; this would account for the variation that has been observed in the hormonal control of normal term parturition seen across viviparous species. In this threshold-limited model, loss of P4/PR-B anti-inflammatory activity coupled with tissue-level inflammation would lead to a local positive-feedback inflammatory state that increases production of PGs to promote myometrial contraction, remodeling of the cervix ECM, and weakening of the amnion membrane. Loss of P4/PR-B would also remove inhibition of ESR1, leading to increased ERα in myometrial and cervical stromal cells. This increase in ERα would allow E2 to increase OTR expression and sensitivity to OT and increase expression of metalloproteinases to remodel the ECM in the cervix. Moreover, loss of P4/PR signaling would deactivate the ZEB transcription factors via the miRNA-200s, leading to derepression of genes encoding CAPs and inflammatory mediators and 20αHSD (via loss of STAT5b). The net effect would be to produce a phenotype in uterine cells that allows uterotonic hormones to induce active labor (FIGURE 7).

FIGURE 7.

FIGURE 7.

The inflammatory threshold hypothesis for the control of parturition timing. During pregnancy, inflammatory stimuli derived from multiple intrinsic (e.g., fetal development, placenta senescence) and extrinsic (e.g., maternal stress, intrauterine infection) factors contribute to a net inflammatory load impacting the uterine tissues. For most of pregnancy the progesterone (P4)/P4 receptor (PR) anti-inflammatory block to labor prevents uterine tissue-level inflammation. Above a threshold level, inflammatory stimuli induce P4 withdrawal via mechanism outlined in FIGURE 4. Loss of the P4/PR block to labor allows a positive-feedback proinflammatory state in the uterine tissues that induces active labor via increased contraction-associated protein (CAP) gene expression and increased production of and sensitivity to prolabor uterotonins. The timing of parturition is determined by the inflammatory load trajectory and the inflammatory load threshold for inflammation-induced P4/PR withdrawal. The threshold is normally reached at term but can be reached earlier in pathological states (e.g., intrauterine infection).

5. UTEROTONIC HORMONES

The parturition process involves uterotonic hormones that directly affect contraction of the myometrium, dilation of the cervix, and weakening of the amnion membrane. Two key uterotonins are PGs and OT. In addition, E2/ERα signaling in myometrial and cervical stromal cells increases responsiveness to PGs and OT and promotes cervix softening. It is notable that active labor in response to induced P4/PR withdrawal (e.g., ovariectomy or RU486 treatment) has a latency, which likely is the time required for gene expression changes and tissue level processes that alter uterotonin production and responsiveness to produce the labor phenotype.

5.1. Estrogen

Animal studies show that E2 increases expression of genes in myometrial, cervical, and decidual cells whose products promote tissue-level changes associated with parturition (151). This includes proteolytic enzymes that remodel the cervix and amnion ECM to cause cervix softening needed for dilation and membrane weakening required for rupture (152, 153). E2 also increases responsiveness to uterotonic hormones, especially OT and PGs, by increasing expression of genes encoding OT and PG receptors in uterine cells (154156) and enhances contraction efficiency by increasing expression of GJA1 to promote formation of gap junctions between myometrial cells (157160) necessary for synchronized contractions.

During the latter half of human pregnancy placental trophoblast cells convert C19 androgen produced by the fetal adrenal cortex to estrogens (E2, estrone, estriol, and estetrol) (161163). Consequently, the uterine tissues are exposed to a high-estrogen milieu for most of human pregnancy (110, 164). Yet for much of this time the uterus is not responsive to prolabor estrogen actions and remains quiescent. This is likely because of dominance of the P4/PR block and P4/PR inhibition of ESR1, thus desensitizing uterine cells to prolabor E2/ERα-mediated actions.

The role of estrogens in the hormonal control of human parturition timing is unclear. Pregnancies affected by abnormally high placental estrogens due to congenital fetal adrenal hyperplasia have normal term parturition (109). The timing and process of parturition are also normal in cases of placental aromatase deficiency (165167), anencephaly (168, 169), placenta sulfatase deficiency (170172), and congenital adrenal lipoid hyperplasia (173, 174), in which maternal estrogen levels are significantly lower than normal. Notably, in such pregnancies E2 production by maternal tissues persists, and therefore the uterine tissues are exposed to some, albeit lowered, estrogenic drive that may be sufficient to promote parturition upon P4/PR withdrawal. A plausible explanation is that for most of human pregnancy inhibition of ESR1 expression by P4/PR desensitizes uterine cells to proparturition effects of estrogens. This is reversed upon P4/PR withdrawal, allowing even lower than normal levels of estrogens to alter the uterine tissues in favor of parturition. Thus, as with other species, in human parturition P4/PR withdrawal and E2/ERα activation appear to be synchronized.

5.2. Oxytocin

OT, a small (9 amino acid) neuropeptide, is produced in the supraoptic and paraventricular nuclei of the hypothalamus and by decidual cells in the gravid uterus and plays a key role in the hormonal control of parturition (for review see Refs. 2, 175). OT production by the decidua and OXTR expressed by myometrial cells are each upregulated by estrogen (176178). OT via the OXTR is a potent uterotonin that induces uterine contractions during the process of parturition. These effects occur mainly during the expulsive phase of active labor. Expression of OXTR in myometrial cells is increased by E2/ERα and P4/PR withdrawal (175, 179181), leading to increased responsiveness to OT (182). Locally produced OT may be a key component of the estrogen proparturition effects on the uterus. Interestingly, in decidual stromal cells OT increases PG production, which could augment contraction of adjacent myometrium (178).

Interaction of OT with receptors on myometrial cells is controlled by a balance between its production by the brain and decidual cells and its inactivation by oxytocinase, a cystine aminopeptidase produced by the decidua, chorion, and placenta (183, 184). For most of pregnancy, metabolism of OT prevails. Secretion of OT from the posterior pituitary occurs as part of the lactation letdown reflex and during labor in response to cervical dilation and vaginal distension (175, 185), referred to as the Ferguson reflex, a neuroendocrine loop that creates positive feedback to augment uterine contractions in response to cervix dilation and vaginal distension. OT secretion from the posterior pituitary may also be controlled in a circadian manner and in response to environmental stressors (186), which could be a mechanism via which psychosocial or other external influences are transduced to influence the process of parturition.

Whether pituitary OT is essential for parturition is questionable, since pregnancy and parturition are normal in women with a dysfunctional posterior pituitary (187), and parturition is delayed but not prevented in clinical studies and animal models of inhibition of OT with OXTR antagonists (188190). Moreover, mice lacking OT have normal term parturition but dysfunctional lactation (191, 192). Overall, current data suggest that the role of OT in parturition is complex and redundant. Although it is a potent stimulatory uterotonin and contributes to the expulsive phase of labor and produces the postpartum tonic contraction of the uterus to facilitate hemostasis, it does not appear to be necessary for the initiation of parturition, and the full parturition cascade can proceed in the absence of OT action. This likely reflects redundancy in the hormonal control of parturition.

5.3. Prostaglandins

PGs, especially PGF and PGE2, play major paracrine roles in the hormonal control of the timing and process of parturition (193196). PGs are synthesized by various cell types including resident immune cells, especially activated macrophages and neutrophils in the placenta, amnion, and chorion-decidua and myometrium. PGs are formed from arachidonic acid by the actions of phospholipase A2 and the prostaglandin-endoperoxide synthase-1 and -2 (PTGS1 and PTGS2) enzymes (197). In the human gestational tissues PTGS2 (aka COX2) is induced by proinflammatory stimuli and is the key rate-limiting enzyme that provides substrate, PGH2, for the PG synthetic pathway. PGH2 is then converted to PGF2a, PGE2 and PGI2, PGD2, and thomboxane A2 by specific isomerases (197).

Tissue levels of PGs are decreased by the 15-hydroxyprostaglandin dehydrogenase (HPGD) enzyme that converts PGs to inactive forms (198). Interestingly, HPGD levels are high in the chorion and decrease during active labor at term and in preterm labor (spontaneous or infection associated) (199, 200). The data suggest that regionalization of HPGD serves to regulate PG access to the myometrium and cervix. In this model, loss of the HPGD chorion barrier may provide a mechanism by which the human fetus triggers parturition, allowing amnion PGs (possibly produced in response to intrauterine infection) to access the uterine tissues.

Clinical and animal studies show that pharmacological inhibition of PG synthesis inhibits labor (194). In humans, PG levels in the amniotic fluid increase during the prelude to labor (201), suggesting that PGs contribute to labor induction. This is applied clinically by exogenous administration of PGF and PGE2 to promote uterine contraction and cervical dilation. In mice, parturition at term is triggered by increased expression and activity of Ptgs1 in the uterine epithelium, which increases PGF release to induce CL regression and 20αHSD-mediated and systemic P4 withdrawal (188). In sheep, increased PGE2 production by placental trophoblast cells increases sensitivity of fetal adrenal cortical cells to ACTH and activity of the fetal HPA axis to produce the cortisol surge that triggers parturition via systemic P4 withdrawal (107). Human parturition involves increased expression of PTGS2 in the gestational tissues; this leads to increased local and amniotic fluid levels of PGF and PGE2 (202204) that may induce functional P4 withdrawal by inhibiting PR transcriptional activity and/or local P4 withdrawal by inducing 20αHSD-mediated P4 metabolism.

Specific actions of PGs on the gravid uterus are controlled by the type of PG synthesized, the extent to which they are locally inactivated by HPGD, and the temporal, spatial, and cell type-specific pattern of PG receptors on target cells. PGF induces myometrial cell contraction via the FP receptor. PGE2 induces myometrial contraction via the EP1 and EP3 receptors and relaxation via the EP2 and EP4 receptors (205, 206). FP and EP3 receptors are present in low quantity in the myometrium for most of pregnancy, likely a consequence of P4/PR inhibition. At parturition, FP receptors increase in the uterine fundus but decline in the lower uterine segment, whereas EP4 expression increases in the lower segment (205, 206). This is consistent with the concept that most of the expulsive force during labor is generated by the fundus, whereas the lower uterine segment must be less contracted to allow movement of the fetus through the dilated cervix.

6. PLACENTAL PEPTIDE HORMONES

The placenta is a remarkable endocrine organ producing multiple hormones whose function is generally to modify maternal physiology in favor of maintaining pregnancy and providing the fetus with oxygen and nutrients. The role of placental hormones in the control of parturition varies among species. As discussed above, the human placenta provides P4 to maintain pregnancy. Whether the placental trophoblast cells and the syncytiotrophoblast participate in the hormonal control of parturition is not clearly apparent. A plethora of hormones are produced by the placenta and are released into the maternal compartment, and most of these affect maternal physiology to favor pregnancy (207). One hypothesis for the contribution of placental signals in parturition is that senescence of placental cells and tissues occurs with advancing gestation, producing local senescence-associated inflammatory factors that contribute to the inflammatory load on the adjacent uterine tissues (135, 208).

Corticotropin-releasing hormone (CRH) produced by cells in the human placenta (syncytiotrophoblast and trophoblast) is thought to play an intermediary role to integrate fetal signals to initiate parturition. In human pregnancy, production of placental CRH, which is identical to CRH produced by the hypothalamus, can be detected in the maternal circulation beginning in the second trimester. Levels increase with advancing gestation, and that increase becomes exponential during the third trimester. The rate of maternal CRH increase can serve as a biomarker to predict proximity to parturition. When compared to the trajectory of maternal CRH in women with a normal term delivery, a higher rate of increase beginning early in the second trimester is predictive of preterm birth and a lower rate of increase is predictive of postterm birth (209, 210). An important concept to emerge from these studies is that events occurring early in pregnancy (e.g., factors affecting placental CRH production) set a trajectory for pregnancy outcome (210, 211). This could reflect a temporal and developmental link between the conceptus and parturition in human pregnancy whereby placental CRH reflects the developmental trajectory of the placenta and/or the fetus (210).

In the human fetus, placental CRH may affect the HPA axis by increasing production of pituitary ACTH and directly affecting fetal adrenal cortical cells by increasing production of dehydroepiandrosterone (DHEA). As DHEA is the principal C19 substrate for estrogen production by the human placenta, this would increase maternal estrogen levels (212). Importantly, unlike the adult HPA axis, in which cortisol inhibits CRH production, cortisol stimulates CRH production by human placental trophoblast cells (213, 214). This may produce a positive feedback loop whereby fetal cortisol increases CRH, which then increases maternal E2 levels (via increased DHEA). E2 could then participate in the hormonal control of parturition. Although plausible, this model must be considered from the perspective of data described above suggesting that the fetal HPA axis and maternal E2 levels do not participate in the hormonal control of human parturition (FIGURE 8).

FIGURE 8.

FIGURE 8.

Proposed model in which maternal and fetal cortisol stimulate corticotropin-releasing hormone (CRH) production by the trophoblast. This would generate positive feedback loops to result in increased estradiol (E2) to promote maternal expression of contraction-associated proteins (CAPs) and contribute to the onset of parturition. Cortisol-induced fetal organ maturation is also positively affected by placental-fetal feedback in this system. DHEA, dehydroepiandrosterone. Image is an adaptation of material from Ref. 215.

