Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Endocrinol., 19 August 2025

Sec. Reproduction

Volume 16 - 2025 | https://doi.org/10.3389/fendo.2025.1637570

This article is part of the Research TopicMaternal Factors and Vascular Remodelling in PregnancyView all 5 articles

The role of placental hormones and metabolites in modulating uterine circulation in physiological and pathological pregnancies

Milena EspositoMilena Esposito1Luana PaulesuLuana Paulesu2Maurizio Mandal,*Maurizio Mandalà1,3*
  • 1Department of Biology, Ecology & Earth Sciences, University of Calabria, Rende, Italy
  • 2Department of Life Sciences, University of Siena, Siena, Italy
  • 3Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States

The adaptation of the uterine circulation during pregnancy is fundamental to ensure an adequate supply of oxygen and nutrients to the fetus, and this process is largely orchestrated by placental hormones/metabolites. In this review, we comprehensively examine the role of placental hormones, growth factors, and proteins in mediating vascular remodeling, vasodilation, and angiogenesis within the uterine circulation under both physiological and pathological conditions. Key molecules such as estrogens, progesterone, relaxin, VEGF, PlGF, and PTHrP, among others, promote structural and functional adaptations of uterine arteries, reduce vascular resistance, and enhance uteroplacental blood flow. Additionally, we discuss the impact of placental dysfunction on the development of pregnancy-related disorders such as preeclampsia, intrauterine growth restriction, gestational diabetes mellitus, and placenta accreta spectrum conditions that share common features of impaired uterine vascular remodeling and altered placental secretome. Furthermore, we explore innovative therapeutic strategies that aim to restore placental and vascular function, including gene therapy, mesenchymal stem cell-based approaches, and targeted nanomedicine. Finally, we highlight the emerging role of placental biomarkers for early diagnosis and risk stratification of vascular complications in pregnancy. Understanding the intricate interplay between placental secretions and the maternal vasculature is critical to advancing the prevention, diagnosis, and treatment of pregnancy complications, ultimately improving maternal and fetal health outcomes.

1 Introduction

Pregnancy requires the adaptation of the maternal uterine vascular system and the development of the placenta to establish the feto-maternal circulation. Within the maternal vascular system, the most prominent adaptation is the remodeling of the spiral uterine arteries (UAs) by trophoblast cells invasion to initiate the placentation process (1). As a result, the spiral UAs enlarge their opening diameters by 5- to 10-fold compared to the non-pregnant state and are transformed into low-resistance conduits that deliver blood at low pressure to the intervillous space (2). The changes in UAs structure include expansion of volume and the outgrowth of newly formed vessels, which are necessary to expand the vascular surface area and create a suitable environment for fetal growth. These morphological adaptations of the uterine circulation are accompanied by physiological changes in uterine vascular reactivity. Indeed, pregnancy is characterized by enhanced vasodilation and a blunted vasoconstrictive response to accommodate the increased utero-placental blood flow (UPBF). These adaptations are accomplished through several mechanisms, including the proliferative and vasodilatory effects of increased levels of circulating growth factors, as well as the effects of pro- and anti-inflammatory cytokines and hormones secreted by the placenta into the maternal bloodstream. Studies have demonstrated the involvement of these placental hormones and metabolites in regulating uterine circulation through pathways specific to endothelium and vascular smooth muscle cells (VSMCs). Alterations in endothelial nitric oxide synthase (eNOS) expression and activity, nitric oxide production (NO), and expression of enzymes involved in prostacyclin (PGI2) production contribute to the uterine artery endothelium-specific responses mediated by these bioactive molecules.

Therefore, the hormones and metabolites secreted by the placenta are crucial for the adaptations of uterine circulation, and hence for the development and survival of the fetus.

The placenta is a temporary endocrine organ that develops immediately after implantation during pregnancy. The placenta becomes fully functional by the 11th week of gestation, and it regulates the bidirectional exchange of nutrients, oxygen and waste products between the fetal and maternal circulation. Despite accounting for less than 1% of maternal body weight, the placenta consumes approximately 40% of the oxygen supplied by the uterus at term (3), providing evidence for its high biosynthetic activity. The placenta secretes into the maternal circulation several hormones and metabolites, including estrogens, progesterone and relaxin, which are critical for the remodeling and the vasodilation of the uterine vasculature necessary to sustain the 40-fold increase in UPBF.

Therefore, placental insufficiency compromises feto-maternal circulation and has been frequently associated with pregnancy complications including preeclampsia (PE), intrauterine growth restriction (IUGR), gestational diabetes mellitus (GDM) and placenta accreta spectrum (PAS) disorders. These complications not only affect pregnancy outcomes but also have long-term consequences on offspring health. Thus, there is a need to maintain tight regulation of hormones/metabolites secreted by the placenta during pregnancy.

Throughout this review, the role of the placenta in mediating uterine vascular adaptations to pregnancy will be discussed in normal and complicated pregnancies such as PE, IUGR, GDM and PAS. Thus, understanding how hormones and proteins secreted by the placenta affect the uterine circulation in both physiological and pathological pregnancies is of interest for improving maternal and fetal outcomes.

2 Physiological regulation of uterine circulation by placental hormones and growth factors

The placenta exhibits massive endocrine activity. The coordinated action of hormones, proteins and growth factors synthesized by the placenta is essential in regulating UA expansion and reducing vascular resistance, supporting the increased UPBF required for fetal development (Figure 1).

Figure 1
Diagram showing the role of hormones and proteins in pregnancy-related vascular changes. It includes pathways for vasodilation, angiogenesis, and vascular remodeling, leading from high to low vascular resistance. Hormones like estrogens, relaxin, and progesterone are involved, affecting elements like nitric oxide and matrix metalloproteinases.

Figure 1. Mechanisms by which placental factors modulate uterine vascular adaptations to pregnancy. The figure illustrates the main placental factors and signaling pathways mediating vasodilation, angiogenesis and uterine vascular remodeling during pregnancy. The coordinated actions, transforming the UA into dilated vessel with low vascular resistance, thus sustaining the increase in UPBF and supporting a proper fetal growth.

The following section summarizes studies conducted on animals, humans, as well as cell culture, evaluating the role of hormones, growth factors and protein synthesized by the placenta, to elucidate their effects and mechanisms of action in processes underlying uterine vascular remodeling during pregnancy (Table 1).

Table 1
www.frontiersin.org

Table 1. Effects of placental factors in mediating vascular adaptations to pregnancy.

2.1 Estrogens

The placenta becomes the primary source of estrogen after week 9 of human pregnancy, when levels of estradiol increase by 50-fold during pregnancy (4). In vitro studies have demonstrated that estradiol-17β induces proliferation, migration and adhesion of endothelial cells (ECs) (58), critical processes underlying uterine angiogenesis and facilitating the increase in UPBF for a healthy pregnancy establishment. These effects have been observed in both human umbilical vein endothelial cells (HUVECs) and uterine arterial endothelial cells (UAECs) (58). In addition, estrogens contribute to uterine vascular remodeling by enhancing the transcriptional activity of serum and glucocorticoid-inducible kinase-1, which in turn modulates matrix metalloproteinases-2 (MMP-2) and E-cadherin expression in human villous samples (9).

Animal studies further demonstrate the role of estrogens in promoting vascular remodeling and hemodynamic adaptation. During early primate pregnancy, estrogens promote trophoblast cell proliferation, differentiation, viability, and invasion of the spiral UA (10). In pregnant rats, estradiol was shown to activate extracellular MMPs inducer, which in turn increased the expression and activity of MMP-2 and MMP-9 in uterus and aorta of late-pregnant rats vs virgin and mid-pregnant rats, supporting its role in vascular remodeling (11). However, in baboons, as gestation advances, the rise of estrogenssuppresses trophoblast uterine invasion (10) by inhibiting vascular endothelial growth factor (VEGF) expression and increasing soluble fms-like tyrosine kinase-1(sFlt-1) levels (12).

Furthermore, several studies have shown that estrogens vasodilate UA (1316), favoring the augmentation in UPBF during pregnancy. In pregnant ewes, systemic infusion of estradiol-17β induced a 5-fold increase in UPBF, mainly mediated by large-conductance Ca2+-activated K+ channel (BKca) and NO (17). In rats, estrogens were shown to act through the G protein-coupled estrogen receptor (GPER) (18, 19), promoting endothelium-dependent vasodilation via the NO–cyclic guanosine monophosphate (NO–cGMP) signaling pathway (20). GPER-mediated vasodilation also involves L-type Ca2+ channels and ERK1/2 activation in VSMCs (16).

In human myometrial arteries, vasodilation in response to 17β-estradiol is mediated by the greater expression of estrogen receptors-α, and -β (ER-α, -β) (15).

The effects of estrogens on uterine circulation during pregnancy have been extensively reviewed elsewhere (18, 21).

2.2 Progesterone

Progesterone is a steroid hormone whose levels during pregnancy are almost 10 times higher than during the luteal phase of the menstrual cycle (22). Around the 10th week of gestation, the placenta takes over progesterone production from the corpus luteum, maintaining serum concentrations of at least 10 ng/ml, which are essential for sustaining a healthy pregnancy (23). Progesterone contributes to thickening and expanding the endometrial lining, thereby enlarging the surface area available for implantation of the fertilized egg (24). It also facilitates trophoblastic invasion of the spiral UA, thereby promoting growth and remodeling of the uterine vasculature and increasing UPBF. Moreover, progesterone induces uterine vessel vasodilation by acting on both ECs and VSMCs. In ECs, it upregulates the expression and activity of eNOS (25) as well as cyclooxygenase-1 and -2 (COX-1, COX-2) (26), increasing the production of NO and PGI2 respectively (27). In VSMCs, progesterone downregulates the protein kinase C pathway (PKC), further contributing to vessel vasodilation (28).

Furthermore, progesterone, along with estrogen, promotes vascular remodeling by enhancing MMPs expression and activity (11). However, the effect of progesterone in regulating UPBF during pregnancy appears controversial (25); therefore, further studies are needed to elucidate its effects and the molecular mechanisms through which progesterone modulates the uterine vascular function during pregnancy.

2.3 Relaxin

Relaxin is produced mainly by the corpus luteum and, during pregnancy, also by the placenta. Relaxin reaches a peak of about 1 ng/ml by the end of the first trimester, then its levels gradually decline and plateau at ~0.6 ng/ml (29). Relaxin contributes to the growth and softening of the cervix, facilitating rapid labor, and it also contributes to uterine vasodilation and increases compliance, favoring the augmentation in UPBF (29). In vitro studies have demonstrated that relaxin stimulates angiogenesis. Experiments on human endometrial stromal cells showed that relaxin treatment induces a variety of angiogenesis-related genes, including VEGF-A (30). Moreover, relaxin has been shown to induce VEGF and basic fibroblast growth factor (31) expression in ischemic wound sites, supporting its pro-angiogenic activity.

Animal studies have provided strong evidence for relaxin role in vascular remodeling and uterine compliance. Kaftanovskaya et al., have demonstrated that relaxin-receptor-deficient mice had reduced lengths of the pubic symphysis and increased collagen density in reproductive tissues (32). Similarly, in pregnant relaxin-mutant mice, UA exhibited increased elastin expression and decreased levels of MMPs and adhesion molecules. Interestingly, 5 days of exogenous relaxin treatment reversed arterial stiffness and improved fetal weight in relaxin-deficient mice (33). Consistent with these findings, neutralization of relaxin in late pregnancy using the monoclonal antibody-1 (MCA-1) has been shown to increase uterine artery stiffness (34). Moreover, relaxin induced vasodilation in UA isolated from pregnant rats, with a greater effect during mid- compared to late pregnancy through the NO-sGC pathway (35). Recent evidence suggests that the vasodilatory responses of relaxin are mediated by its major receptor, the relaxin/insulin-like family peptide 1 receptor (RXFP1), which is most highly expressed in the UA during early pregnancy (34). Newly emerging data support that relaxin binds to RXFP1 activates Gαi/o protein, coupling to phosphatidylinositol-3 kinase/Akt (protein kinase B)-dependent phosphorylation and activation of eNOS (36). Nonetheless, relaxin inhibits spontaneous myometrial contractile activity in mid-gestation, while it has no effect at term (35).

Human studies suggest a potential role of relaxin in regulating uteroplacental vascular function. A positive correlation between serum relaxin levels and UA resistance index at 10–12 weeks of gestation has been observed, suggesting that relaxin contributes to the regulation of the uteroplacental vasculature. Moreover, suppression of circulating relaxin throughout mid-pregnancy abolished the cardiovascular adaptations required to sustain a healthy pregnancy (37). Thus, the sensitivity of the uterus to relaxin is subjected to modulation through pregnancy, suggesting that relaxin sustains an adequate UPBF during mid-gestation, while facilitating labor at term, allowing for vasoconstriction.

In summary, while animal studies show promising roles in vascular remodeling and compliance, human studies are limited and mostly observational, lacking mechanistic insights into RXFP1 signaling in the uterine vasculature during normal and pathological pregnancies

2.4 Vascular endothelial growth factor

In addition to steroid hormones, the placenta secretes a wide range of angiogenic factors, including VEGF, placental growth factor (PlGF) and sFlt-1, which collectively contribute to the regulation of the uterine circulation. In healthy pregnancies, maternal plasma VEGF concentrations are markedly elevated compared to non-pregnant baselines. It rises during the first trimester, peaks at 10–14 weeks, remains elevated up to the 20th week of pregnancy, then declines in the third trimester (38, 39). During the first trimester of pregnancy, VEGF levels increase 4–5 fold (163.2 ± 81.6 pg/mL) compared to the non-pregnant state (18.5 ± 16.8 pg/mL) (40). VEGF contributes to maternal adaptations to pregnancy by increasing vascular permeability, stimulating angiogenesis and inducing vasodilation of the uterine circulation.

The expression of VEGF mRNA in both the uterine endometrium (41) and placenta (42) indicates its active role at the feto-maternal interface, where tightly regulated modulation of vascular permeability and angiogenesis is crucial for the establishment of a successful uteroplacental circulation. VEGF-mediated vascular permeability has been demonstrated in the uterine vasculature and is further enhanced by pregnancy (43, 44). The increase in permeability facilitates the extravasation of plasma proteins into surrounding tissue, facilitating ECs migration and thus the angiogenic process (45). In addition, VEGF induces the expression of serine proteases urokinase, tissue-type plasminogen activators, PA inhibitor 1 (46) and MMPs interstitial collagenase (47), consistent with a pro-degradative environment, which facilitates migration and sprouting of ECs during angiogenesis. Notably, VEGF angiogenic properties are enhanced during pregnancy (48). VEGF role in mediating vasodilation has also been shown in vitro. Indeed, VEGF promotes NO production in cultured UAECs (48) and stimulates PGI2 synthesis in a time- and concentration-dependent manner in UVECs via activation of cytosolic phospholipase A2/p42/p44 MAP kinases pathways (49).

The critical role of VEGF in angiogenesis is supported by evidence from genetically modified pregnant mice lacking VEGF gene or deficient in VEGF receptors, both of which result in impaired vascular development and pregnancy loss (50, 51). VEGF promotes NO production in vivo, in the UA of pregnant ovine (52), while reducing UA contractility and increasing UPBF (5254) short- and long-term. Consistent with these findings, VEGF induced vasodilation of uterine arcuate artery (UAA) of both non-pregnant and pregnant rats, acting through an endothelium-dependent mechanism (55).

2.5 Placental growth factors

PlGF belongs to the VEGF growth factor family, and it binds specifically to VEGFR-1 (56). Plasma PlGF concentrations increase from week 11 to 12 onward to peak at week 30 in healthy pregnancy (57), coinciding with implantation and early vascular development. PlGF is expressed in villous trophoblasts and vascular endothelium in human placenta at term (58), and the correlation between PlGF expression and placental perfusion suggests that PlGF may contribute to ensuring adequate vascular development and function of the placenta early in gestation (59). PlGF contributes to uteroplacental circulation establishment by supporting angiogenesis, immune modulation and trophoblast invasion. Its role in vascular remodeling is supported by a study demonstrating a reduced uterine natural killer population in PlGF null mice (60). This sub-population of natural killer cells is the major participant in the early vascular changes in the pregnant endometrium. Thus, demonstrating that PlGF-mediated remodeling of the UA at the feto-maternal interface is necessary to sustain an adequate UPBF. Supporting this finding, an increased level of PlGF correlates with improved placental perfusion (59). In addition, PlGF potentiates the angiogenic response to VEGF on microvascular ECs (61), thereby promoting the angiogenic process at the feto-maternal interface. PlGF regulates the UPBF, inducing vasodilation, as demonstrated in placental arteries (62) and uterine circulation of rats and humans (63). The mechanism of PlGF-mediated vasodilation has been associated with signaling through VEGFR-1 and NO involvement, according to the vascular bed (63).

2.6 Additional placental hormones and proteins involved in vasodilation, vascular remodeling and angiogenesis of the uterine circulation during pregnancy

In addition to the well-known sex hormones and growth factors discussed above, whose role in pregnancy-associated uterine vascular remodeling has been widely demonstrated and established, several studies have suggested other placental hormones, proteins and growth factors as critical contributors to a successful uterine vascular remodeling.

2.6.1 Parathyroid hormone-related protein

PTHrP is expressed in the placenta and fetal tissues, and its expression increases as gestation progresses (64, 65). It plays a crucial role in placental calcium transport, a function important in maintaining fetal calcium homeostasis. Furthermore, PTHrP regulates uterine vascular tone during pregnancy, inducing vasodilation in the UA of both non-pregnant and pregnant rats (66) and human feto-placental vasculature (67, 68). This vasodilatory effect is mediated through the PTH1 receptor (PTH1R) and involves NO and prostacyclin production, leading to relaxation of VSMCs (66, 69). PTHrP increases the expression of COX-2 and the production of 8-iso-prostaglandin F2α, which in turn can regulate the expression of PTH1R, suggesting a feedback mechanism in VSMCs (70). Alterations in PTHrP expression have been associated with pregnancy complications. For instance, decreased levels of PTHrP have been observed in the placentas of spontaneously hypertensive rats, and they have been shown to correlate with IUGR (71). In conclusion, PTHrP is a multifunctional protein that, through its vasodilatory effects, regulation of vascular tone, and facilitation of calcium transport, ensures adequate UPBF and supports fetal development. However, despite these findings, the precise molecular mechanisms by which PTHrP regulates uterine vascular remodeling remain poorly defined, further human studies evaluating PTHrP levels in pregnancy complication are still lacking. Future research should also clarify whether PTHrP modulation could offer therapeutic benefits in pregnancy disorders.

2.6.2 Insulin growth factor

Insulin growth factor 2 (IGF-2) is abundantly expressed in trophoblast cells and fetal tissue (72). Studies on human uterine microvascular endothelial cells (UMVECs) have demonstrated that IGF-2 promotes ECs migration through the mannose 6-phosphate receptor (73). In addition, IGF-2 has been shown to promote endothelial proliferation and cell survival in placental villous explants, indicating a broader function in placental angiogenesis. The importance of IGF-2 in fetal development is further underscored by studies in mice, where IGF-2 deficiency resulted in growth restriction (74), indirectly supporting its action on UPBF. Thus, IGF-2 contributes to the vascular adaptation to pregnancy mainly by promoting angiogenesis.

Although the role of IGF-2 in placental angiogenesis and fetal growth has been documented gaps remain in understanding its effects on the maternal uterine circulation in the context of vascular tone and UPBF regulation. Further, the molecular pathways through which IGF-2 might influence NO production or expression, as well as PGI2 are largely unexplored. Future studies should clarify these mechanisms particularly through in vivo models and functional assessment of uterine circulation to better define IGF-2 contribution to maternal vascular adaptation during pregnancy.

2.6.3 Pregnancy-associated plasma protein A

PAPP-A is a proteinase highly expressed during pregnancy (75). PAPP-A enhances IGF bioavailability (76), which in turn promotes trophoblast invasion and angiogenesis, leading to a proper vascular remodeling required for adequate uteroplacental perfusion and fetal development. Low maternal serum PAPP-A has been associated with high UA resistance index (77), suggesting PAPP-A involvement in the regulation of UBPF.

Although PAPP-A is associated with improved IGF bioavailability and favorable pregnancy outcomes, come mainly from observational rather than direct mechanistic findings. The mechanisms by which PAPP-A modulates UA vasodilation and vasoconstriction events during pregnancy, as well as the downstream effects of altered PAPP-A expression on uterine microvascular networks beyond the spiral UA remain poorly defined.

2.6.4 Human chorionic gonadotropin

hCG is a glycoprotein hormone produced predominantly by placental syncytiotrophoblast. hCG receptors have been identified on uterine ECs (78, 79), and hCG promotes angiogenesis by stimulating ECs migration and capillary sprouting (80). Evidence from luteal tissue also supports its pro-angiogenic properties, increasing proliferation of ECs and expansion of both endothelial and pericyte compartments, thus contributing to vascular stabilization during pregnancy (81). Additionally, hCG increases migration, proliferation and invasion of trophoblast cells into the maternal decidua and the subsequent remodeling of spiral UA into low-resistance, high-capacitance vessels, essential for adequate placental perfusion (82). Moreover, hCG induces vasodilation of UA through the NO pathway, reducing vascular resistance and enhancing UPBF (83).

However, the downstream intracellular signaling pathways mediating hCG’s pro-angiogenic and vasodilatory actions—beyond NO—are not comprehensively characterized. Longitudinal studies examining hCG fluctuations and uterine hemodynamics throughout pregnancy, along with experimental models manipulating hCG levels, would help clarify its direct role in uterine vascular adaptation and disease susceptibility.

2.6.5 Placental protein 13

PP13, also known as galectin-13, is a member of the galectin family predominantly secreted from placental syncytiotrophoblasts. In addition to its immunomodulatory function (84), PP13 contributes to the vascular remodeling of the spiral UA (85) and uterine veins (UV) (86), ensuring adequate UPBF to the fetus. In vivo studies have demonstrated that PP13 administration lowered blood pressure and promoted placental and fetal growth (86). These findings are further supported by ex vivo evidence demonstrating PP13 vasodilation effects in UA from both rats (8789) and humans (90). The vasodilation action of PP13 is mediated, at least in part, through the NO pathway. By promoting increased UPBF, PP13 supports fetal development and contributes to the maintenance of a healthy pregnancy.

Although the contribution of PP13 in mediating the uterine vascular adaptation to pregnancy has been widely demonstrated in animal models or ex vivo, there is a lack of human in vivo data confirming its role in UPBF regulation.

2.6.6 Placental lactogen and placental growth hormone

PL and GH-V are members of the growth hormone/prolactin family expressed in the human syncytiotrophoblast. Although a direct association between PL and UPBF has not been definitively established (91), maternal circulating levels of PL have been positively associated with fetal weight (92). Consistent studies in animal models have demonstrated that PL deficiency resulted in reduced fetal and placental weight (93), indirectly indicating a potential contribution of PL to adequate UPBF necessary for normal fetal growth. The full-length hormone PL stimulates blood vessel formation in vivo at different stages of CAM development (94), thus sustaining its potential pro-angiogenic effect at the feto-maternal interface. However, the role of PL in mediating the vascular adaptation to pregnancy remains inconclusive and needs to be further elucidated.

GH-V is specifically expressed in the syncytiotrophoblast and invasive EVTs of the human placenta (95). It has been shown to promote EVTs invasive-phenotype in vitro (95), which is an essential process for spiral UA remodeling. Further, its pro-angiogenic effect has been proved in bovine brain capillary cells (94). GH-V correlates with the increases in circulating IGF-1 observed during pregnancy (96) and it is associated with fetal growth (97, 98). However, conflicting results exist on the relationship between maternal GH-V/IGF-1 concentrations and fetal growth during pregnancy (98).

In summary, while PL promote angiogenesis, GH enhances EVT invasion and is associated with IGF-2 increases. However, direct evidence linking PL and GH with uterine hemodynamics in humans is missing. Further, data on receptors and downstream pathways in ECs or VSMCs needs to be clarified.

2.6.7 Leptin

Leptin, a peptide hormone primarily secreted by adipocytes, is also produced by the placenta, particularly by syncytiotrophoblasts (99). Leptin treatment resolves the infertility in obese female mice (100) and it is essential for implantation processes (101). Leptin is likely to play a role in placental development, as its receptors have been identified on placental cytotrophoblasts (CTBs) (102). In vitro studies have demonstrated that leptin promotes CTB invasion (103) through STAT3, PI3K, MAPK pathways (103) and partly by modulating MMPs activity (104). Specifically, leptin increased the secretion of MMP-2 and fetal fibronectin and enhanced the activity of MMP-9 (105). Further, leptin treatment has been shown to induce angiogenesis in UAECs (106), in UVECs and in porcine aortic ECs (107). Although leptin has demonstrated vasodilation effects in the human forearm (108), its direct vasoactive role in the uterine circulation remains to be elucidated. Further, the balance between the pro-angiogenic vs pro-inflammatory effects leptin-mediated in the uterine vasculature is poorly understood, thus further studies need to be addressed.

2.6.8 Corticotropin-releasing hormone

CRH is secreted by syncytiotrophoblast cells of the human placenta into both the fetal and maternal circulation (109). It regulates the hypothalamic-pituitary-adrenal axis and the initiation of labor. Further, CRH acts as a vasodilator of the feto-maternal circulation (110) and UA from pregnant rats via NO–cGMP-EDHF signaling (111). Hence, providing evidence that CRH may act as a regulator of the UPBF. Furthermore, CRH has been shown to attenuate the invasiveness of EVTs, thereby contributing to ensuring controlled remodeling of spiral UA (112). The dual action of promoting vasodilation and regulating trophoblast invasion demonstrates that CRH contributes to the remodeling of the spiral UA. Most of the vasodilatory evidence is from animal models; confirmation in human uterine arteries is limited, further, the interaction with other placental hormones in regulating vascular tone is poorly explored.