7. EVOLUTIONARY PERSPECTIVE

As with all aspects of reproductive biology, the hormonal control of parturition is likely subjected to strong natural selection. Parturition is a major event in the viviparous reproduction process and imparts a significant risk for maternal and neonate survival, and consequently reproductive fitness. As reproductive fitness drives natural selection, parturition traits that optimize reproduction in the context of other traits, some of which are species specific, in the reproduction process, such as seasonality, cyclical or continuous fertility, litter size, and neonate maturity, were likely subjected to strong selection pressure. This explains, at least in part, the diversity among viviparous species in the hormonal control of parturition.

The hormonal control of human parturition is thought to have been influenced by the evolution of encephalization, a trait unique to the hominid lineage that increased the size and complexity of the brain (216218). This concept posits that benefits of encephalization in the hominid lineage produced strong positive selection pressure leading to a progressive increase in the rate of fetal brain growth and the size of the fetal head (FIGURE 9). The obstetric dilemma hypothesis predicts that the benefits of encephalization would be lost when the size of the fetal head exceeds the size of the maternal pelvic outlet (cephalopelvic disproportion), leading to reproduction failure (217, 219, 220). Coupled with the mechanical problems, the metabolic crossover hypothesis (221) suggests that energy required for rapid fetal brain growth is a limiting factor for pregnancy especially when it exceeds the capacity for maternal energy supply. One biological solution to the encephalization-related problems is to alter the hormonal control of parturition to shorten gestation such that the fetus is born before its brain/head size becomes larger than the pelvic outlet and the energy requirements for continued in utero fetal brain growth exceed the capacity for maternal energy supply. As discussed above, the timing of parturition is controlled by multiple drivers mediated by hormonal interactions that converge to induce P4/PR withdrawal. One intriguing possibility is that changes in PGR gene sequence altered the PR to increase sensitivity to PR-mediated functional P4/PR withdrawal. Recent studies of PGR gene sequence variation between human populations indicate that changes around the serine 344/345 locus affect risk for preterm birth (222). The Neanderthal genome suggests differences in the PGR gene compared with Homo sapiens that could have affected reproductive fitness (223). Interestingly, in the Neanderthal PGR has a threonine at position 344 instead of a serine at 344 in humans. Whether these changes affect sensitivity to inflammation-induced phosphorylation remains unknown and the subject of ongoing studies. It is tempting to speculate, however, that in Homo sapiens the presence of a serine at the 344 position increased sensitivity to inflammation-induced parturition via pSer344/345-PRA-mediated functional P4/PR withdrawal that favored a solution to the obstetric dilemma, giving Homo sapiens a reproductive advantage. It is also possible that the change increased the risk for preterm birth, which is counterintuitive in terms of reproductive fitness, since preterm birth increases risk for neonate death and reproduction failure. However, as natural selection operates at the species level and over multiple generations, it is plausible that the benefits of encephalization outweighed the cost of reproduction failure due to preterm birth. Another evolutionary consideration is that although preterm birth imparts cost on the fetus in the current pregnancy, it preserves the female’s capacity for future pregnancies and/or to care for existing offspring. Clearly, pregnancy is a state of physiological stress for the female, especially late in gestation when, in the case of hominids, parturition involves birthing a large-headed fetus whose brain has a high energy demand. From a species-level evolutionary perspective natural selection would favor hormonal signals that trigger parturition to favor survival of the female and neonate, but with preference for the female based on her established capacity for future pregnancy and the possibility that she is responsible for the survival of current offspring.

FIGURE 9.

FIGURE 9.

Visual comparison of the relative proportions of the fetal skull to the maternal pelvis in primates shows a maximal cephalopelvic ratio in humans. The obstetric dilemma hypothesis proposes that the biomechanical limitations of the pelvic outlet (blue outline) and the size of the human head (red outline) necessitate that parturition occur at a time that results in relative altriciality of the human neonate. Image is an adaptation of material from Ref. 2.

8. CLINICAL PERSPECTIVE

Attempts to clinically manipulate the timing of parturition are inherent in the practice of obstetrics. Numerous scenarios exist in which delivery before the onset of spontaneous labor at term, and slowing or halting preterm labor, is beneficial for the parturient and the fetus (224, 225). To this end, leveraging the hormonal mediators of parturition has been a primary focus in strategies to both induce and prevent parturition (FIGURE 10).

FIGURE 10.

FIGURE 10.

The hormonal regulation of parturition provides an opportunity for therapeutic intervention, both for prevention of preterm labor (left) and for induction of labor (right). Interventions that directly target hormonal pathways are listed for the prevention of preterm labor (left) and induction of labor (right). Attempts to prevent preterm labor have targeted progesterone (P4) receptor (PR), oxytocin receptor (OTR), prostaglandin (PG) synthesis, and estrogen receptor (ER). These interventions include some with therapeutic benefit in a limited population (e.g., vaginal P4 for short cervix), some with unclear or unproven benefit (e.g., atosiban), and some harmful and contraindicated in pregnancy [e.g., diethylstilbestrol (DES)]. Medically indicated labor and management of miscarriage, in contrast, utilize all of the listed interventions routinely as the standard of care in common clinical scenarios.

8.1. Induction of Parturition

Medically indicated induction of labor, clinically defined as rhythmic, forceful, and painful contractions of the myometrium, softening and dilation (referred to as ripening) of the cervix, and weakening and possibly rupture of the amnion-chorion membrane, utilizes pharmacological methods that promote cervical ripening and uterine contractions via endogenous uterotonin signaling pathways. The principal agents used are synthetic analogs of OT (e.g., Pitocin) and PGE2 (e.g., dinoprostone) and PGE1 (e.g., misoprostol) (226230). The PG analogs are used for cervical ripening and also promote contractions. In contrast, although OT is effective for inducing myometrial contractions, it is often used as an adjunct for cervical ripening (231). OT and PG analogs are presumed to function via their endogenous uterotonic pathways and possibly override or circumvent the P4/PR block to parturition (120).

RU486 and misoprostol (PGE1) are used for cervical ripening in preparation for medical and surgical abortion and for the management of miscarriage (232, 233). As a PR antagonist, RU486 is a potent abortifacient and in animal models alone is sufficient to induce the full parturition cascade (234), which supports the P4/PR block hypothesis. Clinically, RU486 is used for cervical ripening in term pregnancy in clinical trials (235, 236); RU486 is not broadly used for induction of labor in viable pregnancies. This is likely due, at least in part, to regulatory barriers to its use (237).

The nonpharmacological, mechanical methods of cervical ripening (intracervical balloon catheters and, for pregnancy termination, osmotic dilators) and other obstetric interventions (membrane sweeping and amniotomy) are suspected to act at least in part via stimulation of endogenous PGs (238, 239). These methods of inducing parturition are often used in combination or sequentially in a single patient, and, overall, induction of labor has been demonstrated to decrease the rate of cesarean delivery compared to expectant management (240).

In a minority of patients who undergo cervical ripening and induction of labor, active labor is not achieved and an unplanned cesarean delivery is required (241). The risk factors and clinical scenarios that contribute to failed induction of labor in an individual patient are numerous, and so it follows that the contributing mechanisms of failure are likely complex and heterogeneous. Based on the seesaw and threshold models of parturition initiation, one can surmise that insufficient cervical ripening and failed induction of labor may result from a failure to overcome the P4/PR block. Whether a similar mechanism contributes to dysfunctional spontaneous labor (when parturition is initiated but does not progress normally; also a contributor to unplanned cesarean delivery) is also unknown.

8.2. Prevention of Preterm Parturition

Preterm birth is a substantial cause of neonatal morbidity and mortality. A contemporary epidemiological study has demonstrated that 65% of preterm births from 20 weeks to 36 weeks 6 days of gestation result from spontaneous preterm labor (versus iatrogenic preterm delivery) (242). The clinical presentations of preterm labor are heterogeneous; therefore, it is not surprising that translational and clinical research have failed to identify a universal prophylaxis or therapy for preterm labor. Several therapies have been tested that each attempt to restore uterine/cervical quiescence by exploiting hormonal mediators of parturition.

Because of the central role of the P4/PR block to parturition, progestins have been a major focus over several decades for both early pregnancy maintenance (243, 244) and prevention of preterm labor (for recent reviews on progestin-based prophylaxis for preterm birth prevention see Refs. 245247). Boosting the P4/PR block and/or preventing its withdrawal is an attractive strategy in theory. Indeed, despite early (1950–1960) studies showing that P4 treatment has no tocolytic benefit for women in active labor (248), subsequent, albeit small, trials found that prophylaxis with the caproate salt of 17-hydroxyprogesterone (17HPC) may reduce preterm birth risk (249). Those findings motivated larger clinical trials of 17HPC prophylaxis (250) and vaginal P4 (251) for preterm birth prevention in women with elevated risk due to a history of preterm birth. Both trials produced beneficial outcomes, with a reduced incidence of preterm birth in women receiving progestin prophylaxis. Subsequent trials of 17HPC (252) and vaginal P4 (253), however, showed that neither therapy reduces the risk for preterm birth. Consequently, 17HPC is no longer recommended for preterm birth prevention. Vaginal P4 for women with increased preterm birth risk due to short cervix remains the only progestin-based prophylaxis in clinical use (254, 255). The general failure of progestin supplementation to prevent preterm birth is expected, since levels of P4 in the maternal circulation are persistently high even in pregnancies that end preterm and there is no evidence that preterm birth risk is related to endogenous P4 deficiency that can be corrected clinically (247). Intriguingly, promegestone (aka R5020), a P4 analog that binds the PR with high affinity and is not metabolized by 20αHSD/AKR1C1, delays parturition in a mouse model of inflammation-induced preterm birth (256). Clearly, improved understanding of the mechanism(s) for, and hormonal control of, P4/PR withdrawal in the physiology of human parturition will provide more effective therapeutic strategies to clinically control the timing of birth.

Additional tocolytic strategies to intervene on preterm contractions include atosiban, which functions as an OT receptor antagonist and also has anti-inflammatory effects in myometrial cells (257). This medication has demonstrated safety and is approved for clinical use in Europe; however, its efficacy in improving neonatal outcomes is uncertain, and a clinical trial to address this is ongoing (258). Indomethacin, an inhibitor of prostaglandin synthesis, is also utilized as a tocolytic in limited clinical scenarios for brief durations at early gestational ages; its overall effectiveness for improvement in neonatal morbidity has not been sufficiently demonstrated (259, 260). The use of indomethacin will continue to remain limited because of the risk of premature closure of the fetal ductus arteriosus, a well-known consequence of PG synthesis inhibition in the late fetal period.

The potential for fetal effects of hormonally directed therapies is not limited to PG inhibition. Diethylstilbestrol (DES) is an ER agonist that was used in the 1940s–1960s for, among other indications, prevention of miscarriage and preterm birth. It was ultimately found to cause gynecologic cancers in females who were exposed in utero, congenital anomalies of the reproductive tract in both females and males following in utero exposure, and concerns for third-generation effects (261). This sobering history underscores the importance of appreciation of the holistic maternal, placental, and embryonic/fetal effects of any pregnancy intervention, particularly when manipulating hormonal pathways that have distinct and essential roles in the mother and fetus.

9. CONCLUSIONS

The hormonal control of parturition is complex and redundant, integrating multiple physiological signals. Some of these signals are overt stressors, whereas others allow maternal adaptation to the conceptus and receipt of the fetus’ indication of its readiness for neonatal life. These endocrine and paracrine signals occur consequent to maternal and fetal-placental inputs and collectively result in uterine contractions, cervix softening/dilation, and membrane rupture and eventually in the completion of parturition. Upon this framework, unraveling the hormonal control of parturition must acknowledge that different viviparous species possess diverse controls for parturition timing and process. This diversity is shaped by species-specific natural selection that favors a parturition control system that increases reproductive fitness in the context of the extant constellation of reproductive traits. Despite variability, the P4 block to parturition mediated by the nuclear PRs and the induction of parturition by withdrawal of P4/PR signaling appears to be a conserved trait. Upstream signals that induce P4/PR withdrawal initiate downstream transducers to induce anatomic and physiological changes mediated by uterotonins (e.g., OT, PGs) that complete the parturition process. To better understand the physiology of parturition, analysis of existing data and those from ongoing and future studies must acknowledge and reconcile the diverse and common pathways across species in the context of natural history and evolutionary biology. This approach is yielding novel concepts to advance understanding of the hormonal control of parturition that will translate clinically to address adverse birth timing conditions, especially preterm labor leading to birth of a premature neonate.

GRANTS

This work was funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development (HD097279; HD103676) and the March of Dimes Prematurity Research Center, Collaborative of Ohio.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

S.M. conceived and designed research; S.M. prepared figures; E.H.-S. and S.M. drafted manuscript; E.H.-S. and S.M. edited and revised manuscript; E.H.-S. and S.M. approved final version of manuscript.

REFERENCES

  • 1. Mesiano S. The endocrinology of human pregnancy and fetoplacental neuroendocrine development. In: Yen and Jaffe's Reproductive Endocrinology: Physiology, Pathophysiology, and Clinical Management, edited by Strauss JF, Barbieri RL.. Philadelphia, PA: Elsevier, 2009, p. 249–282. [Google Scholar]
  • 2. Mesiano S, DeFranco E, Muglia L. Parturition. In: Knobil and Neill’s Physiology of Reproduction, edited by Plant TM, Zeleznik A.. London: Academic Press, 2014, p. 1857–1925. [Google Scholar]
  • 3. Casey ML, Macdonald PC. Endocrine changes of pregnancy. In: Williams Textbook of Endocrinology, edited by Wilson JD, Foster DW, Kronenberg HM, Larsen PR.. Philadelphia, PA: W.B. Saunders, 1998, p. 1259. [Google Scholar]
  • 4. Csapo A. The luteo-placental shift, the guardian of pre-natal life. Postgrad Med J 45: 57–64, 1969. doi: 10.1136/pgmj.45.519.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Csapo AI, Pulkkinen M. Indispensability of the human corpus luteum in the maintenance of early pregnancy. Lutectomy evidence. Obstet Gynecol Surv 33: 69–81, 1978. doi: 10.1097/00006254-197802000-00001. [DOI] [PubMed] [Google Scholar]
  • 6. Tuckey RC. Progesterone synthesis by the human placenta. Placenta 26: 273–281, 2005. doi: 10.1016/j.placenta.2004.06.012. [DOI] [PubMed] [Google Scholar]
  • 7. Tulchinsky D, Hobel CJ, Yeager E, Marshall JR. Plasma estrone, estradiol, estriol, progesterone, and 17-hydroxyprogesterone in human pregnancy. I. Normal pregnancy. Am J Obstet Gynecol 112: 1095–1100, 1972. doi: 10.1016/0002-9378(72)90185-8. [DOI] [PubMed] [Google Scholar]
  • 8. Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H, Chambon P. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J 9: 1603–1614, 1990. doi: 10.1002/j.1460-2075.1990.tb08280.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Charo RA. A political history of RU-486. In: Biomedical Politics, edited by Hanna KE. Washington, DC: National Academies Press, 1991. [PubMed] [Google Scholar]
  • 10. Ulmann A, Teutsch G, Philibert D. Ru 486. Sci Am 262: 42–48, 1990. doi: 10.1038/scientificamerican0690-42. [DOI] [PubMed] [Google Scholar]
  • 11. Csapo A. Progesterone block. Am J Anat 98: 273–291, 1956. doi: 10.1002/aja.1000980206. [DOI] [PubMed] [Google Scholar]
  • 12. Wu SP, Anderson ML, Wang T, Zhou L, Emery OM, Li X, DeMayo FJ. Dynamic transcriptome, accessible genome, and PGR cistrome profiles in the human myometrium. FASEB J 34: 2252–2268, 2020. doi: 10.1096/fj.201902654R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Hajagos-Tóth J, Bóta J, Ducza E, Samavati R, Borsodi A, Benyhe S, Gáspár R. The effects of progesterone on the alpha2-adrenergic receptor subtypes in late-pregnant uterine contractions in vitro. Reprod Biol Endocrinol 14: 33, 2016. doi: 10.1186/s12958-016-0166-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Vivat V, Cohen-Tannoudji J, Revelli JP, Muzzin P, Giacobino JP, Maltier JP, Legrand C. Progesterone transcriptionally regulates the beta 2-adrenergic receptor gene in pregnant rat myometrium. J Biol Chem 267: 7975–7978, 1992. doi: 10.1016/S0021-9258(18)42394-0. [DOI] [PubMed] [Google Scholar]
  • 15. Wray S. Uterine contraction and physiological mechanisms of modulation. Am J Physiol Cell Physiol 264: C1–C18, 1993. doi: 10.1152/ajpcell.1993.264.1.C1. [DOI] [PubMed] [Google Scholar]
  • 16. Haluska GJ, West NB, Novy MJ, Brenner RM. Uterine estrogen receptors are increased by RU486 in late pregnant rhesus macaques but not after spontaneous labor. J Clin Endocrinol Metab 70: 181–186, 1990. doi: 10.1210/jcem-70-1-181. [DOI] [PubMed] [Google Scholar]
  • 17. Ilicic M, Butler T, Zakar T, Paul JW. The expression of genes involved in myometrial contractility changes during ex situ culture of pregnant human uterine smooth muscle tissue. J Smooth Muscle Res 53: 73–89, 2017. doi: 10.1540/jsmr.53.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tantengco OA, Richardson LS, Vink J, Kechichian T, Medina PM, Pyles RB, Menon R. Progesterone alters human cervical epithelial and stromal cell transition and migration: implications in cervical remodeling during pregnancy and parturition. Mol Cell Endocrinol 529: 111276, 2021. doi: 10.1016/j.mce.2021.111276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Timmons B, Akins M, Mahendroo M. Cervical remodeling during pregnancy and parturition. Trends Endocrinol Metab 21: 353–361, 2010. doi: 10.1016/j.tem.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Word RA, Li XH, Hnat M, Carrick K. Dynamics of cervical remodeling during pregnancy and parturition: mechanisms and current concepts. Semin Reprod Med 25: 69–79, 2007. doi: 10.1055/s-2006-956777. [DOI] [PubMed] [Google Scholar]
  • 21. Yellon SM. Contributions to the dynamics of cervix remodeling prior to term and preterm birth. Biol Reprod 96: 13–23, 2017. doi: 10.1095/biolreprod.116.142844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Yoshida K, Jayyosi C, Lee N, Mahendroo M, Myers KM. Mechanics of cervical remodelling: insights from rodent models of pregnancy. Interface Focus 9: 20190026, 2019. doi: 10.1098/rsfs.2019.0026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Csapo AJ. The “seesaw” theory of the regulatory mechanism of pregnancy. Am J Obstet Gynecol 121: 578–581, 1975. [PubMed] [Google Scholar]
  • 24. Chappell PE, Lydon JP, Conneely OM, O’Malley BW, Levine JE. Endocrine defects in mice carrying a null mutation for the progesterone receptor gene. Endocrinology 138: 4147–4152, 1997. doi: 10.1210/endo.138.10.5456. [DOI] [PubMed] [Google Scholar]
  • 25. Conneely OM, Mulac-Jericevic B, Lydon JP. Progesterone-dependent regulation of female reproductive activity by two distinct progesterone receptor isoforms. Steroids 68: 771–778, 2003. doi: 10.1016/s0039-128x(03)00126-0. [DOI] [PubMed] [Google Scholar]
  • 26. Lydon JP, DeMayo FJ, Conneely OM, O’Malley BW. Reproductive phenotypes of the progesterone receptor null mutant mouse. J Steroid Biochem Mol Biol 56: 67–77, 1996. doi: 10.1016/0960-0760(95)00254-5. [DOI] [PubMed] [Google Scholar]
  • 27. Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR, Montgomery CA Jr, Shyamala G, Conneely OM, O'Malley BW. Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities. Genes Dev 9: 2266–2278, 1995. doi: 10.1101/gad.9.18.2266. [DOI] [PubMed] [Google Scholar]
  • 28. Mulac-Jericevic B, Lydon JP, DeMayo FJ, Conneely OM. Defective mammary gland morphogenesis in mice lacking the progesterone receptor B isoform. Proc Natl Acad Sci USA 100: 9744–9749, 2003. doi: 10.1073/pnas.1732707100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Mulac-Jericevic B, Mullinax RA, DeMayo FJ, Lydon JP, Conneely OM. Subgroup of reproductive functions of progesterone mediated by progesterone receptor-B isoform. Science 289: 1751–1754, 2000. doi: 10.1126/science.289.5485.1751. [DOI] [PubMed] [Google Scholar]
  • 30. Wu SP, Wang T, Yao ZC, Peavey MC, Li X, Zhou L, Larina IV, DeMayo FJ. Myometrial progesterone receptor determines a transcription program for uterine remodeling and contractions during pregnancy. PNAS Nexus 1: pgac155, 2022. doi: 10.1093/pnasnexus/pgac155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Siiteri PK, Febres F, Clemens LE, Chang RJ, Gondos B, Stites D. Progesterone and maintenance of pregnancy: is progesterone nature's immunosuppressant? Ann NY Acad Sci 286: 384–397, 1977. doi: 10.1111/j.1749-6632.1977.tb29431.x. [DOI] [PubMed] [Google Scholar]
  • 32. Stites DP, Siiteri PK. Steroids as immunosuppressants in pregnancy. Immunol Rev 75: 117–138, 1983. doi: 10.1111/j.1600-065x.1983.tb01093.x. [DOI] [PubMed] [Google Scholar]
  • 33. Bastek JA, Gómez LM, Elovitz MA. The role of inflammation and infection in preterm birth. Clin Perinatol 38: 385–406, 2011. doi: 10.1016/j.clp.2011.06.003. [DOI] [PubMed] [Google Scholar]
  • 34. Keelan JA, Blumenstein M, Helliwell RJ, Sato TA, Marvin KW, Mitchell MD. Cytokines, prostaglandins and parturition—a review. Placenta 24, Suppl A: S33–S46, 2003. doi: 10.1053/plac.2002.0948. [DOI] [PubMed] [Google Scholar]
  • 35. Norman JE, Bollapragada S, Yuan M, Nelson SM. Inflammatory pathways in the mechanism of parturition. BMC Pregnancy Childbirth 7, Suppl 1: S7, 2007. doi: 10.1186/1471-2393-7-S1-S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Osman I, Young A, Jordan F, Greer IA, Norman JE. Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term. J Soc Gynecol Investig 13: 97–103, 2006. doi: 10.1016/j.jsgi.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 37. Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F, Greer IA, Norman JE. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol Hum Reprod 9: 41–45, 2003. doi: 10.1093/molehr/gag001. [DOI] [PubMed] [Google Scholar]
  • 38. Sadowsky DW, Adams KM, Gravett MG, Witkin SS, Novy MJ. Preterm labor is induced by intraamniotic infusions of interleukin-1beta and tumor necrosis factor-alpha but not by interleukin-6 or interleukin-8 in a nonhuman primate model. Am J Obstet Gynecol 195: 1578–1589, 2006. doi: 10.1016/j.ajog.2006.06.072. [DOI] [PubMed] [Google Scholar]
  • 39. Thomson AJ, Telfer JF, Young A, Campbell S, Stewart CJ, Cameron IT, Greer IA, Norman JE. Leukocytes infiltrate the myometrium during human parturition: further evidence that labour is an inflammatory process. Hum Reprod 14: 229–236, 1999. [PubMed] [Google Scholar]
  • 40. Adams Waldorf KM, Rubens CE, Gravett MG. Use of nonhuman primate models to investigate mechanisms of infection-associated preterm birth. BJOG 118: 136–144, 2011. doi: 10.1111/j.1471-0528.2010.02728.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Baggia S, Gravett MG, Witkin SS, Haluska GJ, Novy MJ. Interleukin-1 beta intra-amniotic infusion induces tumor necrosis factor-alpha, prostaglandin production, and preterm contractions in pregnant rhesus monkeys. J Soc Gynecol Investig 3: 121–126, 1996. doi: 10.1177/107155769600300304. [DOI] [PubMed] [Google Scholar]
  • 42. Cappelletti M, Presicce P, Feiyang M, Senthamaraikannan P, Miller LA, Pellegrini M, Sim MS, Jobe AH, Divanovic S, Way SS, Chougnet CA, Kallapur SG. The induction of preterm labor in rhesus macaques is determined by the strength of immune response to intrauterine infection. PLoS Biol 19: e3001385, 2021. doi: 10.1371/journal.pbio.3001385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Gravett MG, Witkin SS, Haluska GJ, Edwards JL, Cook MJ, Novy MJ. An experimental model for intraamniotic infection and preterm labor in rhesus monkeys. Am J Obstet Gynecol 171: 1660–1667, 1994. doi: 10.1016/0002-9378(94)90418-9. [DOI] [PubMed] [Google Scholar]
  • 44. Hirsch E, Saotome I, Hirsh D. A model of intrauterine infection and preterm delivery in mice. Am J Obstet Gynecol 172: 1598–1603, 1995. doi: 10.1016/0002-9378(95)90503-0. [DOI] [PubMed] [Google Scholar]
  • 45. Kim CJ, Romero R, Chaemsaithong P, Chaiyasit N, Yoon BH, Kim YM. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol 213: S29–S52, 2015. doi: 10.1016/j.ajog.2015.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Amini P, Michniuk D, Kuo K, Yi L, Skomorovska-Prokvolit Y, Peters GA, Tan H, Wang J, Malemud CJ, Mesiano S. Human parturition involves phosphorylation of progesterone receptor-A at serine-345 in myometrial cells. Endocrinology 157: 4434–4445, 2016. doi: 10.1210/en.2016-1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Hardy DB, Janowski BA, Corey DR, Mendelson CR. Progesterone receptor plays a major antiinflammatory role in human myometrial cells by antagonism of nuclear factor-kappaB activation of cyclooxygenase 2 expression. Mol Endocrinol 20: 2724–2733, 2006. doi: 10.1210/me.2006-0112. [DOI] [PubMed] [Google Scholar]