2.6.9 Neuropeptide Y

NPY is widely expressed in the central and peripheral nervous systems, and it is also produced by placental and fetal membranes (112). NPY regulates UPBF mainly by inducing vasoconstriction through Y1 receptors located on VSMCs (113). However, UAs isolated from pregnant animals showed a reduced sensitivity to NPY (114), which suggests a finely tuned regulatory mechanism to balance vascular tone and ensure adequate UPBF. In normal placentas, Y2R- the NPY receptor involved in promoting angiogenesis (115) and vasodilation (116)- is the most abundantly expressed among the NPY receptors (117). These changes in receptor expression patterns support a role for NPY in the vascular adaptation of pregnancy. Nonetheless, the specific contribution of NPY and Y2R in regulating UA vascular tone and angiogenesis during pregnancy needs to be further investigated.

2.6.10 Inhibin A

Inhibin A is produced by placental syncytiotrophoblast during pregnancy, and its circulating levels increase as pregnancy progresses. While direct evidence of inhibin A role in uterine vascular remodeling is limited, its elevated levels have been associated with PE and IUGR (118), suggesting a potential role of inhibin A in placental function and uterine perfusion during pregnancy. To the best of our knowledge, no direct evidence supports vasodilation or angiogenic properties of inhibin A on the uterine circulation during pregnancy, although inhibin A induces angiogenesis in pathological processes (119). Therefore, given the similarities between tumors and the human placenta, the pro-angiogenic properties of inhibin A on uterine circulation during gestation remain to be elucidated.

2.6.11 Activin A

Activin A is a member of the transforming growth factor-beta (TGF-β) superfamily and the placenta is the major source of activin A throughout pregnancy (120). The importance of activin A in sustaining a physiologic pregnancy is suggested by the association between its levels and pregnancy complications (121). Activin A receptors are expressed on human trophoblast cells (122) and their role in promoting trophoblast cells invasion, differentiation and migration has been widely demonstrated in HTR8/SV neo cell culture (123126). Further, Activin A has been shown to enhance VEGF expression, thereby promoting angiogenesis (126). Interestingly, in UVECs activin A promotes the expression of adhesion molecules (127), contributing to the maternal endothelial dysfunction observed in women with PE (128). Although direct evidence of a vasodilatory effect of activin A on UA is lacking, its involvement in trophoblast-endothelial crosstalk suggests a role in mediating the adaptations of the uterine circulation to pregnancy.

2.6.12 Pregnancy-specific β-glycoproteins

PSGs are secreted mainly by trophoblast cells throughout gestation (129). During pregnancy, members of the PSGs family regulate the vascular function, promoting angiogenesis and vasodilation. In vitro, treatment with PSG-1 enriched exosomes promotes UVECs proliferation and migration, as well as NO release (130). Consistent with these findings, Qin et al., have demonstrated a role of PSG-9 in increasing NO production through the enhanced expression levels of store-operated calcium entry channels proteins (131). In addition, PSG-1 induces angiogenesis via TGFB-1 and VEGF-A, thus contributing to the establishment of the feto-maternal circulation during pregnancy (132). However, the direct effects of PSGs on uterine artery reactivity and spiral artery remodeling in vivo are largely unexplored. Moreover, the specific contributions of different PSG isoforms beyond PSG-1 and PSG-9 to uterine vascular adaptation remain unclear. Further research is needed to elucidate the receptor-mediated pathways involved and to investigate whether PSGs exert differential effects in normal versus pathological pregnancies.

3 Pathological conditions and dysregulation of uterine circulation

Appropriately timed pregnancy-dependent changes in vasculature are critical for healthy pregnancy outcomes. Thus, alterations in the secretion of placental hormones and metabolites compromise the physiological adaptation of the uterine circulation, leading to reduced UPBF. Most of the pregnancy losses occur early in gestation, when the placenta is established. These losses may be associated with aberrations in remodeling of the uterine vasculature and angiogenesis imbalance at the feto-maternal interface, resulting in pregnancy complications such as PE, IUGR, GDM, and PAS as discussed in the following section.

3.1 PE

PE is defined as new-onset hypertension after 20 weeks of pregnancy, with or without proteinuria and evidence of end-organ damage. Clinically, PE is associated with several maternal and fetal complications (133), and is generally classified in early-onset (<34 weeks of gestation), which tends to be more severe, and late-onset (>34 weeks of gestation), which is usually milder. The placenta has a crucial role in the pathophysiology of both early- and late-onset PE, though the specific mechanisms differ. Early-onset PE is more strongly associated with abnormal placentation, poor uterine perfusion, and IUGR (134), whereas late-onset is primarily attributed to predisposing maternal factors and placental senescence (134).

It is generally accepted that PE originates from the abnormal transformation of the spiral arteries underlying the placenta, due to impaired invasion of the uterine wall by migrating trophoblast cells (135, 136). In addition to incomplete or absent vascular remodeling, signs of vascular damage, resembling atherosclerosis-like lesions, have been described in placental bed samples from women with PE compared to women with healthy pregnancies (135). This maladaptive phenotype leads to narrower maternal uterine vessels and relative placental ischemia, ultimately reducing the UPBF as demonstrated by Doppler ultrasound (137139).

Numerous hormones and proteins of placental origin have been investigated to elucidate the molecular mechanisms underlying the aberrant uterine vascular phenotype observed in PE. It is well-established that PE is associated with a decrease in pro-angiogenic factors such as PlGF and VEGF, coupled with an increase in anti-angiogenic factors such as sFlt-1 and sEng.

Supporting this notion, a reduction in circulating PlGF has been associated with placental hypoperfusion in both human (140) and animal models of PE (141). Placental hypoperfusion, indicative of defective adaptations of the uterine circulation, may be partially explained by a substantial elevation of sFlt-1 observed in PlGf-/- mice (142). Notably, PlGF administration dampened sFlt-1 increase, restoring angiogenic signaling in a PE-like model (141). The anti-angiogenic effects of sFlt-1 are further exacerbated by sEng, which has been shown to induce a severe state of PE in pregnant rats (143). The exacerbation of the PE-like symptoms is further supported by in vitro evidence showing that, in UVEC, sEng treatment contributes to endothelial dysfunction by inhibiting NO production, reducing cell viability, impairing trophoblast invasiveness, as well as suppressing MMPs expression (144).

Thus, PE is characterized by a shift toward an antiangiogenic profile, which disrupts the normal vascular homeostasis, resulting in endothelial cell dysfunction, including decreased NO production, and release of procoagulant proteins. Hence, contributing to hypertension and reduced UPBF.

3.2 IUGR

IUGR is defined as fetal weight estimated to be below the 10th percentile for its gestational age. The most common cause of IUGR is poor placental function and placental ischemia due to deteriorated uteroplacental perfusion (145, 146). A strong association between IUGR and PE has been consistently observed, with IUGR frequently arising in pregnancies complicated by severe PE (147). Histopathological observations revealed a significant decrease in villous vascular density (148) and a maldevelopment of the placental terminal villous tree in placentae from IUGR pregnancies compared to those from normal pregnancies (149). These findings suggest that the aberrant vascular formation is a leading determinant in IUGR, explaining the sub-optimal UPBF. From a molecular standpoint, the angiogenic profile of IUGR placentas presents inconsistencies that may reflect compensatory and pathogenic mechanisms. A study from Barut et al., reported increased placental expression of VEGF-A, basic fibroblast growth factor and eNOS, in placental samples from IUGR mothers compared to control samples collected during the third trimester (150). This result suggests a possibly compensatory upregulation aimed at resolving placental hypoxia. However, Lyall et al. (151), found a reduction in VEGF expression in placental villous tissue from pregnancies complicated by IUGR and PE, indicating that angiogenic insufficiency may dominate in such pathological contexts. Similarly, the expression of PlGF mRNA in placental samples from IUGR pregnancies has been inconsistent, with evidence demonstrating a decrease in PlGF expression only in severe cases of IUGR (152). Study using an ovine model of IUGR has demonstrated an increase in VEGF mRNA levels in the IUGR group compared to the control in early gestation, possibly reflecting early compensatory mechanisms. However, during mid-gestation, the expression of VEGF receptors in fetal tissue was significantly reduced, potentially limiting VEGF and PlGF signaling, thereby impairing angiogenesis and placental vascular development (153).

This finding may reflect a progressive failure of the placenta to sustain proangiogenic signaling under chronic stress, hence affecting UPBF.

3.3 Gestational diabetes mellitus

GDM, defined as hyperglycemia first diagnosed during pregnancy (154), affects up to 20% of pregnancies worldwide (155), and it is frequently associated with uteroplacental insufficiency, as well as increased risk of IUGR and PE (156, 157).

Morphological and structural abnormalities have been documented in human GDM placentas (158, 159) as well as in placentas from hyperglycemic animal model, resulting in IUGR (160). Placentas from hyperglycemic-induced animal model exhibited disrupted trophoblast invasion, inadequate spiral UA remodeling (160), as well as dysregulation in PGI2 levels (161, 162) and decreased levels of VEGF and PlGF (163, 164).

Collectively, the molecular, structural and functional abnormalities observed in GDM pregnancy suggest a defective vascularization due to impaired placental development in GDM pregnancy, affecting the uteroplacental circulation and thus compromising the UPBF.

3.4 Placenta accreta spectrum

PAS encompasses a spectrum of conditions characterized by abnormal placental adhesion to the uterine wall, which fails to detach at birth (165). The vast majority of PAS cases are attributed to scarring from cesarean births, which disrupts the endometrial-myometrial interface and promotes aberrant implantation (166168). In PAS, EVTs excessively invade the myometrium (169), often within a rigid, collagen-rich extracellular matrix (170), accompanied by inflammation and hypoxia in uterus (171, 172). The absence of proper decidualization and EVT invasiveness prevents the remodeling of the uterine spiral artery in PAS (173), resulting in an abnormal pulsatile and high-velocity blood flow at the feto-maternal interface (174). Histopathologic analysis has revealed excessive vascularity in scarred uterine tissue in PAS (173) (175) and a lack of structural integrity of the vessel, potentially due to Von Willebrand factor suppression as demonstrated in ECs of PAS epiplacental artery (176).

The excessive vascularization is supported by upregulation of VEGF, angiopoietin-2 (177) and PlGF (178) in PAS lysates, and reduced expression of antiangiogenic factors such as vascular endothelial growth factor receptor-2 (VEGFR-2), endothelial cell tyrosine kinase receptor (Tie-2), and sflt-1 in syncytiotrophoblast cells from PAS placenta specimens (177). While evidence on VEGF levels in PAS remains controversial (178), hormonal regulation of angiogenic signaling may contribute to this imbalance. Indeed, the relaxin gene is upregulated in the basal plate of PAS placentas, together with RFXP1 in both the basal plate and villous trophoblast (179).

Animal models have recapitulated this condition, including placental dysplasia, incomplete remodeling of the spiral arteries, deep trophoblast invasion at the feto-maternal interface and reduced placental perfusion, as well as imbalances in angiogenic and anti-angiogenic factors within the placenta and in peripheral blood (180).

4 Therapeutic interventions

Understanding the molecular mechanisms by which placental hormones and proteins regulate UA adaptations and functions during pregnancy offers novel opportunities for therapeutic strategies in the context of pregnancy diseases. The administration of pregnancy-related hormones and proteins involved in uterine vascular adaptations during healthy gestation may restore the maternal vascular homeostasis in pathological conditions. Remarkably, as naturally occurring molecules synthesized during pregnancy, their administration is less likely to cause adverse effects on maternal and fetal health during pregnancy. As previously discussed, hormones and metabolites secreted by the placenta exert pleiotropic actions during pregnancy, promoting the uterine vascular adaptations to pregnancy. Thus, the preclinical evidence makes them particularly attractive candidates for therapeutic use.

For example, relaxin infusion in women at >40 weeks of gestation did not induce adverse side effects and demonstrated a modest reduction in systolic blood pressure (181). However, relaxin research has advanced to clinical trials primarily in the context of acute heart failure patients, demonstrating good tolerability, improved renal function, and enhanced systemic perfusion following its infusion (182, 183).

Progesterone is another placental hormone with emerging therapeutic potential. Evidence for progesterone beneficial effects during pregnancy arises from the PROMISE trial (184) and the PRISM trial (185), in which first trimester initiation of vaginal progesterone prevented pregnancy loss, lowering the risk of PE and the risk of pregnancy hypertensive disorders (186). Similarly, hCG administration induces a significant improvement in the pregnancy success rate in women with oligomenorrhea (187). Although there were no adverse effects of administering hCG during pregnancy, the evidence supporting its supplementation to prevent recurrent miscarriage remains equivocal (188).

Hence, hormones and proteins secreted by the placenta are naturally occurring molecules that normally circulate in the maternal bloodstream during pregnancy. Although the number of pregnancy-specific clinical trials remains limited, the existing evidence from preclinical and clinical studies indicates a favorable safety profile and mechanistic plausibility for their use during pregnancy. Thus, their administration to women with complicated pregnancy would be expected to ameliorate the maternal vascular dysfunction observed in some pregnancy-related disorders, enabling the activation of several beneficial pathways through the administration of one therapeutic. However, future clinical trials specifically designed to evaluate efficacy, timing of administration, and optimal dosing in women with pregnancy diseases characterized by placental dysfunction will be crucial for improving our understanding of innovative therapeutic interventions.

4.1 Placental-target drug delivery systems

The ability of most drugs to reach therapeutic concentrations is limited by the placenta, which acts as a selective barrier between fetal and maternal circulation. To face these challenges, a range of methods for placental-targeted drug delivery have been developed. These include stem cell-based therapies, lipid nanoparticles therapies, and gene therapies.

4.1.1 Stem cell-based therapies

Stem cells are undifferentiated cells, capable of self-renewal and of differentiating into several specialized cells (189). These cells have been isolated from embryonic tissues, placenta, and amniotic fluid (190). Stem cell-based therapies using mesenchymal stromal cells have been shown to hold potential in promoting vascular health (191). In vitro, transplantation of mesenchymal stem cells genetically modified to express the heme-oxygenase 1 gene promoted placental vascularization and restored angiogenic balance (192). Accordingly, in a PE-like animal model, transplantation of mesenchymal stem cells modified to express the heme-oxygenase 1 gene alleviates PE symptoms, promoting angiogenesis and improving placental perfusion (193). More recently, exosomes derived from human umbilical cord mesenchymal stem cells have been shown to induce similar therapeutic effects as mesenchymal cells themselves. For instance, in animal models of PE, exosome-derived mesenchymal cells decreased blood pressure and proteinuria (194196), and decreased the death rate of the fetuses (195) as well as the fetal birth weight (196). Furthermore, human umbilical cord mesenchymal stem cells-derived exosomes have been shown to rescue sFlt-1-induced HUVECs dysfunction in vitro (196).

4.1.2 Lipid nanoparticles therapies

The lipid nanoparticles (LNPs) are nanostructured lipid carriers that encapsulate nucleic acids for efficient intracellular delivery (197). LNPs can be classified into liposomes, solid lipid nanoparticles and nanostructured lipid carriers. Drug-loaded LNPs have been successfully employed in the treatment of both acute and chronic disorders (198). However, the use of LNPs-based therapies during pregnancy requires careful consideration, as it introduces unique challenges to drug delivery. A recent study demonstrates that the structural composition and delivery route of LNPs during pregnancy critically influence mRNA delivery efficiency, maternal immune activation and fetal outcomes (199). Thus, there is a need to design pregnancy-adapted LNPs for safe and effective placental-targeted therapies. For example, LNPs encapsulating VEGF-mRNA triggered vasodilation in the placentas of pregnant mice (200), resolved maternal hypertension and partially restored placental vasculature, the local and systemic immune response, and serum levels of sFlt-1 (201). Similarly, LNPs delivering PlGF mRNA to the placenta in pregnant mice achieved efficient protein expression without maternal or fetal toxicity, further supporting their potential therapeutic action in treating placental-related dysfunction (202).

4.1.3 Gene therapy

Gene therapies typically require a vector to introduce gene material into target cells. The vector can be either viral or non-viral, and both have proven effective for transporting therapeutic agents directly to specific organs and cells, including the placenta. Major viral vectors used for gene therapies are adenovirus, lentivirus and adeno-associated virus, which differ in efficiency, duration of gene expression and immunogenicity. For example, in pregnant mice, the use of RGD fiber-mutant adenoviral vectors enhanced placental tropism and gene transfer efficiency by 10- to 100-fold compared to conventional vectors and sustained transgene expression for at least 7 days (203). Notably, RGD fiber-mutant adenovirus vectors did not induce placental dysfunction or fetal loss (203), suggesting a favorable safety profile for RGD fiber-mutant adenoviral vectors during pregnancy. Intra-arterial administration of adenoviral vectors encoding the VEGF gene has been associated with enhanced fetal growth (204, 205) and increased UPBF (52) in animal models.

Among non-viral vectors, nanostructured delivery systems complexed with the IGF-1 gene have shown promising results in pregnancy models. In vivo studies in pregnant mice demonstrated that this vector effectively delivered the gene in the placenta and alleviated IUGR (206). Similarly, in guinea pigs, IGF-1 gene delivery promoted vascular remodeling, enhancing the expression of growth factors (207) and increasing the fetal capillary volume density along with fetal glucose transport (208). Accordingly, in ex vivo human placental explants, perfusion with the IGF-1 vector increased human IGF1 expression in villous fragments and enhanced translocation of glucose transporters (209).

In addition to gene transfection approaches, gene silencing strategies using small interfering RNAs represent other possibilities in the treatment of pregnancy-related disorders. In this regard, the use of siRNA for sFlt-1 has been shown to lower maternal blood pressure and to reduce proteinuria in an animal model of PE (210, 211). Thus, comprehensively, these approaches may have the potential to improve the hemodynamic disturbance observed in pregnancy complications such as PE and IUGR.

5 Predictive biomarkers and diagnostics

A wide range of serum biomarkers have been investigated as potential markers for early screening of pregnancy complications (see Tables 25).

Table 2
www.frontiersin.org

Table 2. Biomarkers associated with PE categorized by trimester of pregnancy at screening.

Table 3
www.frontiersin.org

Table 3. Biomarkers associated with IUGR categorized by trimester of pregnancy at screening.

Table 4
www.frontiersin.org

Table 4. Biomarkers associated with GDM categorized by trimester of pregnancy at screening.

Table 5
www.frontiersin.org

Table 5. Biomarkers associated with PAS categorized by trimester of pregnancy at screening.

5.1 Biomarkers for diagnosing PE

5.1.1 Hormonal and protein biomarkers

PAPP-A levels <10th percentile in the first trimester are associated with an increased risk of PE (209, 212). Its predictive value improves when combined with other markers, such as β-hCG (213215) and UA pulsatility index (216).

In the early second trimester of pregnancy (16–18 weeks of gestation), elevated β-hCG levels have been associated with PE and particularly severe PE (217). However, other studies demonstrate inconsistent findings (216, 218). A recent meta-analysis confirmed increased levels of serum β-hCG in the early second trimester, but not during the first trimester, in pregnancies later complicated by PE compared to healthy pregnancies (219).

Another biomarker used to predict PE is A-Disintegrin and Metalloprotease-12 (ADAM-12) decreased levels, in association with PAPP-A, correlate with the severity of IUGR (220), though findings are inconsistent for ADAM-12 association with PE and related disorders (221). Conflicting evidence, such as increased ADAM-12 levels reported in PE and HELLP syndrome (222), underscores the need for further validation.

Research has also been conducted on the use of PP13 in the prediction of PE. Low first-trimester levels are associated with PE risk (223225), while increased amounts of PP13 carried via the placental-associated extracellular vesicles have been reported in late gestation (226). Combined with PAPP-A and the free leptin index, PP13 can reach a detection rate of 40% at a 10% false positive rate (227). Inhibin A levels are elevated during both the first trimester (228) and the second trimester (229) in pregnancies complicated by PE, and its levels correlate with the incidence and severity of PE (229). However, to increase its predictive value, the level of inhibin-A is used in combination with other biomarkers such as PAPP-A (228), endoglin and PlGF (230). Additional hormones have also been explored for PE screening. For example, relaxin levels decrease during the first trimester of pregnancy and have been associated with late-onset PE (231), though relaxin levels may not reflect disease severity (232).

Similarly, decreased serum levels of PTHrP (233) E2 and progesterone (234) are lower in preeclamptic pregnancies compared to healthy pregnancies.

A reduction in PL levels has also been associated with PE onset, making it a reliable marker of placental function in the second half of pregnancy (235).

Conversely, leptin concentration is significantly increased in PE, both early and late in gestation (236, 237).

CRH levels increase in late pregnancy in women with PE (238, 239), with a sensitivity of 92.5% and specificity of 82.5% as a single predictive marker for PE (238).

Elevated serum NPY levels have also been reported in PE during the third trimester (240). Interestingly, another study found a decrease in NPY2 receptor rather than NPY expression in late pregnancy (117).

Finally, the role of activin A remains unclear. Although no significant difference between PE and healthy pregnancy has been demonstrated in its levels through gestation (241), a slight increase in the first trimester was observed in another study; however, the association remains too weak to support its use in PE screening (242).

5.1.2 Growth factors biomarkers

A recent study found that the levels of IGF-1 and its receptor in the serum and in placental tissue were significantly lower in the preeclamptic group compared to a control group. IGF-1 exhibited a sensitivity of 86% and a specificity of 100% for diagnosing PE, while serum IGF-1R showed a sensitivity and a specificity of 77% for diagnosing PE (243). Angiogenic factors have emerged as a useful tool for the identification of patients at risk for pregnancy complications. The lowering of PlGF alone (244) or in association with VEGF (245), as well as in association with sFlt-1 (246), has been associated with PE. In addition, increased sFlt-1, sFlt-1/PlGF ratio, and a decline in PlGF levels have also been associated with PE (247). Further, PlGF in association with PAPP-A may predict early onset PE during the third trimester (228).

5.2 Biomarkers for diagnosing IUGR

Hormonal and protein biomarkers - Low levels of PAPP-A during the first trimester have been consistently associated with IUGR (248, 249). The predictive value improves when PAPP-A is combined with UA Doppler pulsatility index (249).

While some studies found no significant association between β-hCG and the occurrence of IUGR (249), a meta-analysis reported increased levels of β-hCG during the second trimester in pregnancy complicated by IUGR (250).

PP13 has shown potential as a predictive biomarker for IUGR, particularly when assessed during the second trimester, where PP13 levels are significantly. However, its predictive accuracy is higher for PE than for IUGR (251). Nonetheless, maternal serum PP13 in the first trimester may predict the risk of developing IUGR, PE, or PE complicated by IUGR (252). However, PP13 role in predicting IUGR remains contradictory (253).

A positive correlation between maternal PL levels and estimated fetal weight has been reported in a cross-sectional study, suggesting its potential utility in monitoring fetal growth (235, 254). Importantly, PL reduction has been significantly associated not only with the onset of PE, but also with asymmetrical IUGR, supporting its role as a marker of placental dysfunction (235).

Elevated maternal serum leptin concentrations at delivery have been documented in pregnancies complicated by IUGR compared to normal pregnancies (255). Similarly, elevated CRH levels in the third trimester of pregnancy have been associated with a 3.6-fold increase in IUGR risk (256). Molecular studies using in silico approaches further support these findings, showing increased RNA expression of PAPP-A2 and CRH in both maternal blood and placental tissue from IUGR pregnancies (257).

Additionally, higher levels of inhibin A and B have been observed in fetuses affected by IUGR compared to controls (258). Lastly, lower levels of serum PSG-1 have been reported in the late second trimester of IUGR pregnancies (259).

Growth factors biomarkers During the third trimester, pregnancies with IUGR showed significantly lower levels of free and total PlGF (260). A meta-analysis revealed that pregnant women with IUGR and PE may have higher sFlt-1/PlGF ratio. A sFlt-1/PlGF ratio >33 showed strong predictive capacity in FGR (261). Nonetheless, similar to Flt-1/PlGF, sEng has high predictive value for IUGR along with PE and HELLP (262). IUGR pregnancies, along with PE, showed significantly elevated sEng concentrations and a strong positive correlation with sFlt-1 (263). Remarkably, placental transcript abundance for VEGF was significantly lower in IUGR placentae compared to healthy placentae (264). Additionally, increased sEng levels have been consistently associated with GDM, PE, and IUGR across different stages of gestation (265, 266). Despite these associations, a meta-analysis by Conde-Agudelo et al., including 53 studies and 39,974 women, concluded a minimal predictive accuracy of angiogenic factors for IUGR (267).

5.3 Biomarkers for diagnosing GDM

5.3.1 Hormonal and protein biomarkers

Plasma levels of relaxin-2 have been found to increase in both the first- (268) and third-trimester of pregnancy in women with GDM (269).

The association between β-HCG and GDM appears more controversial. One study reported that the incidence of GDM was increased in women with elevated β-HCG levels during the second trimester of pregnancy (250, 270). In contrast, Liu et al., found that increased β-HCG levels in early pregnancy were associated with a lower risk of GDM (271), raising the possibility of a time course-dependent profile in the predictive capacity. Supporting this complexity, a machine learning study demonstrated improved prediction accuracy for GDM when β-HCG was used in combination with PAPP-A (272).

Although PAPP-A alone may not be a definitive predictive marker for GDM (273, 274) low levels could support the recommendation for early screening as part of a broader diagnostic approach. Low serum PAPP-A MoM levels are significantly associated with the development of GDM,

Increased serum levels of PP13 in the early second trimester were significantly associated with GDM, with a detection rate of 92.3% at 80% specificity (275). This preliminary study may support PP13 as an early screening biomarker, though its implementation in clinical practice requires validation.

PL has also been implicated in GDM. A meta-analysis showed that in women with type 1 GDM, PL is decreased in early pregnancy and increased in late pregnancy (276), suggesting that PL dynamics may reflect placental adaptation to altered glycemic states.