  • 48. Lei K, Georgiou EX, Chen L, Yulia A, Sooranna SR, Brosens JJ, Bennett PR, Johnson MR. Progesterone and the repression of myometrial inflammation: the roles of MKP-1 and the AP-1 system. Mol Endocrinol 29: 1454–1467, 2015. doi: 10.1210/me.2015-1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Patel B, Peters GA, Skomorovska-Prokvolit Y, Yi L, Tan H, Yousef A, Wang J, Mesiano S. Control of progesterone receptor-A transrepressive activity in myometrial cells: implications for the control of human parturition. Reprod Sci 25: 214–221, 2018. doi: 10.1177/1933719117716775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Peters GA, Yi L, Skomorovska-Prokvolit Y, Patel B, Amini P, Tan H, Mesiano S. Inflammatory stimuli increase progesterone receptor-A stability and transrepressive activity in myometrial cells. Endocrinology 158: 158–169, 2017. doi: 10.1210/en.2016-1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Stanfield Z, Amini P, Wang J, Yi L, Tan H, Chance MR, Koyutürk M, Mesiano S. Interplay of transcriptional signaling by progesterone, cyclic AMP, and inflammation in myometrial cells: implications for the control of human parturition. Mol Hum Reprod 25: 408–422, 2019. doi: 10.1093/molehr/gaz028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Tan H, Yi L, Rote NS, Hurd WW, Mesiano S. Progesterone receptor-A and -B have opposite effects on proinflammatory gene expression in human myometrial cells: implications for progesterone actions in human pregnancy and parturition. J Clin Endocrinol Metab 97: E719–E730, 2012. doi: 10.1210/jc.2011-3251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Tibbetts TA, Conneely OM, O’Malley BW. Progesterone via its receptor antagonizes the pro-inflammatory activity of estrogen in the mouse uterus. Biol Reprod 60: 1158–1165, 1999. doi: 10.1095/biolreprod60.5.1158. [DOI] [PubMed] [Google Scholar]
  • 54. Allport VC, Pieber D, Slater DM, Newton R, White JO, Bennett PR. Human labour is associated with nuclear factor-kappaB activity which mediates cyclo-oxygenase-2 expression and is involved with the ‘functional progesterone withdrawal’. Mol Hum Reprod 7: 581–586, 2001. doi: 10.1093/molehr/7.6.581. [DOI] [PubMed] [Google Scholar]
  • 55. Khanjani S, Terzidou V, Johnson MR, Bennett PR. NFkappaB and AP-1 drive human myometrial IL8 expression. Mediators Inflamm 2012: 504952, 2012. doi: 10.1155/2012/504952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lindström TM, Bennett PR. The role of nuclear factor kappa B in human labour. Reproduction 130: 569–581, 2005. doi: 10.1530/rep.1.00197. [DOI] [PubMed] [Google Scholar]
  • 57. Kalkhoven E, Wissink S, van der Saag PT, van der Burg B. Negative interaction between the RelA(p65) subunit of NF-kappaB and the progesterone receptor. J Biol Chem 271: 6217–6224, 1996. doi: 10.1074/jbc.271.11.6217. [DOI] [PubMed] [Google Scholar]
  • 58. Renthal NE, Williams KC, Montalbano AP, Chen CC, Gao L, Mendelson CR. Molecular regulation of parturition: a myometrial perspective. Cold Spring Harb Perspect Med 5: a023069, 2015. doi: 10.1101/cshperspect.a023069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Giangrande PH, Kimbrel EA, Edwards DP, McDonnell DP. The opposing transcriptional activities of the two isoforms of the human progesterone receptor are due to differential cofactor binding. Mol Cell Biol 20: 3102–3115, 2000. doi: 10.1128/MCB.20.9.3102-3115.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Jacobsen BM, Horwitz KB. Progesterone receptors, their isoforms and progesterone regulated transcription. Mol Cell Endocrinol 357: 18–29, 2012. doi: 10.1016/j.mce.2011.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Nadeem L, Shynlova O, Matysiak-Zablocki E, Mesiano S, Dong X, Lye S. Molecular evidence of functional progesterone withdrawal in human myometrium. Nat Commun 7: 11565, 2016. doi: 10.1038/ncomms11565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Kappelmann M, Bosserhoff A, Kuphal S. AP-1/c-Jun transcription factors: regulation and function in malignant melanoma. Eur J Cell Biol 93: 76–81, 2014. doi: 10.1016/j.ejcb.2013.10.003. [DOI] [PubMed] [Google Scholar]
  • 63. Karin M, Liu Z, Zandi E. AP-1 function and regulation. Curr Opin Cell Biol 9: 240–246, 1997. doi: 10.1016/s0955-0674(97)80068-3. [DOI] [PubMed] [Google Scholar]
  • 64. Mitchell JA, Lye SJ. Differential activation of the connexin 43 promoter by dimers of activator protein-1 transcription factors in myometrial cells. Endocrinology 146: 2048–2054, 2005. doi: 10.1210/en.2004-1066. [DOI] [PubMed] [Google Scholar]
  • 65. Mitchell JA, Lye SJ. Differential expression of activator protein-1 transcription factors in pregnant rat myometrium. Biol Reprod 67: 240–246, 2002. doi: 10.1095/biolreprod67.1.240. [DOI] [PubMed] [Google Scholar]
  • 66. Renthal NE, Chen CC, Williams KC, Gerard RD, Prange-Kiel J, Mendelson CR. miR-200 family and targets, ZEB1 and ZEB2, modulate uterine quiescence and contractility during pregnancy and labor. Proc Natl Acad Sci USA 107: 20828–20833, 2010. doi: 10.1073/pnas.1008301107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Williams KC, Renthal NE, Gerard RD, Mendelson CR. The microRNA (miR)-199a/214 cluster mediates opposing effects of progesterone and estrogen on uterine contractility during pregnancy and labor. Mol Endocrinol 26: 1857–1867, 2012. doi: 10.1210/me.2012-1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Muter J, Kong CS, Brosens JJ. The role of decidual subpopulations in implantation, menstruation and miscarriage. Front Reprod Health 3: 804921, 2021. doi: 10.3389/frph.2021.804921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Okada H, Nakajima T, Sanezumi M, Ikuta A, Yasuda K, Kanzaki H. Progesterone enhances interleukin-15 production in human endometrial stromal cells in vitro. J Clin Endocrinol Metab 85: 4765–4770, 2000. doi: 10.1210/jcem.85.12.7023. [DOI] [PubMed] [Google Scholar]
  • 70. Okada H, Nakajima T, Yasuda K, Kanzaki H. Interleukin-1 inhibits interleukin-15 production by progesterone during in vitro decidualization in human. J Reprod Immunol 61: 3–12, 2004. doi: 10.1016/j.jri.2003.10.002. [DOI] [PubMed] [Google Scholar]
  • 71. Nadeem L, Balendran R, Dorogin A, Mesiano S, Shynlova O, Lye S. Pro-inflammatory signals induce 20α-HSD expression in myometrial cells: a key mechanism for local progesterone withdrawal. J Cell Mol Med 25: 6773–6785, 2021. doi: 10.1111/jcmm.16681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Penning TM. Molecular endocrinology of hydroxysteroid dehydrogenases. Endocr Rev 18: 281–305, 1997. doi: 10.1210/edrv.18.3.0302. [DOI] [PubMed] [Google Scholar]
  • 73. Penning TM, Drury JE. Human aldo-keto reductases: function, gene regulation, and single nucleotide polymorphisms. Arch Biochem Biophys 464: 241–250, 2007. doi: 10.1016/j.abb.2007.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Wiest WG, Kidwell WR, Balogh K Jr.. Progesterone catabolism in the rat ovary: a regulatory mechanism for progestational potency during pregnancy. Endocrinology 82: 844–859, 1968. doi: 10.1210/endo-82-4-844. [DOI] [PubMed] [Google Scholar]
  • 75. Liggins GC, Fairclough RJ, Grieves SA, Kendall JZ, Knox BS. The mechanism of initiation of parturition in the ewe. Recent Prog Horm Res 29: 111–159, 1973. doi: 10.1016/b978-0-12-571129-6.50007-5. [DOI] [PubMed] [Google Scholar]
  • 76. Pointis G, Rao B, Latreille MT, Mignot TM, Cedard L. Progesterone levels in the circulating blood of the ovarian and uterine veins during gestation in the mouse. Biol Reprod 24: 801–805, 1981. doi: 10.1095/biolreprod24.4.801. [DOI] [PubMed] [Google Scholar]
  • 77. Williams KC, Renthal NE, Condon JC, Gerard RD, Mendelson CR. MicroRNA-200a serves a key role in the decline of progesterone receptor function leading to term and preterm labor. Proc Natl Acad Sci USA 109: 7529–7534, 2012. doi: 10.1073/pnas.1200650109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Stocco CO, Chedrese J, Deis RP. Luteal expression of cytochrome P450 side-chain cleavage, steroidogenic acute regulatory protein, 3beta-hydroxysteroid dehydrogenase, and 20alpha-hydroxysteroid dehydrogenase genes in late pregnant rats: effect of luteinizing hormone and RU486. Biol Reprod 65: 1114–1119, 2001. doi: 10.1095/biolreprod65.4.1114. [DOI] [PubMed] [Google Scholar]
  • 79. Stocco CO, Zhong L, Sugimoto Y, Ichikawa A, Lau LF, Gibori G. Prostaglandin F2alpha-induced expression of 20alpha-hydroxysteroid dehydrogenase involves the transcription factor NUR77. J Biol Chem 275: 37202–37211, 2000. doi: 10.1074/jbc.M006016200. [DOI] [PubMed] [Google Scholar]
  • 80. Ishida M, Choi JH, Hirabayashi K, Matsuwaki T, Suzuki M, Yamanouchi K, Horai R, Sudo K, Iwakura Y, Nishihara M. Reproductive phenotypes in mice with targeted disruption of the 20alpha-hydroxysteroid dehydrogenase gene. J Reprod Dev 53: 499–508, 2007. doi: 10.1262/jrd.18125. [DOI] [PubMed] [Google Scholar]
  • 81. Piekorz RP, Gingras S, Hoffmeyer A, Ihle JN, Weinstein Y. Regulation of progesterone levels during pregnancy and parturition by signal transducer and activator of transcription 5 and 20alpha-hydroxysteroid dehydrogenase. Mol Endocrinol 19: 431–440, 2005. doi: 10.1210/me.2004-0302. [DOI] [PubMed] [Google Scholar]
  • 82. Mahendroo MS, Porter A, Russell DW, Word RA. The parturition defect in steroid 5alpha-reductase type 1 knockout mice is due to impaired cervical ripening. Mol Endocrinol 13: 981–992, 1999. doi: 10.1210/mend.13.6.0307. [DOI] [PubMed] [Google Scholar]
  • 83. Paul M, Zakar T, Phung J, Gregson A, Barreda AP, Butler TA, Walker FR, Pennell C, Smith R, Paul JW. 20alpha-Hydroxysteroid dehydrogenase expression in the human myometrium at term and preterm birth: relationships to fetal sex and maternal body mass index. Reprod Sci 30: 2512–2523, 2023. doi: 10.1007/s43032-023-01183-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Junkermann H, Runnebaum B, Lisboa BP. New progesterone metabolites in human myometrium. Steroids 30: 1–14, 1977. doi: 10.1016/0039-128x(77)90131-3. [DOI] [PubMed] [Google Scholar]
  • 85. Runnebaum B, Zander J. Progesterone and 20 alpha-dihydroprogesterone in human myometrium during pregnancy. Acta Endocrinol Suppl (Copenh) 150: 3–45, 1971. [PubMed] [Google Scholar]
  • 86. Richer JK, Lange CA, Manning NG, Owen G, Powell R, Horwitz KB. Convergence of progesterone with growth factor and cytokine signaling in breast cancer. Progesterone receptors regulate signal transducers and activators of transcription expression and activity. J Biol Chem 273: 31317–31326, 1998. doi: 10.1074/jbc.273.47.31317. [DOI] [PubMed] [Google Scholar]
  • 87. Giangrande PH, McDonnell DP. The A and B isoforms of the human progesterone receptor: two functionally different transcription factors encoded by a single gene. Recent Prog Horm Res 54: 291–313, 1999. [PubMed] [Google Scholar]
  • 88. Vegeto E, Shahbaz MM, Wen DX, Goldman ME, O’Malley BW, McDonnell DP. Human progesterone receptor A form is a cell-and promoter-specific repressor of human progesterone receptor B function. Mol Endocrinol 7: 1244–1255, 1993. doi: 10.1210/mend.7.10.8264658. [DOI] [PubMed] [Google Scholar]
  • 89. Ke W, Chen C, Luo H, Tang J, Zhang Y, Gao W, Yang X, Tian Z, Chang Q, Liang Z. Histone deacetylase 1 regulates the expression of progesterone receptor A during human parturition by occupying the progesterone receptor A promoter. Reprod Sci 23: 955–964, 2016. doi: 10.1177/1933719115625848. [DOI] [PubMed] [Google Scholar]
  • 90. Merlino AA, Welsh TN, Tan H, Yi LJ, Cannon V, Mercer BM, Mesiano S. Nuclear progesterone receptors in the human pregnancy myometrium: evidence that parturition involves functional progesterone withdrawal mediated by increased expression of progesterone receptor-A. J Clin Endocrinol Metab 92: 1927–1933, 2007. doi: 10.1210/jc.2007-0077. [DOI] [PubMed] [Google Scholar]
  • 91. 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. J Clin Endocrinol Metab 87: 2924–2930, 2002. doi: 10.1210/jcem.87.6.8609. [DOI] [PubMed] [Google Scholar]
  • 92. Peavey MC, Wu SP, Li R, Liu J, Emery OM, Wang T, Zhou L, Wetendorf M, Yallampalli C, Gibbons WE, Lydon JP, DeMayo FJ. Progesterone receptor isoform B regulates the Oxtr-Plcl2-Trpc3 pathway to suppress uterine contractility. Proc Natl Acad Sci USA 118: e2011643118, 2021. doi: 10.1073/pnas.2011643118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Pieber D, Allport VC, Hills F, Johnson M, Bennett PR. Interaction between progesterone receptor isoforms in myometrial cells in human labour. Mol Hum Reprod 7: 875–879, 2001. doi: 10.1093/molehr/7.9.875. [DOI] [PubMed] [Google Scholar]
  • 94. Condon JC, Jeyasuria P, Faust JM, Wilson JW, Mendelson CR. A decline in the levels of progesterone receptor coactivators in the pregnant uterus at term may antagonize progesterone receptor function and contribute to the initiation of parturition. Proc Natl Acad Sci USA 100: 9518–9523, 2003. doi: 10.1073/pnas.1633616100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Dong X, Shylnova O, Challis JR, Lye SJ. Identification and characterization of the protein-associated splicing factor as a negative co-regulator of the progesterone receptor. J Biol Chem 280: 13329–13340, 2005. doi: 10.1074/jbc.M409187200. [DOI] [PubMed] [Google Scholar]
  • 96. Henderson D, Wilson T. Reduced binding of progesterone receptor to its nuclear response element after human labor onset. Am J Obstet Gynecol 185: 579–585, 2001. doi: 10.1067/mob.2001.116753. [DOI] [PubMed] [Google Scholar]
  • 97. Chai SY, Smith R, Fitter JT, Mitchell C, Pan X, Ilicic M, Maiti K, Zakar T, Madsen G. Increased progesterone receptor A expression in labouring human myometrium is associated with decreased promoter occupancy by the histone demethylase JARID1A. Mol Hum Reprod 20: 442–453, 2014. doi: 10.1093/molehr/gau005. [DOI] [PubMed] [Google Scholar]
  • 98. Chai SY, Smith R, Zakar T, Mitchell C, Madsen G. Term myometrium is characterized by increased activating epigenetic modifications at the progesterone receptor-A promoter. Mol Hum Reprod 18: 401–409, 2012. doi: 10.1093/molehr/gas012. [DOI] [PubMed] [Google Scholar]
  • 99. Abdel-Hafiz HA, Horwitz KB. Post-translational modifications of the progesterone receptors. J Steroid Biochem Mol Biol 140: 80–89, 2014. doi: 10.1016/j.jsbmb.2013.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Khan JA, Amazit L, Bellance C, Guiochon-Mantel A, Lombès M, Loosfelt H. p38 and p42/44 MAPKs differentially regulate progesterone receptor A and B isoform stabilization. Mol Endocrinol 25: 1710–1724, 2011. doi: 10.1210/me.2011-1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Nadeem L, Shynlova O, Mesiano S, Lye S. Progesterone via its type-A receptor promotes myometrial gap junction coupling. Sci Rep 7: 13357, 2017. doi: 10.1038/s41598-017-13488-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Sugimoto Y, Segi E, Tsuboi K, Ichikawa A, Narumiya S. Female reproduction in mice lacking the prostaglandin F receptor. Roles of prostaglandin and oxytocin receptors in parturition. Adv Exp Med Biol 449: 317–321, 1998. doi: 10.1007/978-1-4615-4871-3_39. [DOI] [PubMed] [Google Scholar]
  • 103. Sugimoto Y, Yamasaki A, Segi E, Tsuboi K, Aze Y, Nishimura T, Oida H, Yoshida N, Tanaka T, Katsuyama M, Hasumoto K, Murata T, Hirata M, Ushikubi F, Negishi M, Ichikawa A, Narumiya S. Failure of parturition in mice lacking the prostaglandin F receptor. Science 277: 681–683, 1997. doi: 10.1126/science.277.5326.681. [DOI] [PubMed] [Google Scholar]
  • 104. Yellon SM, Ebner CA, Sugimoto Y. Parturition and recruitment of macrophages in cervix of mice lacking the prostaglandin F receptor. Biol Reprod 78: 438–444, 2008. doi: 10.1095/biolreprod.107.063404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Menon R, Bonney EA, Condon J, Mesiano S, Taylor RN. Novel concepts on pregnancy clocks and alarms: redundancy and synergy in human parturition. Hum Reprod Update 22: 535–560, 2016. doi: 10.1093/humupd/dmw022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Thorburn GD, Challis JR. Endocrine control of parturition. Physiol Rev 59: 863–918, 1979. doi: 10.1152/physrev.1979.59.4.863. [DOI] [PubMed] [Google Scholar]
  • 107. Thorburn GD. The placenta, prostaglandins and parturition: a review. Reprod Fertil Dev 3: 277–294, 1991. doi: 10.1071/rd9910277. [DOI] [PubMed] [Google Scholar]
  • 108. Honnebier WJ, Swaab DF. The influence of anencephaly upon intrauterine growth of fetus and placenta and upon gestation length. J Obstet Gynaecol Br Commonw 80: 577–588, 1973. doi: 10.1111/j.1471-0528.1973.tb16030.x. [DOI] [PubMed] [Google Scholar]
  • 109. Price HV, Cone BA, Keogh M. Length of gestation in congenital adrenal hyperplasia. J Obstet Gynaecol Br Commonw 78: 430–434, 1971. doi: 10.1111/j.1471-0528.1971.tb00297.x. [DOI] [PubMed] [Google Scholar]
  • 110. Walsh S, Kittinger G, Novy M. Maternal peripheral concentrations of estradiol, estrone, cortisol, and progesterone during late pregnancy in rhesus monkeys (Macaca mulatta) and after experimental fetal anencephaly and fetal death. Am J Obstet Gynecol 135: 37–42, 1979. [PubMed] [Google Scholar]
  • 111. Hirsch E, Muhle R. Intrauterine bacterial inoculation induces labor in the mouse by mechanisms other than progesterone withdrawal. Biol Reprod 67: 1337–1341, 2002. doi: 10.1095/biolreprod67.4.1337. [DOI] [PubMed] [Google Scholar]
  • 112. Romero R, Mazor M, Tartakovsky B. Systemic administration of interleukin-1 induces preterm parturition in mice. Am J Obstet Gynecol 165: 969–971, 1991. doi: 10.1016/0002-9378(91)90450-6. [DOI] [PubMed] [Google Scholar]
  • 113. Keirse MJ, Hicks BR, Mitchell MD, Turnbull AC. Increase of the prostaglandin precursor, arachidonic acid, in amniotic fluid during spontaneous labour. Br J Obstet Gynaecol 84: 937–940, 1977. doi: 10.1111/j.1471-0528.1977.tb12524.x. [DOI] [PubMed] [Google Scholar]
  • 114. Kredentser JV, Embree JE, McCoshen JA. Prostaglandin F2 alpha output by amnion-chorion-decidua: relationship with labor and prostaglandin E2 concentration at the amniotic surface. Am J Obstet Gynecol 173: 199–204, 1995. doi: 10.1016/0002-9378(95)90190-6. [DOI] [PubMed] [Google Scholar]
  • 115. Lee SE, Park IS, Romero R, Yoon BH. Amniotic fluid prostaglandin F2 increases even in sterile amniotic fluid and is an independent predictor of impending delivery in preterm premature rupture of membranes. J Matern Fetal Neonatal Med 22: 880–886, 2009. doi: 10.1080/14767050902994648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lee SE, Romero R, Park IS, Seong HS, Park CW, Yoon BH. Amniotic fluid prostaglandin concentrations increase before the onset of spontaneous labor at term. J Matern Fetal Neonatal Med 21: 89–94, 2008. doi: 10.1080/14767050701830514. [DOI] [PubMed] [Google Scholar]
  • 117. Mitchell MD, Romero RJ, Edwin SS, Trautman MS. Prostaglandins and parturition. Reprod Fertil Dev 7: 623–632, 1995. doi: 10.1071/rd9950623. [DOI] [PubMed] [Google Scholar]
  • 118. Jain JK, Mishell DR Jr.. A comparison of intravaginal misoprostol with prostaglandin E2 for termination of second-trimester pregnancy. N Engl J Med 331: 290–293, 1994. doi: 10.1056/NEJM199408043310502. [DOI] [PubMed] [Google Scholar]
  • 119. Robins J, Mann LI. Midtrimester pregnancy termination by intramuscular injection of a 15-methyl analogue of prostaglandin F2 alpha. Am J Obstet Gynecol 123: 625–631, 1975. doi: 10.1016/0002-9378(75)90886-8. [DOI] [PubMed] [Google Scholar]
  • 120. Madsen G, Zakar T, Ku CY, Sanborn BM, Smith R, Mesiano S. Prostaglandins differentially modulate progesterone receptor-A and -B expression in human myometrial cells: evidence for prostaglandin-induced functional progesterone withdrawal. J Clin Endocrinol Metab 89: 1010–1013, 2004. doi: 10.1210/jc.2003-031037. [DOI] [PubMed] [Google Scholar]
  • 121. Keelan JA. Intrauterine inflammatory activation, functional progesterone withdrawal, and the timing of term and preterm birth. J Reprod Immunol 125: 89–99, 2018. doi: 10.1016/j.jri.2017.12.004. [DOI] [PubMed] [Google Scholar]
  • 122. Andrews WW, Hauth JC, Goldenberg RL. Infection and preterm birth. Am J Perinatol 17: 357–365, 2000. doi: 10.1055/s-2000-13448. [DOI] [PubMed] [Google Scholar]
  • 123. Gibbs RS, Romero R, Hillier SL, Eschenbach DA, Sweet RL. A review of premature birth and subclinical infection. Am J Obstet Gynecol 166: 1515–1528, 1992. doi: 10.1016/0002-9378(92)91628-n. [DOI] [PubMed] [Google Scholar]
  • 124. Goldenberg RL, Culhane JF. Infection as a cause of preterm birth. Clin Perinatol 30: 677–700, 2003. doi: 10.1016/s0095-5108(03)00110-6. [DOI] [PubMed] [Google Scholar]
  • 125. Romero R, Espinoza J, Gonçalves LF, Kusanovic JP, Friel L, Hassan S. The role of inflammation and infection in preterm birth. Semin Reprod Med 25: 21–39, 2007. doi: 10.1055/s-2006-956773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Csapo A, Erdos T, De Mattos CR, Gramss E, Moscowitz C. Stretch-induced uterine growth, protein synthesis and function. Nature 207: 1378–1379, 1965. doi: 10.1038/2071378a0. [DOI] [PubMed] [Google Scholar]
  • 127. Hua R, Pease JE, Sooranna SR, Viney JM, Nelson SM, Myatt L, Bennett PR, Johnson MR. Stretch and inflammatory cytokines drive myometrial chemokine expression via NF-kappaB activation. Endocrinology 153: 481–491, 2012. doi: 10.1210/en.2011-1506. [DOI] [PubMed] [Google Scholar]
  • 128. Lee YH, Shynlova O, Lye SJ. Stretch-induced human myometrial cytokines enhance immune cell recruitment via endothelial activation. Cell Mol Immunol 12: 231–242, 2015. doi: 10.1038/cmi.2014.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Sooranna SR, Engineer N, Liang Z, Bennett PR, Johnson MR; Imperial College Parturition Research Group. Stretch and interleukin 1 beta: pro-labour factors with similar mitogen-activated protein kinase effects but differential patterns of transcription factor activation and gene expression. J Cell Physiol 212: 195–206, 2007. doi: 10.1002/jcp.21019. [DOI] [PubMed] [Google Scholar]
  • 130. Sooranna SR, Lee Y, Kim LU, Mohan AR, Bennett PR, Johnson MR. Mechanical stretch activates type 2 cyclooxygenase via activator protein-1 transcription factor in human myometrial cells. Mol Hum Reprod 10: 109–113, 2004. doi: 10.1093/molehr/gah021. [DOI] [PubMed] [Google Scholar]
  • 131. Cox LS, Redman C. The role of cellular senescence in ageing of the placenta. Placenta 52: 139–145, 2017. doi: 10.1016/j.placenta.2017.01.116. [DOI] [PubMed] [Google Scholar]