In addition, increased leptin levels were observed in the third trimester of pregnancies complicated by GDM compared to normoglycemic pregnancies (277, 278). Accordingly, a meta-analysis confirmed these data, revealing increased leptin concentrations in GDM cases regardless of gestational age, with no trimester-dependent variation in its level (279).

5.3.2 Growth factors biomarkers

Similar, GDM has been associated with increased levels of sFlt-1 (280). However, the predictive value of PlGF in GDM remains uncertain (244, 281). Accordingly, increased levels of VEGF-A, endoglin, endothelin-1, and angiopoietin-2 have also been documented in GDM placentas compared to controls (265). In a longitudinal study, the sFlt-1 levels were higher in the first trimester in GDM women as compared to non-GDM women. Placental PlGF and Flt-1 levels were lower in the GDM group, and they were negatively associated with placental dimensions, revealing that an imbalance in these growth factors may affect placental development, hence pregnancy outcomes in GDM (282). Additionally, increased sEng levels have been consistently associated with GDM (283).

5.4 Biomarkers for diagnosing PAS

Hormonal and protein biomarkers - Increased levels of PAPP-A during the first trimester of pregnancy have been observed in women at higher risk of PAS (284, 285). In contrast, another study reported that PAPP-A levels were lower in PAS cases, while β-hCG levels were increased compared to the control group. Interestingly, combining these biomarkers in predictive models substantially improved diagnostic accuracy over single-marker use, with a sensitivity of 100% and specificity of 72% (286). Studies have also examined the diagnostic value of hyperglycosylated human CG. One study found that levels of this hormone were lower in PAS cases compared to controls in both the second and third trimesters of pregnancy. However, its diagnostic performance was limited, showing only a modest sensitivity and specificity (287).

Another hormone of interest is PL. Research has shown that during the third trimester, PL mRNA levels are increased in the plasma of women with invasive placentation (288). These results are in agreement with the study from Jing et al., which confirmed that maternal plasma PL mRNA concentrations were significantly increased in PAS cases compared to controls (289).

In addition to protein hormones, alpha-fetoprotein (AFP), a glycoprotein produced by the fetal liver and transported to maternal serum through the placenta or by diffusion across fetal membranes, has also emerged as a potential biomarker for PAS. A retrospective case-control study found increased levels of AFP in the PAS group during the second trimester, with a sensitivity of 71% and a specificity of 46% (290). Similar results were obtained in a subsequent study, which demonstrated a positive association between elevated second-trimester serum AFP and placenta accreta, suggesting its potential use as a screening tool for pregnancy at high risk of PAS (291).

Growth factors biomarkers - PAS is characterized by an excessive neovascularization, thus angiogenic factors have also been studied as potential biomarkers. A study found significantly increased maternal serum PlGF levels in PAS cases compared to the healthy group during the third trimester, while no differences were found in serum levels of VEGF (178). In the same study, PlGF is elevated in maternal serum and the placental bed of patients with PAS disorders compared to those without PAS (178).

In contrast, studies investigating sFlt-1 levels have yielded less conclusive results. One study reported no association between third-trimester sFlt-1 levels and PAS disorders (292). Another study suggested that although sFlt-1 and PlGF expression slightly differ depending on the depth of placental invasion, there is no direct correlation between serum levels and PlGF and sFlt-1 expression in the placenta (293).

Although these biomarkers show potential for detecting adverse pregnancy outcomes, large-scale studies are needed to conclude whether they could be used in early screening for pregnancy complications such as IUGR, PE, GDM, and PAS.

6 Future research directions

There is still much to learn about how the placental hormones and metabolites contribute to maintaining basal and activity-dependent perfusion of the uterine circulation during pregnancy. Therefore, it is critical for basic science and clinical studies to elucidate the precise molecular mechanisms by which hormones, growth factors, and proteins secreted by the placenta modulate uterine vascular adaptation to pregnancy. A deeper understanding of these pathways is essential to inform clinical strategies for early diagnosis, risk stratification, and targeted therapy. Placental hormones and metabolites represent promising therapeutic targets for the prevention and treatment of pregnancy complications, including PE and IUGR. Integrative approaches combining translational models, omics technologies, and longitudinal clinical studies will be critical to unlocking their potential and improving maternal and fetal outcomes.

7 Conclusion

The endocrine function of the placenta is central to the hemodynamic adaptations of the uterine circulation during pregnancy. The placental hormones and metabolites appear to have a substantial effect on the uterine circulation, including altering ECs vasodilator production, decreasing vascular resistance, and promoting the remodeling of the UA. Placental hormones and metabolites also promote the angiogenic process, ultimately contributing to the increase in UPBF necessary to sustain normal fetal growth. Most of these changes occur in the first trimester of pregnancy, a crucial period where abnormalities in the secretion of placental hormones and metabolites may reflect early signs of placental and uterine vascular impairment. Understanding how the secretion of placental hormones and metabolites is altered during gestation, both under normal and pathological conditions, is important for the treatment and prevention of pregnancy complications.

Author contributions

ME: Conceptualization, Writing – original draft. LP: Validation, Conceptualization, Writing – review & editing. MM: Supervision, Writing – review & editing, Conceptualization.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This work was funded by the Next Generation EU-Italian NRRP, Mission 4, Component 2, Investment 1.5, call for the creation and strengthening of ‘Innovation Ecosystems’, building “Territorial R&D Leaders (Directorial Decree n. 2021/3277)—project Tech4You-Technologies for climate change adaptation and quality of life improvement”, no. ECS0000009. This work reflects only the authors’ views and opinions; neither the Ministry for University and Research nor the European Commission can be considered responsible for them.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

ADAM-12, A-disintegrin and metalloprotease-12; CRH, Corticotropin-releasing hormone; cGMP, Cyclic guanosine monophosphate; CTBs, Cytotrophoblasts; ECs, Endothelial cells; ER-α, ER –β, Estrogen receptor-α, -β; EVTs, Extravillous trophoblasts; GPER, G-protein coupled estrogen receptor; GH, Gestational hypertension; GDM, Gestational diabetes mellitus; hCG, Human chorionic gonadotropin; IGFs, Insulin growth factors; IUGR, Intrauterine growth restriction; LNPs, Lipid nanoparticles; MUA, Main uterine artery; MMPs, Metalloproteinases; NPY, Neuropeptide Y; NO, Nitric oxide; PTHrP, Parathyroid hormone-related protein; PlGF, Placental growth factor; PGH, Placental growth hormone; PL, Placental lactogen; PP13, Placental protein 13; PE, Preeclampsia; PAPP-A, Pregnancy-Associated Plasma Protein A; PSGs, Pregnancy-specific β-glycoproteins; PGI2, Prostaglandin I2; RUA, Radial uterine artery; SGA, Small for gestational age; VSMCs, Vascular smooth muscle cells; sEng, Soluble endoglin; sFlt-1, Soluble fms-like tyrosine kinase; UVECs, Umbilical vein endothelial cells; UAA, Uterine arcuate artery; UAECs, Uterine arterial endothelial cells; UA, Uterine artery; UBF, Uterine blood flow; UMVECs, Uterine microvascular endothelial cells; UPBF, Uteroplacental blood flow; UV, Uterine vein; VEGF, Vascular endothelial growth factor.

References

1. Osol G and Mandala M. Maternal uterine vascular remodeling during pregnancy. Physiology. (2009) 24:58–71. doi: 10.1152/physiol.00033.2008

PubMed Abstract | Crossref Full Text | Google Scholar

2. Burton GJ, Woods AW, Jauniaux E, and Kingdom JCP. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta. (2009) 30:473–82. doi: 10.1016/j.placenta.2009.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

3. Carter AM. Placental oxygen consumption. Part I: in vivo studies–a review. Placenta. (2000) 21 Suppl A:S31–7. doi: 10.1053/plac.1999.0513

PubMed Abstract | Crossref Full Text | Google Scholar

4. Karvaly G, Kovács K, Gyarmatig M, Gerszi D, Nagy S, Jalal DA, et al. Reference data on estrogen metabolome in healthy pregnancy. Mol Cell Probes. (2024) 74:101953. doi: 10.1016/j.mcp.2024.101953

PubMed Abstract | Crossref Full Text | Google Scholar

5. Morales DE, McGowan KA, Grant DS, Maheshwari S, Bhartiya D, Cid MC, et al. Estrogen promotes angiogenic activity in human umbilical vein endothelial cells in vitro and in a murine model. Circulation. (1995) 91:755–63. doi: 10.1161/01.CIR.91.3.755

PubMed Abstract | Crossref Full Text | Google Scholar

6. Jobe SO, Ramadoss J, Koch JM, Jiang Y, Zheng J, and Ronald R. Estradiol-17beta and its cytochrome P450- and catechol-O-methyltransferase-derived metabolites stimulate proliferation in uterine artery endothelial cells: role of estrogen receptor-alpha versus estrogen receptor-beta. Hypertension. (2010) 55:1005–11. doi: 10.1161/HYPERTENSIONAHA.109.146399

PubMed Abstract | Crossref Full Text | Google Scholar

7. Oviedo PJ, Sobrino A, Laguna-Fernandez A, Novella S, Tarín JJ, García-Pérez MA, et al. Estradiol induces endothelial cell migration and proliferation through estrogen receptor-enhanced RhoA/ROCK pathway. Mol Cell Endocrinol. (2011) 335:96–103. doi: 10.1016/j.mce.2010.06.020

PubMed Abstract | Crossref Full Text | Google Scholar

8. Cid MC, Schnaper HW, and Kleinman HK. Estrogens and the vascular endothelium. Ann N Y Acad Sci. (2002) 966:143–57. doi: 10.1111/j.1749-6632.2002.tb04211.x

PubMed Abstract | Crossref Full Text | Google Scholar

9. He WH, Jin MM, Liu AP, Zhou Y, Hu XL, Zhu YM, et al. Estradiol promotes trophoblast viability and invasion by activating SGK1. BioMed Pharmacother. (2019) 117:109092. doi: 10.1016/j.biopha.2019.109092

PubMed Abstract | Crossref Full Text | Google Scholar

10. Albrecht ED, Bonagura TW, Burleigh DW, Enders AC, Aberdeen GW, and Pepe GJ. Suppression of extravillous trophoblast invasion of uterine spiral arteries by estrogen during early baboon pregnancy. Placenta. (2006) 27:483–90. doi: 10.1016/j.placenta.2005.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

11. Dang Y, Li W, Tran V, and Khalil RA. EMMPRIN-mediated induction of uterine and vascular matrix metalloproteinases during pregnancy and in response to estrogen and progesterone. Biochem Pharmacol. (2013) 86:734–47. doi: 10.1016/j.bcp.2013.06.030

PubMed Abstract | Crossref Full Text | Google Scholar

12. Bonagura TW, Pepe GJ, Enders AC, and Albrecht ED. Suppression of extravillous trophoblast vascular endothelial growth factor expression and uterine spiral artery invasion by estrogen during early baboon pregnancy. Endocrinology. (2008) 149:5078–87. doi: 10.1210/en.2008-0116

PubMed Abstract | Crossref Full Text | Google Scholar

13. Storment JM, Meyer M, and Osol G. Estrogen augments the vasodilatory effects of vascular endothelial growth factor in the uterine circulation of the rat. Am J Obstet Gynecol. (2000) 183:449–53. doi: 10.1067/mob.2000.105910

PubMed Abstract | Crossref Full Text | Google Scholar

14. Scott PA, Tremblay A, Brochu M, and St-Louis J. Vasorelaxant action of 17 -estradiol in rat uterine arteries: role of nitric oxide synthases and estrogen receptors. Am J Physiol Heart Circ Physiol. (2007) 293:H3713–9. doi: 10.1152/ajpheart.00736.2007

PubMed Abstract | Crossref Full Text | Google Scholar

15. Corcoran JJ, Nicholson C, Sweeney M, Charnock JC, Robson SC, Westwood M, et al. Human uterine and placental arteries exhibit tissue-specific acute responses to 17β-estradiol and estrogen-receptor-specific agonists. Mol Hum Reprod. (2014) 20:433–41. doi: 10.1093/molehr/gat095

PubMed Abstract | Crossref Full Text | Google Scholar

16. Tropea T, Rigiracciolo D, Esposito M, Maggiolini M, and Mandalà M. G-protein-coupled estrogen receptor expression in rat uterine artery is increased by pregnancy and induces dilation in a Ca2+ and ERK1/2 dependent manner. Int J Mol Sci. (2022) 23:5996. doi: 10.3390/ijms23115996

PubMed Abstract | Crossref Full Text | Google Scholar

17. Rosenfeld CR, Roy T, and Cox BE. Mechanisms modulating estrogen-induced uterine vasodilation. Vascul Pharmacol. (2002) 38:115–25. doi: 10.1016/S0306-3623(02)00135-0

PubMed Abstract | Crossref Full Text | Google Scholar

18. Bai J, Qi QR, Li Y, Day R, Makhoul J, Magness RR, et al. Estrogen receptors and estrogen-induced uterine vasodilation in pregnancy. Int J Mol Sci. (2020) 21:4349. doi: 10.3390/ijms21124349

PubMed Abstract | Crossref Full Text | Google Scholar

19. Pastore MB, Jobe SO, Ramadoss J, and Magness RR. Estrogen receptor-α and estrogen receptor-β in the uterine vascular endothelium during pregnancy: functional implications for regulating uterine blood flow. Semin Reprod Med. (2012) 30:46–61. doi: 10.1055/s-0031-1299597

PubMed Abstract | Crossref Full Text | Google Scholar

20. Tropea T, De Francesco EM, Rigiracciolo D, Maggiolini M, Wareing M, Osol G, et al. Pregnancy Augments G Protein Estrogen Receptor (GPER) Induced Vasodilation in Rat Uterine Arteries via the Nitric Oxide - cGMP Signaling Pathway. PloS One. (2015) 10:e0141997. doi: 10.1371/journal.pone.0141997

PubMed Abstract | Crossref Full Text | Google Scholar

21. Mandalà M. Influence of estrogens on uterine vascular adaptation in normal and preeclamptic pregnancies. Int J Mol Sci. (2020) 21:2592. doi: 10.3390/ijms21072592

PubMed Abstract | Crossref Full Text | Google Scholar

22. Albrecht ED and Pepe GJ. Placental steroid hormone biosynthesis in primate pregnancy. Endocr Rev. (1990) 11:124–50. doi: 10.1210/edrv-11-1-124

PubMed Abstract | Crossref Full Text | Google Scholar

23. Neumann K, Depenbusch M, Schultze-Mosgau A, and Griesinger G. Strong variation in progesterone production of the placenta in early pregnancy - what are the clinical implications? Reprod BioMed Online. (2020) 41:748–9. doi: 10.1016/j.rbmo.2020.07.009

PubMed Abstract | Crossref Full Text | Google Scholar

24. Taraborrelli S. Physiology, production and action of progesterone. Acta Obstet Gynecol Scand. (2015) 94 Suppl 161:8–16. doi: 10.1111/aogs.12771

PubMed Abstract | Crossref Full Text | Google Scholar

25. Rupnow HL, Phernetton TM, Shaw CE, Modrick ML, Bird IM, and Magness RR. Endothelial vasodilator production by uterine and systemic arteries. VII. Estrogen and progesterone effects on eNOS. Am J Physiol Heart Circ Physiol. (2001) 280:H1699–705. doi: 10.1152/ajpheart.2001.280.4.H1699

PubMed Abstract | Crossref Full Text | Google Scholar

26. Hermenegildo C, Oviedo PJ, García-Martínez MC, García-Pérez MA, Tarín JJ, and Cano A. Progestogens stimulate prostacyclin production by human endothelial cells. Hum Reprod. (2005) 20:1554–61. doi: 10.1093/humrep/deh803

PubMed Abstract | Crossref Full Text | Google Scholar

27. Rupnow HL, Phernetton TM, Modrick ML, Wiltbank MC, Bird IM, and Magness RR. Endothelial vasodilator production by uterine and systemic arteries. VIII. Estrogen and progesterone effects on cPLA2, COX-1, and PGIS protein expression1. Biol Reprod. (2002) 66:468–74. doi: 10.1095/biolreprod66.2.468

PubMed Abstract | Crossref Full Text | Google Scholar

28. Xiao D, Huang X, Yang S, and Zhang L. Direct chronic effect of steroid hormones in attenuating uterine arterial myogenic tone: role of protein kinase c/extracellular signal-regulated kinase 1/2. Hypertension. (2009) 54:352–8. doi: 10.1161/HYPERTENSIONAHA.109.130781

PubMed Abstract | Crossref Full Text | Google Scholar

29. Conrad KP. Emerging role of relaxin in the maternal adaptations to normal pregnancy: implications for preeclampsia. Semin Nephrol. (2011) 31:15–32. doi: 10.1016/j.semnephrol.2010.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

30. Marshall SA, Ng L, Unemori EN, Girling JE, and Parry LJ. Relaxin deficiency results in increased expression of angiogenesis- and remodelling-related genes in the uterus of early pregnant mice but does not affect endometrial angiogenesis prior to implantation. Reprod Biol Endocrinol. (2016) 14:11. doi: 10.1186/s12958-016-0148-y

PubMed Abstract | Crossref Full Text | Google Scholar

31. Unemori EN, Lewis M, Constant J, Arnold G, Grove BH, and Normand J. Relaxin induces vascular endothelial growth factor expression and angiogenesis selectively at wound sites. Wound Repair Regener. (2000) 8:361–70. doi: 10.1111/j.1524-475X.2000.00361.x

PubMed Abstract | Crossref Full Text | Google Scholar

32. Kaftanovskaya EM, Huang Z, Lopez C, Conrad K, and Agoulnik AI. Conditional deletion of the relaxin receptor gene in cells of smooth muscle lineage affects lower reproductive tract in pregnant mice. Biol Reprod. (2015) 92:91. doi: 10.1095/biolreprod.114.127209

PubMed Abstract | Crossref Full Text | Google Scholar

33. Gooi J, Richardson ML, Jelinic M, Girling JE, Wlodek ME, Tare M, et al. Enhanced uterine artery stiffness in aged pregnant relaxin mutant mice is reversed with exogenous relaxin treatment. Biol Reprod. (2013) 89. doi: 10.1095/biolreprod.113.108118

PubMed Abstract | Crossref Full Text | Google Scholar

34. Vodstrcil LA, Tare M, Novak J, Dragomir N, Ramirez RJ, Wlodek ME, et al. Relaxin mediates uterine artery compliance during pregnancy and increases uterine blood flow. FASEB J. (2012) 26:4035–44. doi: 10.1096/fj.12-210567

PubMed Abstract | Crossref Full Text | Google Scholar

35. Longo M, Jain V, Vedernikov YP, Garfield RE, and Saade GR. Effects of recombinant human relaxin on pregnant rat uterine artery and myometrium in vitro. Am J Obstet Gynecol. (2003) 188:1468–74. doi: 10.1067/mob.2003.454

PubMed Abstract | Crossref Full Text | Google Scholar

36. McGuane JT, Debrah JE, Sautina L, Jarajapu YPR, Novak J, Rubin JP, et al. Relaxin induces rapid dilation of rodent small renal and human subcutaneous arteries via PI3 kinase and nitric oxide. Endocrinology. (2011) 152:2786–96. doi: 10.1210/en.2010-1126

PubMed Abstract | Crossref Full Text | Google Scholar

37. Debrah DO, Novak J, Matthews JE, Ramirez RJ, Shroff SG, and Conrad KP. Relaxin is essential for systemic vasodilation and increased global arterial compliance during early pregnancy in conscious rats. Endocrinology. (2006) 147:5126–31. doi: 10.1210/en.2006-0567

PubMed Abstract | Crossref Full Text | Google Scholar

38. Evans PW, Wheeler T, Anthony FW, and Osmond C. A longitudinal study of maternal serum vascular endothelial growth factor in early pregnancy. Hum Reprod. (1998) 13:1057–62. doi: 10.1093/humrep/13.4.1057

PubMed Abstract | Crossref Full Text | Google Scholar

39. Wheeler T, Evans PW, Anthony FW, Godfrey KM, Howe DT, and Osmond C. Relationship between maternal serum vascular endothelial growth factor concentration in early pregnancy and fetal and placental growth. Hum Reprod. (1999) 14:1619–23. doi: 10.1093/humrep/14.6.1619

PubMed Abstract | Crossref Full Text | Google Scholar

40. Zamudio S, Kovalenko O, Echalar L, Torricos T, Al-Khan A, Alvarez M, et al. Evidence for extraplacental sources of circulating angiogenic growth effectors in human pregnancy. Placenta. (2013) 34:1170–6. doi: 10.1016/j.placenta.2013.09.016

PubMed Abstract | Crossref Full Text | Google Scholar

41. Charnock-Jones DS, Sharkey AM, Rajput-Williams J, Burch D, Schofield JP, Fountain SA, et al. Identification and localization of alternately spliced mRNAs for vascular endothelial growth factor in human uterus and estrogen regulation in endometrial carcinoma cell lines. . Biol Reprod. (1993) 48:1120–8. doi: 10.1095/biolreprod48.5.1120

PubMed Abstract | Crossref Full Text | Google Scholar

42. Anthony FW, Wheeler T, Elcock CL, Pickett M, and Thomas EJ. Short report: identification of a specific pattern of vascular endothelial growth factor mRNA expression in human placenta and cultured placental fibroblasts. Placenta. (1994) 15:557–61. doi: 10.1016/S0143-4004(05)80424-2

PubMed Abstract | Crossref Full Text | Google Scholar

43. Cullinan-Bove K and Koos RD. Vascular endothelial growth factor/vascular permeability factor expression in the rat uterus: rapid stimulation by estrogen correlates with estrogen-induced increases in uterine capillary permeability and growth. Endocrinology. (1993) 133:829–37. doi: 10.1210/endo.133.2.8344219

PubMed Abstract | Crossref Full Text | Google Scholar

44. Celia G and Osol G. Uterine venous permeability in the rat is altered in response to pregnancy, vascular endothelial growth factor, and venous constriction. Endothelium. (2005) 12:81–8. doi: 10.1080/10623320590933879

PubMed Abstract | Crossref Full Text | Google Scholar

45. Roberts WG and Palade GE. Increased microvascular permeability and endothelial fenestration induced by vascular endothelial growth factor. J Cell Sci. (1995) 108:2369–79. doi: 10.1242/jcs.108.6.2369

PubMed Abstract | Crossref Full Text | Google Scholar

46. Pepper MS, Ferrara N, Orci L, and Montesano R. Vascular endothelial growth factor (VEGF) induces plasminogen activators and plasminogen activator inhibitor-1 in microvascular endothelial cells. Biochem Biophys Res Commun. (1991) 181:902–6. doi: 10.1016/0006-291X(91)91276-I

PubMed Abstract | Crossref Full Text | Google Scholar

47. Unemori EN, et al. Vascular endothelial growth factor induces interstitial collagenase expression in human endothelial cells. J Cell Physiol. (1992) 153:557–62. doi: 10.1002/jcp.1041530317

PubMed Abstract | Crossref Full Text | Google Scholar

48. Zhang HH, Chen JC, Sheibani L, Lechuga TJ, and Chen DB. Pregnancy augments VEGF-stimulated in vitro angiogenesis and vasodilator (NO and H2S) production in human uterine artery endothelial cells. J Clin Endocrinol Metab. (2017) 102:2382–93. doi: 10.1210/jc.2017-00437

PubMed Abstract | Crossref Full Text | Google Scholar

49. Wheeler-Jones C, Abu-Ghazaleh R, Cospedal R, Houliston RA, Martin J, and Zachary I. Vascular endothelial growth factor stimulates prostacyclin production and activation of cytosolic phospholipase A2 in endothelial cells via p42/p44 mitogen-activated protein kinase. FEBS Lett. (1997) 420:28–32. doi: 10.1016/S0014-5793(97)01481-6

PubMed Abstract | Crossref Full Text | Google Scholar

50. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, et al. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. (1995) 376:62–6. doi: 10.1038/376062a0

PubMed Abstract | Crossref Full Text | Google Scholar

51. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. (1996) 380:435–9. doi: 10.1038/380435a0

PubMed Abstract | Crossref Full Text | Google Scholar

52. David AL, Torondel B, Zachary I, Wigley V, Nader KA, Mehta V, et al. Local delivery of VEGF adenovirus to the uterine artery increases vasorelaxation and uterine blood flow in the pregnant sheep. Gene Ther. (2008) 15:1344–50. doi: 10.1038/gt.2008.102

PubMed Abstract | Crossref Full Text | Google Scholar

53. Mehta V, et al. Long-term increase in uterine blood flow is achieved by local overexpression of VEGF-A(165) in the uterine arteries of pregnant sheep. Gene Ther. (2012) 19:925–35. doi: 10.1038/gt.2011.158

PubMed Abstract | Crossref Full Text | Google Scholar

54. Mehta V, Abi-Nader KN, Shangaris P, Shaw SWS, Filippi E, Benjamin E, et al. Local over-expression of VEGF-DΔNΔC in the uterine arteries of pregnant sheep results in long-term changes in uterine artery contractility and angiogenesis. PloS One. (2014) 9:e100021. doi: 10.1371/journal.pone.0100021

PubMed Abstract | Crossref Full Text | Google Scholar

55. Ni Y, May V, Braas K, and Osol G. Pregnancy augments uteroplacental vascular endothelial growth factor gene expression and vasodilator effects. Am J Physiol. (1997) 273:H938–44. doi: 10.1152/ajpheart.1997.273.2.H938

PubMed Abstract | Crossref Full Text | Google Scholar

56. Dewerchin M and Carmeliet P. PlGF: a multitasking cytokine with disease-restricted activity. Cold Spring Harb Perspect Med. (2012) 2. doi: 10.1101/cshperspect.a011056

PubMed Abstract | Crossref Full Text | Google Scholar

57. Saffer C, Olson G, Boggess KA, Beyerlein R, Eubank C, and Sibai BM. Determination of placental growth factor (PlGF) levels in healthy pregnant women without signs or symptoms of preeclampsia. Pregnancy Hypertens. (2013) 3:124–32. doi: 10.1016/j.preghy.2013.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