  • 132. Feng L, Allen TK, Marinello WP, Murtha AP. Roles of progesterone receptor membrane component 1 in oxidative stress-induced aging in chorion cells. Reprod Sci 26: 394–403, 2018. doi: 10.1177/1933719118776790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Maiti K, Sultana Z, Aitken RJ, Morris J, Park F, Andrew B, Riley SC, Smith R. Evidence that fetal death is associated with placental aging. Am J Obstet Gynecol 217: 441.e1–441.e14, 2017. doi: 10.1016/j.ajog.2017.06.015. [DOI] [PubMed] [Google Scholar]
  • 134. Manna S, McCarthy C, McCarthy FP. Placental ageing in adverse pregnancy outcomes: telomere shortening, cell senescence, and mitochondrial dysfunction. Oxid Med Cell Longev 2019: 3095383, 2019. doi: 10.1155/2019/3095383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Menon R, Moore JJ. Fetal membranes, not a mere appendage of the placenta, but a critical part of the fetal-maternal interface controlling parturition. Obstet Gynecol Clin North Am 47: 147–162, 2020. doi: 10.1016/j.ogc.2019.10.004. [DOI] [PubMed] [Google Scholar]
  • 136. Sultana Z, Maiti K, Aitken J, Morris J, Dedman L, Smith R. Oxidative stress, placental ageing-related pathologies and adverse pregnancy outcomes. Am J Reprod Immunol 77: 5, 2017. doi: 10.1111/aji.12653. [DOI] [PubMed] [Google Scholar]
  • 137. Padula AM, Monk C, Brennan PA, Borders A, Barrett ES, McEvoy CT, Foss S, Desai P, Alshawabkeh A, Wurth R, Salafia C, Fichorova R, Varshavsky J, Kress A, Woodruff TJ, Morello-Frosch R. A review of maternal prenatal exposures to environmental chemicals and psychosocial stressors-implications for research on perinatal outcomes in the ECHO program. J Perinatol 40: 10–24, 2020. doi: 10.1038/s41372-019-0510-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Eick SM, Goin DE, Cushing L, DeMicco E, Smith S, Park JS, Padula AM, Woodruff TJ, Morello-Frosch R. Joint effects of prenatal exposure to per- and poly-fluoroalkyl substances and psychosocial stressors on corticotropin-releasing hormone during pregnancy. J Expo Sci Environ Epidemiol 32: 27–36, 2022. doi: 10.1038/s41370-021-00322-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med 342: 1500–1507, 2000. doi: 10.1056/NEJM200005183422007. [DOI] [PubMed] [Google Scholar]
  • 140. Fettweis JM, Serrano MG, Brooks JP, Edwards DJ, Girerd PH, Parikh HI, , et al. The vaginal microbiome and preterm birth. Nat Med 25: 1012–1021, 2019. doi: 10.1038/s41591-019-0450-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Gudnadottir U, Debelius JW, Du J, Hugerth LW, Danielsson H, Schuppe-Koistinen I, Fransson E, Brusselaers N. The vaginal microbiome and the risk of preterm birth: a systematic review and network meta-analysis. Sci Rep 12: 7926, 2022. doi: 10.1038/s41598-022-12007-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Siewiera J, McIntyre TI, Cautivo KM, Mahiddine K, Rideaux D, Molofsky AB, Erlebacher A. Circumvention of luteolysis reveals parturition pathways in mice dependent upon innate type 2 immunity. Immunity 56: 606–619.e7, 2023. doi: 10.1016/j.immuni.2023.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Condon JC, Jeyasuria P, Faust JM, Mendelson CR. Surfactant protein secreted by the maturing mouse fetal lung acts as a hormone that signals the initiation of parturition. Proc Natl Acad Sci USA 101: 4978–4983, 2004. doi: 10.1073/pnas.0401124101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Kim CJ, Kim JS, Kim YM, Cushenberry E, Richani K, Espinoza J, Romero R. Fetal macrophages are not present in the myometrium of women with labor at term. Am J Obstet Gynecol 195: 829–833, 2006. doi: 10.1016/j.ajog.2006.06.052. [DOI] [PubMed] [Google Scholar]
  • 145. Bayar E, Bennett PR, Chan D, Sykes L, MacIntyre DA. The pregnancy microbiome and preterm birth. Semin Immunopathol 42: 487–499, 2020. doi: 10.1007/s00281-020-00817-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Leo AP, Oskotsky TT, Oskotsky B, Wibrand C, Roldan A, Tang A, Ha CW, Wong RJ, Minot SS, Andreoletti G, Kosti I, Theis KR, Ng S, Lee YS, Diaz-Gimeno P, Bennett PR, MacIntyre DA, Lynch SV, Romero R, Tarca AL, Stevenson DK, Aghaeepour N, Golob J, Sirota M. VMAP: Vaginal Microbiome Atlas During Pregnancy (Preprint). medRxiv 2023.03.21.23286947, 2023. doi: 10.1101/2023.03.21.23286947. [DOI] [PMC free article] [PubMed]
  • 147. Liao J, Shenhav L, Urban JA, Serrano M, Zhu B, Buck GA, Korem T. Microdiversity of the vaginal microbiome is associated with preterm birth (Preprint). bioRxiv 2023.01.13.523991, 2023. doi: 10.1101/2023.01.13.523991. [DOI] [PMC free article] [PubMed]
  • 148. Pruski P, Correia GD, Lewis HV, Capuccini K, Inglese P, Chan D, Brown RG, Kindinger L, Lee YS, Smith A, Marchesi J, McDonald JA, Cameron S, Alexander-Hardiman K, David AL, Stock SJ, Norman JE, Terzidou V, Teoh TG, Sykes L, Bennett PR, Takats Z, MacIntyre DA. Direct on-swab metabolic profiling of vaginal microbiome host interactions during pregnancy and preterm birth. Nat Commun 12: 5967, 2021. doi: 10.1038/s41467-021-26215-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Stout MJ, Zhou Y, Wylie KM, Tarr PI, Macones GA, Tuuli MG. Early pregnancy vaginal microbiome trends and preterm birth. Am J Obstet Gynecol 217: 356.e1–356.e18, 2017. doi: 10.1016/j.ajog.2017.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Chan D, Bennett PR, Lee YS, Kundu S, Teoh TG, Adan M, Ahmed S, Brown RG, David AL, Lewis HV, Gimeno-Molina B, Norman JE, Stock SJ, Terzidou V, Kropf P, Botto M, MacIntyre DA, Sykes L. Microbial-driven preterm labour involves crosstalk between the innate and adaptive immune response. Nat Commun 13: 975, 2022. doi: 10.1038/s41467-022-28620-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Pepe GJ, Albrecht ED. Actions of placental and fetal adrenal steroid hormones in primate pregnancy. Endocr Rev 16: 608–648, 1995. doi: 10.1210/edrv-16-5-608. [DOI] [PubMed] [Google Scholar]
  • 152. Rajabi M, Solomon S, Poole AR. Hormonal regulation of interstitial collagenase in the uterine cervix of the pregnant guinea pig. Endocrinology 128: 863–871, 1991. doi: 10.1210/endo-128-2-863. [DOI] [PubMed] [Google Scholar]
  • 153. Rajabi MR, Dodge GR, Solomon S, Poole AR. Immunochemical and immunohistochemical evidence of estrogen-mediated collagenolysis as a mechanism of cervical dilatation in the guinea pig at parturition. Endocrinology 128: 371–378, 1991. doi: 10.1210/endo-128-1-371. [DOI] [PubMed] [Google Scholar]
  • 154. Nissenson R, Fluoret G, Hechter O. Opposing effects of estradiol and progesterone on oxytocin receptors in rabbit uterus. Proc Natl Acad Sci USA 75: 2044–2048, 1978. doi: 10.1073/pnas.75.4.2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Pinto RM, Lerner U, Glauberman M, Pontelli H. Influence of estradiol-17-beta upon the oxytocic action of oxytocin in the pregnant human uterus. Am J Obstet Gynecol 96: 857–862, 1966. doi: 10.1016/0002-9378(66)90682-x. [DOI] [PubMed] [Google Scholar]
  • 156. Pinto RM, Lerner U, Pontelli H, Rabow W. Effect of estradiol-17-beta on oxytocin-induced contraction of the three separate layers of human pregnant myometrium. Am J Obstet Gynecol 97: 881–887, 1967. doi: 10.1016/0002-9378(67)90511-x. [DOI] [PubMed] [Google Scholar]
  • 157. Kilarski WM, Fu X, Bäckström T, Roomans GM, Ulmsten U. Progesterone, oestradiol and oxytocin and their in vitro effect on maintaining the number of gap junction plaques in human myometrium at term. Acta Physiol Scand 157: 461–469, 1996. doi: 10.1046/j.1365-201X.1996.500247000.x. [DOI] [PubMed] [Google Scholar]
  • 158. Kilarski WM, Hongpaisan J, Semik D, Roomans GM. Effect of progesterone and oestradiol on expression of connexin43 in cultured human myometrium cells. Folia Histochem Cytobiol 38: 3–9, 2000. [PubMed] [Google Scholar]
  • 159. Lye SJ, Nicholson BJ, Mascarenhas M, MacKenzie L, Petrocelli T. Increased expression of connexin-43 in the rat myometrium during labor is associated with an increase in the plasma estrogen:progesterone ratio. Endocrinology 132: 2380–2386, 1993. doi: 10.1210/endo.132.6.8389279. [DOI] [PubMed] [Google Scholar]
  • 160. Petrocelli T, Lye SJ. Regulation of transcripts encoding the myometrial gap junction protein, connexin-43, by estrogen and progesterone. Endocrinology 133: 284–290, 1993. doi: 10.1210/endo.133.1.8391423. [DOI] [PubMed] [Google Scholar]
  • 161. Bolte E, Mancuso S, Eriksson G, Wiqvist N, Diczfalusy E. Studies on the aromatization of neutral steroids in pregnant women: I. Aromatization of C-19 steroids by placenta perfused in situ. Acta Endocrinol (Copenh) 45: 535–559, 1964. doi: 10.1530/acta.0.0450535. [DOI] [PubMed] [Google Scholar]
  • 162. Siiteri P, MacDonald P. The utilization of circulating dehydroepiandrosterone sulfate for estrogen synthesis during human pregnancy. Steroids 2: 713–730, 1963. doi: 10.1016/0039-128X(63)90044-8. [DOI] [Google Scholar]
  • 163. Siiteri PK, MacDonald PC. Placental estrogen biosynthesis during human pregnancy. J Clin Endocrinol Metab 26: 751–761, 1966. doi: 10.1210/jcem-26-7-751. [DOI] [PubMed] [Google Scholar]
  • 164. Pasqualini JR, Kincl F. Hormones and the Fetus. Oxford: Pergamin Press, 1985. [Google Scholar]
  • 165. Harada N. Genetic analysis of human placental aromatase deficiency. J Steroid Biochem Mol Biol 44: 331–340, 1993. doi: 10.1016/0960-0760(93)90236-p. [DOI] [PubMed] [Google Scholar]
  • 166. Mullis PE, Yoshimura N, Kuhlmann B, Lippuner K, Jaeger P, Harada H. Aromatase deficiency in a female who is compound heterozygote for two new point mutations in the P450arom gene: impact of estrogens on hypergonadotropic hypogonadism, multicystic ovaries, and bone densitometry in childhood. J Clin Endocrinol Metab 82: 1739–1745, 1997. doi: 10.1210/jcem.82.6.3994. [DOI] [PubMed] [Google Scholar]
  • 167. Shozu M, Akasofu K, Harada T, Kubota Y. A new cause of female pseudohermaphroditism: placental aromatase deficiency. J Clin Endocrinol Metab 72: 560–566, 1991. doi: 10.1210/jcem-72-3-560. [DOI] [PubMed] [Google Scholar]
  • 168. Diczfalusy E. Endocrine functions of the human fetoplacental unit. Fed Proc 23: 791–798, 1964. [PubMed] [Google Scholar]
  • 169. Frandsen VA, Stakemann G. The site of production of oestrogenic hormones in human pregnancy. Hormone excretion in pregnancy with anencephalic foetus. Acta Endocrinol (Copenh) 38: 383–391, 1961. doi: 10.1530/acta.0.0380383. [DOI] [PubMed] [Google Scholar]
  • 170. France J. Steroid sulfatase deficiency. J Steroid Biochem 11: 647–651, 1979. doi: 10.1016/0022-4731(79)90094-3. [DOI] [PubMed] [Google Scholar]
  • 171. France JT, Liggins GC. Placental sulfatase deficiency. J Clin Endocrinol Metab 29: 138–141, 1969. doi: 10.1210/jcem-29-1-138. [DOI] [PubMed] [Google Scholar]
  • 172. France JT, Seddon RJ, Liggins GC. A study of pregnancy with low estrogen production due to placental sulfatase deficiency. J Clin Endocrinol Metab 36: 1–9, 1973. doi: 10.1210/jcem-36-1-1. [DOI] [PubMed] [Google Scholar]
  • 173. Bose HS, Sugawara T, Strauss JF, Miller WL; International Congenital Lipoid Adrenal Hyperplasia Consortium. The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. N Engl J Med 335: 1870–1878, 1996. doi: 10.1056/NEJM199612193352503. [DOI] [PubMed] [Google Scholar]