58. Vuorela P, Hatva E, Lymboussaki A, Kaipainen A, Joukov V, Persico MG, et al. Expression of vascular endothelial growth factor and placenta growth factor in human placenta. Biol Reprod. (1997) 56:489–94. doi: 10.1095/biolreprod56.2.489

PubMed Abstract | Crossref Full Text | Google Scholar

59. Welch PC, Amankwah KS, Miller P, McAsey ME, and Torry DS. Correlations of placental perfusion and PlGF protein expression in early human pregnancy. Am J Obstet Gynecol. (2006) 194:1625–9. doi: 10.1016/j.ajog.2006.01.012

PubMed Abstract | Crossref Full Text | Google Scholar

60. Tayade C, Hilchie D, He H, Fang Y, Moons L, Carmeliet P, et al. Genetic deletion of placenta growth factor in mice alters uterine NK cells. J Immunol. (2007) 178:4267–75. doi: 10.4049/jimmunol.178.7.4267

PubMed Abstract | Crossref Full Text | Google Scholar

61. Park JE, Chen HH, Winer J, Houck KA, and Ferrara N. Placenta growth factor. Potentiation of vascular endothelial growth factor bioactivity, in vitro and in vivo, and high affinity binding to Flt-1 but not to Flk-1/KDR. J Biol Chem. (1994) 269:25646–54. doi: 10.1016/S0021-9258(18)47298-5

PubMed Abstract | Crossref Full Text | Google Scholar

62. Szukiewicz D, Szewczyk G, Watroba M, Kurowska E, and Maslinski S. Isolated placental vessel response to vascular endothelial growth factor and placenta growth factor in normal and growth-restricted pregnancy. Gynecologic Obstetric Invest. (2005) 59:102–7. doi: 10.1159/000082622

PubMed Abstract | Crossref Full Text | Google Scholar

63. Osol G, Celia G, Gokina N, Barron C, Chien E, Mandala M, et al. Placental growth factor is a potent vasodilator of rat and human resistance arteries. Am J Physiology-Heart Circulatory Physiol. (2008) 294:H1381–7. doi: 10.1152/ajpheart.00922.2007

PubMed Abstract | Crossref Full Text | Google Scholar

64. Curtis NE, King RG, Moseley JM, Ho PWM, Rice GE, and Wlodek ME. Intrauterine expression of parathyroid hormone-related protein in normal and pre-eclamptic pregnancies. Placenta. (1998) 19:595–601. doi: 10.1016/S0143-4004(98)90020-0

PubMed Abstract | Crossref Full Text | Google Scholar

65. Dunne FP, Ratcliffe W, Mansour P, and Heath DA. Parathyroid hormone related protein (PTHrP) gene expression in fetal and extra-embryonic tissues of early pregnancy. Hum Reprod. (1994) 9:149–56. doi: 10.1093/oxfordjournals.humrep.a138306

PubMed Abstract | Crossref Full Text | Google Scholar

66. Meziani F, Van Overloop B, Schneider F, and Gairard A. Parathyroid hormone-related protein-induced relaxation of rat uterine arteries: influence of the endothelium during gestation. J Soc Gynecol Investig. (2005) 12:14–9. doi: 10.1016/j.jsgi.2004.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

67. Macgill K, Moseley JM, Martin TJ, Brennecke SP, Rice GE, and Wlodek ME. Vascular effects of PTHrP (1-34) and PTH (1-34) in the human fetal-placental circulation. Placenta. (1997) 18:587–92. doi: 10.1016/0143-4004(77)90014-5

PubMed Abstract | Crossref Full Text | Google Scholar

68. Mandsager NT, Brewer AS, and Myatt L. Vasodilator effects of parathyroid hormone, parathyroid hormone-related protein, and calcitonin gene-related peptide in the human fetal-placental circulation. J Soc Gynecol Investig. (1994) 1:19–24. doi: 10.1177/107155769400100105

PubMed Abstract | Crossref Full Text | Google Scholar

69. Rashid G, Bernheim J, Green J, and Benchetrit S. Parathyroid hormone stimulates the endothelial nitric oxide synthase through protein kinase A and C pathways. Nephrol Dialysis Transplant. (2007) 22:2831–7. doi: 10.1093/ndt/gfm269

PubMed Abstract | Crossref Full Text | Google Scholar

70. Meziani F, Tesse A, Welsch S, Kremer H, Barthelmebs M, Andriantsitohaina R, et al. Expression and biological activity of parathyroid hormone-related peptide in pregnant rat uterine artery: any role for 8-iso-prostaglandin F2α? Endocrinology. (2008) 149:626–33. doi: 10.1210/en.2007-0568

PubMed Abstract | Crossref Full Text | Google Scholar

71. Wlodek ME, Di Nicolantonio R, Westcott KT, Farrugia W, Ho PWM, and Moseley JM. PTH/PTHrP receptor and mid-molecule PTHrP regulation of intrauterine PTHrP: PTH/PTHrP receptor antagonism increases SHR fetal weight. Placenta. (2004) 25:53–61. doi: 10.1016/j.placenta.2003.08.001

PubMed Abstract | Crossref Full Text | Google Scholar

72. Han VK, Bassett N, Walton J, and Challis JR. The expression of insulin-like growth factor (IGF) and IGF-binding protein (IGFBP) genes in the human placenta and membranes: evidence for IGF-IGFBP interactions at the feto-maternal interface. J Clin Endocrinol Metab. (1996) 81:2680–93. doi: 10.1210/jcem.81.7.8675597

PubMed Abstract | Crossref Full Text | Google Scholar

73. Herr F, Liang OD, Herrero J, Lang U, Preissner KT, Han VKM, et al. Possible angiogenic roles of insulin-like growth factor II and its receptors in uterine vascular adaptation to pregnancy. J Clin Endocrinol Metab. (2003) 88:4811–7. doi: 10.1210/jc.2003-030243

PubMed Abstract | Crossref Full Text | Google Scholar

74. DeChiara TM, Efstratiadis A, and Robertson EJ. A growth-deficiency phenotype in heterozygous mice carrying an insulin-like growth factor II gene disrupted by targeting. Nature. (1990) 345:78–80. doi: 10.1038/345078a0

PubMed Abstract | Crossref Full Text | Google Scholar

75. Qin Q-P, Christiansen M, Oxvig C, Pettersson K, Sottrup-Jensen L, Koch C, et al. Double-monoclonal immunofluorometric assays for pregnancy-associated plasma protein A/proeosinophil major basic protein (PAPP-A/proMBP) complex in first-trimester maternal serum screening for Down syndrome. Clin Chem. (1997) 43:2323–32. doi: 10.1093/clinchem/43.12.2323

PubMed Abstract | Crossref Full Text | Google Scholar

76. Monget P and Oxvig C. PAPP-A and the IGF system. Ann Endocrinol (Paris). (2016) 77:90–6. doi: 10.1016/j.ando.2016.04.015

PubMed Abstract | Crossref Full Text | Google Scholar

77. Balcı S. Predictive values of maternal serum PAPP-A level, uterine artery Doppler velocimetry, and fetal biometric measurements for poor pregnancy and poor neonatal outcomes in pregnant women. J Turk Ger Gynecol Assoc. (2016) 17:143–9. doi: 10.5152/jtgga.2016.16040

PubMed Abstract | Crossref Full Text | Google Scholar

78. Lei ZM, Reshef E, and Rao V. The expression of human chorionic gonadotropin/luteinizing hormone receptors in human endometrial and myometrial blood vessels. J Clin Endocrinol Metab. (1992) 75:651–9. doi: 10.1210/jcem.75.2.1379262

PubMed Abstract | Crossref Full Text | Google Scholar

79. Toth P, Li X, Rao CV, Lincoln SR, Sanfilippo JS, Spinnato JA, et al. Expression of functional human chorionic gonadotropin/human luteinizing hormone receptor gene in human uterine arteries. J Clin Endocrinol Metab. (1994) 79:307–15. doi: 10.1210/jcem.79.1.8027246

PubMed Abstract | Crossref Full Text | Google Scholar

80. Zygmunt M, Herr F, Keller-Schoenwetter S, Kunzi-Rapp K, Münstedt K, Rao CV, et al. Characterization of human chorionic gonadotropin as a novel angiogenic factor. J Clin Endocrinol Metab. (2002) 87:5290–6. doi: 10.1210/jc.2002-020642

PubMed Abstract | Crossref Full Text | Google Scholar

81. Wulff C, Dickson SE, Duncan WC, and Fraser HM. Angiogenesis in the human corpus luteum: simulated early pregnancy by HCG treatment is associated with both angiogenesis and vessel stabilization. Hum Reprod. (2001) 16:2515–24. doi: 10.1093/humrep/16.12.2515

PubMed Abstract | Crossref Full Text | Google Scholar

82. Prast J, Saleh L, Husslein H, Sonderegger S, Helmer H, and Knöfler M. Human chorionic gonadotropin stimulates trophoblast invasion through extracellularly regulated kinase and AKT signaling. Endocrinology. (2008) 149:979–87. doi: 10.1210/en.2007-1282

PubMed Abstract | Crossref Full Text | Google Scholar

83. Hermsteiner M, Zoltan DR, Doetsch J, Rascher W, and Kuenzel W. Human chorionic gonadotropin dilates uterine and mesenteric resistance arteries in pregnant and nonpregnant rats. Pflügers Archiv. (1999) 439:186–94. doi: 10.1007/s004249900151

PubMed Abstract | Crossref Full Text | Google Scholar

84. Vokalova L, Balogh A, Toth E, Van Breda SV, Schäfer G, Hoesli I, et al. Placental protein 13 (Galectin-13) polarizes neutrophils toward an immune regulatory phenotype. Front Immunol. (2020) 11:145. doi: 10.3389/fimmu.2020.00145

PubMed Abstract | Crossref Full Text | Google Scholar

85. Kliman HJ, Sammar M, Grimpel YI, Lynch SK, Milano KM, Pick E, et al. Placental protein 13 and decidual zones of necrosis: an immunologic diversion that may be linked to preeclampsia. Reprod Sci. (2012) 19:16–30. doi: 10.1177/1933719111424445

PubMed Abstract | Crossref Full Text | Google Scholar

86. Gizurarson S, Sigurdardottir ER, Meiri H, Huppertz B, Sammar M, Sharabi-Nov A, et al. Placental protein 13 administration to pregnant rats lowers blood pressure and augments fetal growth and venous remodeling. Fetal Diagn Ther. (2016) 39:56–63. doi: 10.1159/000381914

PubMed Abstract | Crossref Full Text | Google Scholar

87. Drobnjak T, Gizurarson S, Gokina NI, Meiri H, Mandalà M, Huppertz B, et al. Placental protein 13 (PP13)-induced vasodilation of resistance arteries from pregnant and nonpregnant rats occurs via endothelial-signaling pathways. Hypertens Pregnancy. (2017) 36:186–95. doi: 10.1080/10641955.2017.1295052

PubMed Abstract | Crossref Full Text | Google Scholar

88. Drobnjak T, Jónsdóttir AM, Helgadóttir H, Runólfsdóttir MS, Meiri H, Sammar M, et al. Placental protein 13 (PP13) stimulates rat uterine vessels after slow subcutaneous administration. Int J Womens Health. (2019) 11:213–22. doi: 10.2147/IJWH.S188303

PubMed Abstract | Crossref Full Text | Google Scholar

89. Gatto M, Esposito M, Gizurarson S, Henrion D, and Mandalà M. Placental protein 13 dilation of pregnant rat uterine vein is endothelium dependent and involves nitric oxide/calcium activated potassium channels signals. Placenta. (2022) 126:233–8. doi: 10.1016/j.placenta.2022.07.001

PubMed Abstract | Crossref Full Text | Google Scholar

90. Gatto M, Esposito M, Morelli M, De Rose S, Gizurarson S, Meiri H, et al. Placental protein 13: vasomodulatory effects on human uterine arteries and potential implications for preeclampsia. Int J Mol Sci. (2024) 25:7522. doi: 10.3390/ijms25147522

PubMed Abstract | Crossref Full Text | Google Scholar

91. Hayden TJ, Buttle HL, Rees PL, Smith SV, and Forsyth IA. Simultaneous determinations of uterine blood flow and plasma concentrations of placental lactogen in late-pregnant goats. J Reprod Fertil. (1983) 69:503–10. doi: 10.1530/jrf.0.0690503

PubMed Abstract | Crossref Full Text | Google Scholar

92. Houghton DJ, Shackleton P, Obiekwe BC, and Chard T. Relationship of maternal and fetal levels of human placental lactogen to the weight and sex of the fetus. Placenta. (1984) 5:455–8. doi: 10.1016/S0143-4004(84)80026-0

PubMed Abstract | Crossref Full Text | Google Scholar

93. Baker CM, Goetzmann LN, Cantlon JD, Jeckel KM, Winger QA, and Anthony RV. Development of ovine chorionic somatomammotropin hormone-deficient pregnancies. Am J Physiology-Regulatory Integr Comp Physiol. (2016) 310:R837–46. doi: 10.1152/ajpregu.00311.2015