  • 174. Saenger P, Klonari Z, Black SM, Compagnone N, Mellon SH, Fleischer A, Abrams CA, Shackelton CH, Miller WL. Prenatal diagnosis of congenital lipoid adrenal hyperplasia. J Clin Endocrinol Metab 80: 200–205, 1995. doi: 10.1210/jcem.80.1.7829612. [DOI] [PubMed] [Google Scholar]
  • 175. Blanks AM, Thornton S. The role of oxytocin in parturition. BJOG 110, Suppl 20: 46–51, 2003. doi: 10.1016/s1470-0328(03)00024-7. [DOI] [PubMed] [Google Scholar]
  • 176. Chibbar R, Wong S, Miller FD, Mitchell BF. Estrogen stimulates oxytocin gene expression in human chorio-decidua. J Clin Endocrinol Metab 80: 567–572, 1995. doi: 10.1210/jcem.80.2.7852522. [DOI] [PubMed] [Google Scholar]
  • 177. Mitchell BF, Chibbar R. Synthesis and metabolism of oxytocin in late gestation in human decidua. Adv Exp Med Biol 395: 365–380, 1995. [PubMed] [Google Scholar]
  • 178. Wilson T, Liggins GC, Whittaker DJ. Oxytocin stimulates the release of arachidonic acid and prostaglandin F2 alpha from human decidual cells. Prostaglandins 35: 771–780, 1988. doi: 10.1016/0090-6980(88)90149-9. [DOI] [PubMed] [Google Scholar]
  • 179. Fuchs AR, Fuchs F, Husslein P, Soloff MS. Oxytocin receptors in the human uterus during pregnancy and parturition. Am J Obstet Gynecol 150: 734–741, 1984. doi: 10.1016/0002-9378(84)90677-x. [DOI] [PubMed] [Google Scholar]
  • 180. 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 215: 1396–1398, 1982. doi: 10.1126/science.6278592. [DOI] [PubMed] [Google Scholar]
  • 181. Fuchs AR, Periyasamy S, Alexandrova M, Soloff MS. Correlation between oxytocin receptor concentration and responsiveness to oxytocin in pregnant rat myometrium: effects of ovarian steroids. Endocrinology 113: 742–749, 1983. doi: 10.1210/endo-113-2-742. [DOI] [PubMed] [Google Scholar]
  • 182. Mitchell BF, Schmid B. Oxytocin and its receptor in the process of parturition. J Soc Gynecol Investig 8: 122–133, 2001. [PubMed] [Google Scholar]
  • 183. 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 Endocrinol (Copenh) 114: 458–464, 1987. doi: 10.1530/acta.0.1140458. [DOI] [PubMed] [Google Scholar]
  • 184. Mitchell BF, Wong S. Metabolism of oxytocin in human decidua, chorion, and placenta. J Clin Endocrinol Metab 80: 2729–2733, 1995. doi: 10.1210/jcem.80.9.7673416. [DOI] [PubMed] [Google Scholar]
  • 185. Russell JA, Leng G. Sex, parturition and motherhood without oxytocin? J Endocrinol 157: 343–359, 1998. doi: 10.1677/joe.0.1570343. [DOI] [PubMed] [Google Scholar]
  • 186. Giraldi A, Enevoldsen AS, Wagner G. Oxytocin and the initiation of parturition. A review. Dan Med Bull 37: 377–383, 1990. [PubMed] [Google Scholar]
  • 187. Chard T. Fetal and maternal oxytocin in human parturition. Am J Perinatol 6: 145–152, 1989. doi: 10.1055/s-2007-999566. [DOI] [PubMed] [Google Scholar]
  • 188. Gross GA, Imamura T, Luedke C, Vogt SK, Olson LM, Nelson DM, Sadovsky Y, Muglia LJ. Opposing actions of prostaglandins and oxytocin determine the onset of murine labor. Proc Natl Acad Sci USA 95: 11875–11879, 1998. doi: 10.1073/pnas.95.20.11875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Renfree MB, Parry LJ, Shaw G. Infusion with an oxytocin receptor antagonist delays parturition in a marsupial. J Reprod Fertil 108: 131–137, 1996. doi: 10.1530/jrf.0.1080131. [DOI] [PubMed] [Google Scholar]
  • 190. Schellenberg JC. The effect of oxytocin receptor blockade on parturition in guinea pigs. J Clin Invest 95: 13–19, 1995. doi: 10.1172/JCI117629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. 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 93: 11699–11704, 1996. doi: 10.1073/pnas.93.21.11699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. 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. J Neuroendocrinol 8: 847–853, 1996. doi: 10.1046/j.1365-2826.1996.05266.x. [DOI] [PubMed] [Google Scholar]
  • 193. Olson DM. The role of prostaglandins in the initiation of parturition. Best Pract Res Clin Obstet Gynaecol 17: 717–730, 2003. doi: 10.1016/s1521-6934(03)00069-5. [DOI] [PubMed] [Google Scholar]
  • 194. Olson DM, Ammann C. Role of the prostaglandins in labour and prostaglandin receptor inhibitors in the prevention of preterm labour. Front Biosci 12: 1329–1343, 2007. doi: 10.2741/2151. [DOI] [PubMed] [Google Scholar]
  • 195. Patel FA, Challis JR. Prostaglandins and uterine activity. Front Horm Res 27: 31–56, 2001. doi: 10.1159/000061040. [DOI] [PubMed] [Google Scholar]
  • 196. Winchester SK, Imamura T, Gross GA, Muglia LM, Vogt SK, Wright J, Watanabe K, Tai HH, Muglia LJ. Coordinate regulation of prostaglandin metabolism for induction of parturition in mice. Endocrinology 143: 2593–2598, 2002. doi: 10.1210/endo.143.7.8926. [DOI] [PubMed] [Google Scholar]
  • 197. Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 69: 145–182, 2000. doi: 10.1146/annurev.biochem.69.1.145. [DOI] [PubMed] [Google Scholar]
  • 198. Tai HH, Ensor CM, Zhou H, Yan F. Structure and function of human NAD+-linked 15-hydroxyprostaglandin dehydrogenase. Adv Exp Med Biol 507: 245–250, 2002. doi: 10.1007/978-1-4615-0193-0_37. [DOI] [PubMed] [Google Scholar]
  • 199. van Meir CA, Matthews SG, Keirse MJ, Ramirez MM, Bocking A, Challis JR. 15-Hydroxyprostaglandin dehydrogenase: implications in preterm labor with and without ascending infection. J Clin Endocrinol Metab 82: 969–976, 1997. doi: 10.1210/jcem.82.3.3812. [DOI] [PubMed] [Google Scholar]
  • 200. Van Meir CA, Ramirez MM, Matthews SG, Calder AA, Keirse MJ, Challis JR. Chorionic prostaglandin catabolism is decreased in the lower uterine segment with term labour. Placenta 18: 109–114, 1997. doi: 10.1016/s0143-4004(97)90081-3. [DOI] [PubMed] [Google Scholar]
  • 201. Romero R, Munoz H, Gomez R, Parra M, Polanco M, Valverde V, Hasbun J, Garrido J, Ghezzi F, Mazor M, Tolosa JE, Mitchell MD. Increase in prostaglandin bioavailability precedes the onset of human parturition. Prostaglandins Leukot Essent Fatty Acids 54: 187–191, 1996. doi: 10.1016/s0952-3278(96)90015-0. [DOI] [PubMed] [Google Scholar]
  • 202. Gibb W, Sun M. Localization of prostaglandin H synthase type 2 protein and mRNA in term human fetal membranes and decidua. J Endocrinol 150: 497–503, 1996. doi: 10.1677/joe.0.1500497. [DOI] [PubMed] [Google Scholar]
  • 203. Hirst JJ, Teixeira FJ, Zakar T, Olson DM. Prostaglandin H synthase-2 expression increases in human gestational tissues with spontaneous labour onset. Reprod Fertil Dev 7: 633–637, 1995. doi: 10.1071/rd9950633. [DOI] [PubMed] [Google Scholar]
  • 204. Slater DM, Berger LC, Newton R, Moore GE, Bennett PR. Expression of cyclooxygenase types 1 and 2 in human fetal membranes at term. Am J Obstet Gynecol 172: 77–82, 1995. doi: 10.1016/0002-9378(95)90087-x. [DOI] [PubMed] [Google Scholar]
  • 205. Grigsby PL, Sooranna SR, Adu-Amankwa B, Pitzer B, Brockman DE, Johnson MR, Myatt L. Regional expression of prostaglandin E2 and F2alpha receptors in human myometrium, amnion, and choriodecidua with advancing gestation and labor. Biol Reprod 75: 297–305, 2006. doi: 10.1095/biolreprod.106.051987. [DOI] [PubMed] [Google Scholar]
  • 206. Myatt L, Lye SJ. Expression, localization and function of prostaglandin receptors in myometrium. Prostaglandins Leukot Essent Fatty Acids 70: 137–148, 2004. doi: 10.1016/j.plefa.2003.04.004. [DOI] [PubMed] [Google Scholar]
  • 207. Vannuccini S, Bocchi C, Severi FM, Challis JR, Petraglia F. Endocrinology of human parturition. Ann Endocrinol (Paris) 77: 105–113, 2016. doi: 10.1016/j.ando.2016.04.025. [DOI] [PubMed] [Google Scholar]
  • 208. Farfán-Labonne B, Leff-Gelman P, Pellón-Díaz G, Camacho-Arroyo I. Cellular senescence in normal and adverse pregnancy. Reprod Biol 23: 100734, 2023. doi: 10.1016/j.repbio.2023.100734. [DOI] [PubMed] [Google Scholar]
  • 209. Leung T, Chung T, Madsen G, Lam P, Sahota D, Smith R. Rate of rise in maternal plasma corticotrophin-releasing hormone and its relation to gestational length. BJOG 108: 527–532, 2001. doi: 10.1111/j.1471-0528.2001.00112.x. [DOI] [PubMed] [Google Scholar]
  • 210. McLean M, Bisits A, Davies J, Woods R, Lowry P, Smith R. A placental clock controlling the length of human pregnancy. Nat Med 1: 460–463, 1995. doi: 10.1038/nm0595-460. [DOI] [PubMed] [Google Scholar]
  • 211. McLean M, Bisits A, Davies J, Walters W, Hackshaw A, De Voss K, Smith R. Predicting risk of preterm delivery by second-trimester measurement of maternal plasma corticotropin-releasing hormone and alpha-fetoprotein concentrations. Am J Obstet Gynecol 181: 207–215, 1999. doi: 10.1016/s0002-9378(99)70461-8. [DOI] [PubMed] [Google Scholar]
  • 212. Smith R, Mesiano S, Chan EC, Brown S, Jaffe RB. Corticotropin-releasing hormone directly and preferentially stimulates dehydroepiandrosterone sulfate secretion by human fetal adrenal cortical cells. J Clin Endocrinol Metab 83: 2916–2920, 1998. doi: 10.1210/jcem.83.8.5020. [DOI] [PubMed] [Google Scholar]
  • 213. Sirianni R, Rehman KS, Carr BR, Parker CR Jr, Rainey WE. Corticotropin-releasing hormone directly stimulates cortisol and the cortisol biosynthetic pathway in human fetal adrenal cells. J Clin Endocrinol Metab 90: 279–285, 2005. doi: 10.1210/jc.2004-0865. [DOI] [PubMed] [Google Scholar]
  • 214. Thomson M, Smith R. The action of hypothalamic and placental corticotropin releasing factor on the corticotrope. Mol Cell Endocrinol 62: 1–12, 1989. doi: 10.1016/0303-7207(89)90107-x. [DOI] [PubMed] [Google Scholar]
  • 215. Robertson R (editor). DeGroot’s Endocrinology (8th ed.). Amsterdam: Elsevier, 2022. [Google Scholar]
  • 216. Plunkett J, Doniger S, Orabona G, Morgan T, Haataja R, Hallman M, Puttonen H, Menon R, Kuczynski E, Norwitz E, Snegovskikh V, Palotie A, Peltonen L, Fellman V, DeFranco EA, Chaudhari BP, McGregor TL, McElroy JJ, Oetjens MT, Teramo K, Borecki I, Fay J, Muglia L. An evolutionary genomic approach to identify genes involved in human birth timing. PLoS Genet 7: e1001365, 2011. doi: 10.1371/journal.pgen.1001365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Rosenberg KR, Trevathan WR. The evolution of human birth. Sci Am 285: 72–77, 2001. doi: 10.1038/scientificamerican1101-72. [DOI] [PubMed] [Google Scholar]
  • 218. Trevathan WR. Human Birth. an Evolutionary Perspective. Hawthorne, NY: Aldine De Gryter, 1987. [Google Scholar]
  • 219. Rightmire GP. Brain size and encephalization in early to mid-Pleistocene Homo. Am J Phys Anthropol 124: 109–123, 2004. doi: 10.1002/ajpa.10346. [DOI] [PubMed] [Google Scholar]
  • 220. Rosenberg K, Trevathan W. Birth, obstetrics and human evolution. BJOG 109: 1199–1206, 2002. doi: 10.1046/j.1471-0528.2002.00010.x. [DOI] [PubMed] [Google Scholar]
  • 221. Dunsworth HM, Warrener AG, Deacon T, Ellison PT, Pontzer H. Metabolic hypothesis for human altriciality. Proc Natl Acad Sci USA 109: 15212–15216, 2012. doi: 10.1073/pnas.1205282109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Li J, Hong X, Mesiano S, Muglia LJ, Wang X, Snyder M, Stevenson DK, Shaw GM. Natural selection has differentiated the progesterone receptor among human populations. Am J Hum Genet 103: 45–57, 2018. doi: 10.1016/j.ajhg.2018.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Zeberg H, Kelso J, Paabo S. The Neandertal progesterone receptor. Mol Biol Evol 37: 2655–2660, 2020. doi: 10.1093/molbev/msaa119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.American College of Obstetricians and Gynecologists’ Committee on Obstetric Practice, Society for Maternal-Fetal Medicine. Medically Indicated Late-Preterm and Early-Term Deliveries: ACOG Committee Opinion, Number 831. Obstet Gynecol 138: e35–e39, 2021. doi: 10.1097/AOG.0000000000004447. [DOI] [PubMed] [Google Scholar]
  • 225. Lawn JE, Ohuma EO, Bradley E, Idueta LS, Hazel E, Okwaraji YB, Erchick DJ, Yargawa J, Katz J, Lee ACC, Diaz M, Salasibew M, Requejo J, Hayashi C, Moller AB, Borghi E, Black RE, Blencowe H; Lancet Small Vulnerable Newborn Steering Committee, WHO/UNICEF Preterm Birth Estimates Group, National Vulnerable Newborn Measurement Group, Subnational Vulnerable Newborn Measurement Group. Small babies, big risks: global estimates of prevalence and mortality for vulnerable newborns to accelerate change and improve counting. Lancet 401: 1707–1719, 2023. doi: 10.1016/S0140-6736(23)00522-6. [DOI] [PubMed] [Google Scholar]
  • 226. Allen R, O’Brien BM. Uses of misoprostol in obstetrics and gynecology. Rev Obstet Gynecol 2: 159–168, 2009. [PMC free article] [PubMed] [Google Scholar]
  • 227. Baxi LV, Petrie RH, Caritis SN. Induction of labor with low-dose prostaglandin F2 alpha and oxytocin. Am J Obstet Gynecol 136: 28–31, 1980. doi: 10.1016/0002-9378(80)90559-1. [DOI] [PubMed] [Google Scholar]
  • 228. Mozurkewich EL, Chilimigras JL, Berman DR, Perni UC, Romero VC, King VJ, Keeton KL. Methods of induction of labour: a systematic review. BMC Pregnancy Childbirth 11: 84, 2011. doi: 10.1186/1471-2393-11-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Shyken JM, Petrie RH. Oxytocin to induce labor. Clin Obstet Gynecol 38: 232–245, 1995. doi: 10.1097/00003081-199506000-00006. [DOI] [PubMed] [Google Scholar]
  • 230. Xi M, Gerriets V. Prostaglandin E2 (Dinoprostone). Treasure Island, FL: StatPearls, 2023. [PubMed] [Google Scholar]
  • 231. Andrikopoulou M, Bushman ET, Rice MM, Grobman WA, Reddy UM, Silver RM, El-Sayed YY, Rouse DJ, Saade GR, Thorp JM Jr, Chauhan SP, Costantine MM, Chien EK, Casey BM, Srinivas SK, Swamy GK, Simhan HN, Eunice Kennedy Shriver National Institute of Child Health Human Development Maternal-Fetal Medicine Units Network. Maternal and neonatal outcomes in nulliparous participants undergoing labor induction by cervical ripening method. Am J Perinatol 40: 1061–1070, 2023. doi: 10.1055/s-0041-1732379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Diedrich JT, Drey EA, Newmann SJ. Society of Family Planning clinical recommendations: cervical preparation for dilation and evacuation at 20-24 weeks’ gestation. Contraception 101: 286–292, 2020. doi: 10.1016/j.contraception.2020.01.002. [DOI] [PubMed] [Google Scholar]
  • 233. Ralph JA, Shulman LP. Adjunctive agents for cervical preparation in second trimester surgical abortion. Adv Ther 36: 1246–1251, 2019. doi: 10.1007/s12325-019-00953-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Dudley DJ, Branch DW, Edwin SS, Mitchell MD. Induction of preterm birth in mice by RU486. Biol Reprod 55: 992–995, 1996. doi: 10.1095/biolreprod55.5.992. [DOI] [PubMed] [Google Scholar]
  • 235. Hapangama D, Neilson JP. Mifepristone for induction of labour. Cochrane Database Syst Rev 2009: CD002865, 2009. doi: 10.1002/14651858.CD002865.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Hcini N, Jolivet A, Pomar L, McHirgui A, Maamri F, Elcadhi Y, Lambert V, Carles G. Cervical maturation using mifepristone in women with normal pregnancies at or beyond term. Eur J Obstet Gynecol Reprod Biol 248: 58–62, 2020. doi: 10.1016/j.ejogrb.2020.03.020. [DOI] [PubMed] [Google Scholar]
  • 237. Hagey JM, Givens M, Bryant AG. Clinical update on uses for mifepristone in obstetrics and gynecology. Obstet Gynecol Surv 77: 611–623, 2022. doi: 10.1097/OGX.0000000000001063. [DOI] [PubMed] [Google Scholar]
  • 238. de Vaan MD, Ten Eikelder ML, Jozwiak M, Palmer KR, Davies-Tuck M, Bloemenkamp KW, Mol BW, Boulvain M. Mechanical methods for induction of labour. Cochrane Database Syst Rev 3: CD001233, 2023. doi: 10.1002/14651858.CD001233.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Keirse MJ, Thiery M, Parewijck W, Mitchell MD. Chronic stimulation of uterine prostaglandin synthesis during cervical ripening before the onset of labor. Prostaglandins 25: 671–682, 1983. doi: 10.1016/0090-6980(83)90121-1. [DOI] [PubMed] [Google Scholar]
  • 240. Grobman WA, Rice MM, Reddy UM, Tita AT, Silver RM, Mallett G, Hill K, Thom EA, El-Sayed YY, Perez-Delboy A, Rouse DJ, Saade GR, Boggess KA, Chauhan SP, Iams JD, Chien EK, Casey BM, Gibbs RS, Srinivas SK, Swamy GK, Simhan HN, Macones GA; Eunice Kennedy Shriver National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Labor induction versus expectant management in low-risk nulliparous women. N Engl J Med 379: 513–523, 2018. doi: 10.1056/NEJMoa1800566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Ayala NK, Rouse DJ. Failed induction of labor. Am J Obstet Gynecol 230: S769–S774, 2024. doi: 10.1016/j.ajog.2021.06.103. [DOI] [PubMed] [Google Scholar]
  • 242. Mensah NA, Fassett MJ, Shi JM, Kawatkar AA, Xie F, Chiu VY, Yeh M, Avila CC, Khadka N, Sacks DA, Getahun D. Examining recent trends in spontaneous and iatrogenic preterm birth across race and ethnicity in a large managed care population. Am J Obstet Gynecol 228: 736.e1–736.e15, 2023. doi: 10.1016/j.ajog.2022.11.1288. [DOI] [PubMed] [Google Scholar]
  • 243. Jiang Y, Wang L, Shen H, Wang B, Wu J, Hu K, Wang Y, Ma B, Zhang X. The effect of progesterone supplementation for luteal phase support in natural cycle frozen embryo transfer: a systematic review and meta-analysis based on randomized controlled trials. Fertil Steril 119: 597–605, 2023. doi: 10.1016/j.fertnstert.2022.12.035. [DOI] [PubMed] [Google Scholar]
  • 244.Practice Committee of the American Society for Reproductive Medicine. Progesterone supplementation during the luteal phase and in early pregnancy in the treatment of infertility: an educational bulletin. Fertil Steril 89: 789–792, 2008. doi: 10.1016/j.fertnstert.2008.02.012. [DOI] [PubMed] [Google Scholar]
  • 245. Mesiano S. Progesterone—historical perspective. J Steroid Biochem Mol Biol 223: 106157, 2022. doi: 10.1016/j.jsbmb.2022.106157. [DOI] [PubMed] [Google Scholar]
  • 246. Mesiano SA, Peters GA, Amini P, Wilson RA, Tochtrop GP, van Den Akker F. Progestin therapy to prevent preterm birth: history and effectiveness of current strategies and development of novel approaches. Placenta 79: 46–52, 2019., doi: 10.1016/j.placenta.2019.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Weatherborn M, Mesiano S. Rationale for current and future progestin-based therapies to prevent preterm birth. Best Pract Res Clin Obstet Gynaecol 52: 114–125, 2018. doi: 10.1016/j.bpobgyn.2018.03.008. [DOI] [PubMed] [Google Scholar]
  • 248. Hendricks CH, Brenner WE, Gabel RA, Kerenyi T. The effect of progesterone administered intra-amniotically in late human pregnancy. In: Brook Lodge Symposium: Progesterone. Augusta, MI: Upjohn Company, 1961, p. 53–64. [Google Scholar]
  • 249. Keirse MJ. Progestogen administration in pregnancy may prevent preterm delivery. Br J Obstet Gynaecol 97: 149–154, 1990. doi: 10.1111/j.1471-0528.1990.tb01740.x. [DOI] [PubMed] [Google Scholar]
  • 250. Meis PJ, Klebanoff M, Thom E, Dombrowski MP, Sibai B, Moawad AH, Spong CY, Hauth JC, Miodovnik M, Varner MW, Leveno KJ, Caritis SN, Iams JD, Wapner RJ, Conway D, O’Sullivan MJ, Carpenter M, Mercer B, Ramin SM, Thorp JM, Peaceman AM, Gabbe S; National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Prevention of recurrent preterm delivery by 17 alpha-hydroxyprogesterone caproate. N Engl J Med 348: 2379–2385, 2003. doi: 10.1056/NEJMoa035140. [DOI] [PubMed] [Google Scholar]
  • 251. da Fonseca EB, Bittar RE, Carvalho MH, Zugaib M. Prophylactic administration of progesterone by vaginal suppository to reduce the incidence of spontaneous preterm birth in women at increased risk: a randomized placebo-controlled double-blind study. Am J Obstet Gynecol 188: 419–424, 2003. doi: 10.1067/mob.2003.41. [DOI] [PubMed] [Google Scholar]
  • 252. Blackwell SC, Gyamfi-Bannerman C, Biggio JR Jr, Chauhan SP, Hughes BL, Louis JM, Manuck TA, Miller HS, Das AF, Saade GR, Nielsen P, Baker J, Yuzko OM, Reznichenko GI, Reznichenko NY, Pekarev O, Tatarova N, Gudeman J, Birch R, Jozwiakowski MJ, Duncan M, Williams L, Krop J. 17-OHPC to prevent recurrent preterm birth in singleton gestations (PROLONG Study): a multicenter, international, randomized double-blind trial. Am J Perinatol 37: 127–136, 2020. doi: 10.1055/s-0039-3400227. [DOI] [PubMed] [Google Scholar]
  • 253. Norman JE, Marlow N, Messow CM, Shennan A, Bennett PR, Thornton S, Robson SC, McConnachie A, Petrou S, Sebire NJ, Lavender T, Whyte S, Norrie J. Does progesterone prophylaxis to prevent preterm labour improve outcome? A randomised double-blind placebo-controlled trial (OPPTIMUM). Health Technol Assess 22: 1–304, 2018. doi: 10.3310/hta22350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Conde-Agudelo A, Romero R. Vaginal progesterone for the prevention of preterm birth: who can benefit and who cannot? Evidence-based recommendations for clinical use. J Perinat Med 51: 125–134, 2023. doi: 10.1515/jpm-2022-0462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Heyborne K. Reassessing preterm birth prevention after the withdrawal of 17-alpha hydroxyprogesterone caproate. Obstet Gynecol 142: 493–501, 2023. doi: 10.1097/AOG.0000000000005290. [DOI] [PubMed] [Google Scholar]
  • 256. Shynlova O, Nadeem L, Dorogin A, Mesiano S, Lye SJ. The selective progesterone receptor modulator-promegestone-delays term parturition and prevents systemic inflammation-mediated preterm birth in mice. Am J Obstet Gynecol 226: 249.e1–249.e21, 2022. doi: 10.1016/j.ajog.2021.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Kim SH, Riaposova L, Ahmed H, Pohl O, Chollet A, Gotteland JP, Hanyaloglu A, Bennett PR, Terzidou V. Oxytocin receptor antagonists, atosiban and nolasiban, inhibit prostaglandin F2alpha-induced contractions and inflammatory responses in human myometrium. Sci Rep 9: 5792, 2019. doi: 10.1038/s41598-019-42181-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Klumper J, Breebaart W, Roos C, Naaktgeboren CA, van der Post J, Bosmans J, van Kaam A, Schuit E, Mol BW, Baalman J, McAuliffe F, Thornton J, Kok M, Oudijk MA. Study protocol for a randomised trial for atosiban versus placebo in threatened preterm birth: the APOSTEL 8 study. BMJ Open 9: e029101, 2019. doi: 10.1136/bmjopen-2019-029101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Premkumar A, Sinha N, Miller ES, Peaceman AM. Perioperative use of cefazolin and indomethacin for physical examination-indicated cerclages to improve gestational latency. Obstet Gynecol 135: 1409–1416, 2020. doi: 10.1097/AOG.0000000000003874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Reinebrant HE, Pileggi-Castro C, Romero CL, Dos Santos RA, Kumar S, Souza JP, Flenady V. Cyclo-oxygenase (COX) inhibitors for treating preterm labour. Cochrane Database Syst Rev 2015: CD001992, 2015. doi: 10.1002/14651858.CD001992.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Zamora-Leon P. Are the effects of DES over? A tragic lesson from the past. Int J Environ Res Public Health 18, 2021. doi: 10.3390/ijerph181910309. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiological Reviews are provided here courtesy of American Physiological Society

RESOURCES