PubMed Abstract | Crossref Full Text | Google Scholar

94. Struman I, Bentzien F, Lee H, Mainfroid V, D’Angelo G, Goffin V, et al. Opposing actions of intact and N-terminal fragments of the human prolactin/growth hormone family members on angiogenesis: an efficient mechanism for the regulation of angiogenesis. Proc Natl Acad Sci U.S.A. (1999) 96:1246–51. doi: 10.1073/pnas.96.4.1246

PubMed Abstract | Crossref Full Text | Google Scholar

95. Lacroix M-C, Guibourdenche J, Fournier T, Laurendeau I, Igout A, Goffin V, et al. Stimulation of human trophoblast invasion by placental growth hormone. Endocrinology. (2005) 146:2434–44. doi: 10.1210/en.2004-1550

PubMed Abstract | Crossref Full Text | Google Scholar

96. Caufriez A, Frankenne F, Hennen G, and Copinschi G. Regulation of maternal IGF-I by placental GH in normal and abnormal human pregnancies. Am J Physiol. (1993) 265:E572–7. doi: 10.1152/ajpendo.1993.265.4.E572

PubMed Abstract | Crossref Full Text | Google Scholar

97. Koutsaki M, Sifakis S, Zaravinos A, Koutroulakis D, Koukoura O, and Spandidos DA. Decreased placental expression of hPGH, IGF-I and IGFBP-1 in pregnancies complicated by fetal growth restriction. Growth Hormone IGF Res. (2011) 21:31–6. doi: 10.1016/j.ghir.2010.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

98. Chellakooty M, Vangsgaard K, Larsen T, Scheike T, Falck-Larsen J, Legarth J, et al. A longitudinal study of intrauterine growth and the placental growth hormone (GH)-insulin-like growth factor I axis in maternal circulation: association between placental GH and fetal growth. J Clin Endocrinol Metab. (2004) 89:384–91. doi: 10.1210/jc.2003-030282

PubMed Abstract | Crossref Full Text | Google Scholar

99. Masuzaki H, Ogawa Y, Sagawa N, Hosoda K, Matsumoto T, Mise H, et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat Med. (1997) 3:1029–33. doi: 10.1038/nm0997-1029

PubMed Abstract | Crossref Full Text | Google Scholar

100. Chehab FF, Lim ME, and Lu R. Correction of the sterility defect in homozygous obese female mice by treatment with the human recombinant leptin. Nat Genet. (1996) 12:318–20. doi: 10.1038/ng0396-318

PubMed Abstract | Crossref Full Text | Google Scholar

101. Malik NM, Carter ND, Murray JF, Scaramuzzi RJ, Wilson CA, and Stock MJ. Leptin requirement for conception, implantation, and gestation in the mouse. Endocrinology. (2001) 142:5198–202. doi: 10.1210/endo.142.12.8535

PubMed Abstract | Crossref Full Text | Google Scholar

102. Henson MC, Swan KF, and O’Neil JS. Expression of placental leptin and leptin receptor transcripts in early pregnancy and at term. Obstet Gynecol. (1998) 92:1020–8. doi: 10.1016/s0029-7844(98)00299-3

PubMed Abstract | Crossref Full Text | Google Scholar

103. Rumer KK, Sehgal S, Kramer A, Bogart KP, and Winn VD. The effects of leptin on human cytotrophoblast invasion are gestational age and dose-dependent. Front Endocrinol (Lausanne). (2024) 15:1386309. doi: 10.3389/fendo.2024.1386309

PubMed Abstract | Crossref Full Text | Google Scholar

104. Schulz LC and Widmaier EP. The effect of leptin on mouse trophoblast cell invasion1. Biol Reprod. (2004) 71:1963–7. doi: 10.1095/biolreprod.104.032722

PubMed Abstract | Crossref Full Text | Google Scholar

105. Castellucci M, De Matteis R, Meisser A, Cancello R, Monsurrò V, Islami D, et al. Leptin modulates extracellular matrix molecules and metalloproteinases: possible implications for trophoblast invasion. Mol Hum Reprod. (2000) 6:951–8. doi: 10.1093/molehr/6.10.951

PubMed Abstract | Crossref Full Text | Google Scholar

106. Vargas VE, Villalon Landeros R, Lopez GE, Zheng J, and Magness RR. Uterine artery leptin receptors during the ovarian cycle and pregnancy regulate angiogenesis in ovine uterine artery endothelial cells†. Biol Reprod. (2017) 96:866–76. doi: 10.1093/biolre/iox008

PubMed Abstract | Crossref Full Text | Google Scholar

107. Bouloumié A, Drexler HCA, Lafontan M, and Busse R. Leptin, the product of ob gene, promotes angiogenesis. Circ Res. (1998) 83:1059–66. doi: 10.1161/01.RES.83.10.1059

PubMed Abstract | Crossref Full Text | Google Scholar

108. Nakagawa K, Higashi Y, Sasaki S, Ohshima T, Matsuura H, and Kajiyama G. Leptin causes vasodilation in humans. Hypertension. (2000) 36:725–5. doi: 10.1161/hyp.36.suppl_1.725-a

Crossref Full Text | Google Scholar

109. Riley SC, Walton JC, Herlick JM, and Challis JRG. The localization and distribution of corticotropin-releasing hormone in the human placenta and fetal membranes throughout gestation. J Clin Endocrinol Metab. (1991) 72:1001–7. doi: 10.1210/jcem-72-5-1001

PubMed Abstract | Crossref Full Text | Google Scholar

110. Clifton VL, Read MA, Leitch IM, Boura AL, Robinson PJ, and Smith R. Corticotropin-releasing hormone-induced vasodilatation in the human fetal placental circulation. J Clin Endocrinol Metab. (1994) 79:666–9. doi: 10.1210/jcem.79.2.8045990

PubMed Abstract | Crossref Full Text | Google Scholar

111. Jain V, Vedernikov YP, Saade GR, Chwalisz K, and Garfield RE. Endothelium-dependent and -independent mechanisms of vasorelaxation by corticotropin-releasing factor in pregnant rat uterine artery. J Pharmacol Exp Ther. (1999) 288:407–13. doi: 10.1016/S0022-3565(24)37970-4

PubMed Abstract | Crossref Full Text | Google Scholar

112. Bamberger AM, Minas V, Kalantaridou SN, Radde J, Sadeghian H, Löning T, et al. Corticotropin-releasing hormone modulates human trophoblast invasion through carcinoembryonic antigen-related cell adhesion molecule-1 regulation. Am J Pathol. (2006) 168:141–50. doi: 10.2353/ajpath.2006.050167

PubMed Abstract | Crossref Full Text | Google Scholar

113. Fried G and Samuelson U. Endothelin and neuropeptide Y are vasoconstrictors in human uterine blood vessels. Am J Obstet Gynecol. (1991) 164:1330–6. doi: 10.1016/0002-9378(91)90709-Z

PubMed Abstract | Crossref Full Text | Google Scholar

114. Jovanovic S, Grbovic L, and Jovanovic A. Pregnancy is associated with altered response to neuropeptide Y in uterine artery. Mol Hum Reprod. (2000) 6:352–60. doi: 10.1093/molehr/6.4.352

PubMed Abstract | Crossref Full Text | Google Scholar

115. Lee EW, Michalkiewicz M, Kitlinska J, Kalezic I, Switalska H, Yoo P, et al. Neuropeptide Y induces ischemic angiogenesis and restores function of ischemic skeletal muscles. J Clin Invest. (2003) 111:1853–62. doi: 10.1172/JCI16929

PubMed Abstract | Crossref Full Text | Google Scholar

116. Taylor JC, Laughlin MH, and Terjung RL. Neuropeptide Y (NPY) Y2 receptor (Y2R) mediates dilation in peripheral collateral vessels. FASEB J. (2007) 21:A1213–3. doi: 10.1096/fasebj.21.6.A1213-a

Crossref Full Text | Google Scholar

117. Klinjampa R, Sitticharoon C, Souvannavong-Vilivong X, Sripong C, Keadkraichaiwat I, Churintaraphan M, et al. Placental Neuropeptide Y (NPY) and NPY receptors expressions and serum NPY levels in preeclampsia. Exp Biol Med (Maywood). (2019) 244:380–8. doi: 10.1177/1535370219831437

PubMed Abstract | Crossref Full Text | Google Scholar

118. Sharabi-Nov A, Premru Sršen T, Kumer K, Vodušek VF, Fabjan T, Tul N, et al. Maternal Serum Inhibin-A Augments the Value of Maternal Serum PlGF and of sFlt-1/PlGF Ratio in the Prediction of Preeclampsia and/or FGR Near Delivery—A Secondary Analysis. Reprod Med. (2021) 2:35–49. doi: 10.3390/reprodmed2010005

Crossref Full Text | Google Scholar

119. Singh P, Jenkins LM, Horst B, Alers V, Pradhan S, Kaur P, et al. Inhibin is a novel paracrine factor for tumor angiogenesis and metastasis. Cancer Res. (2018) 78:2978–89. doi: 10.1158/0008-5472.CAN-17-2316

PubMed Abstract | Crossref Full Text | Google Scholar

120. O’Connor AE, McFarlane JR, Hayward S, Yohkaichiya T, Groome NP, and de Kretser DM. Serum activin A and follistatin concentrations during human pregnancy: a cross-sectional and longitudinal study. Hum Reprod. (1999) 14:827–32. doi: 10.1093/humrep/14.3.827

PubMed Abstract | Crossref Full Text | Google Scholar

121. Florio P, Ciarmela P, Luisi S, Palumbo MA, Lambert-Messerlian G, Severi FM, et al. Pre-eclampsia with fetal growth restriction: placental and serum activin A and inhibin A levels. Gynecological Endocrinol. (2002) 16:365–72. doi: 10.1080/gye.16.5.365.372

PubMed Abstract | Crossref Full Text | Google Scholar

122. He ZY, Liu HC, Mele CA, Barmat L, Veeck LL, Davis O, et al. Expression of inhibin/activin subunits and their receptors and binding proteins in human preimplantation embryos. J Assist Reprod Genet. (1999) 16:73–80. doi: 10.1023/A:1022564722353

PubMed Abstract | Crossref Full Text | Google Scholar

123. Zhu S, Li Z, Cui L, Ban Y, Leung PCK, Li Y, et al. Activin A increases human trophoblast invasion by upregulating integrin β1 through ALK4. FASEB J. (2021) 35:e21220. doi: 10.1096/fj.202001604R

PubMed Abstract | Crossref Full Text | Google Scholar

124. Sun F, Cheng L, Guo L, Su S, Li Y, and Yan J. Activin A promotes human trophoblast invasion by upregulating integrin β3 via ALK4-SMAD4 signaling. Placenta. (2022) 129:62–9. doi: 10.1016/j.placenta.2022.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

125. Lan X, Guo L, Hu C, Zhang Q, Deng J, Wang Y, et al. Fibronectin mediates activin A-promoted human trophoblast migration and acquisition of endothelial-like phenotype. Cell Communication Signaling. (2024) 22:61. doi: 10.1186/s12964-023-01463-z

PubMed Abstract | Crossref Full Text | Google Scholar

126. Li Y, Zhu H, Klausen C, Peng B, and Leung PCK. Vascular endothelial growth factor-A (VEGF-A) mediates activin A-induced human trophoblast endothelial-like tube formation. Endocrinology. (2015) 156:4257–68. doi: 10.1210/en.2015-1228

PubMed Abstract | Crossref Full Text | Google Scholar

127. Hobson SR, Acharya R, Lim R, Chan ST, Mockler J, and Wallace EM. Role of activin A in the pathogenesis of endothelial cell dysfunction in preeclampsia. Pregnancy Hypertension: Int J Women’s Cardiovasc Health. (2016) 6:130–3. doi: 10.1016/j.preghy.2016.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

128. Barber C, Yap Y, Hannan NJ, Wallace EM, and Marshall SA. Activin A causes endothelial dysfunction of mouse aorta and human aortic cells. Reproduction. (2022) 163:145–55. doi: 10.1530/REP-21-0368

PubMed Abstract | Crossref Full Text | Google Scholar

129. Moore T and Dveksler GS. Pregnancy-specific glycoproteins: complex gene families regulating maternal-fetal interactions. Int J Dev Biol. (2014) 58:273–80. doi: 10.1387/ijdb.130329gd

PubMed Abstract | Crossref Full Text | Google Scholar

130. Jia L, Huang X, Peng H, Jia Y, Zhang R, Wei Y, et al. Pregnancy-specific beta-1-glycoprotein 1-enriched exosomes are involved in the regulation of vascular endothelial cell function during pregnancy. Placenta. (2023) 139:138–47. doi: 10.1016/j.placenta.2023.06.014

PubMed Abstract | Crossref Full Text | Google Scholar

131. Qin Y, Meng Q, Wang Q, Wu M, Fang Y, Tu C, et al. Pregnancy-specific glycoprotein 9 enhances store-operated calcium entry and nitric oxide release in human umbilical vein endothelial cells. Diagnostics (Basel). (2023) 13:1134. doi: 10.3390/diagnostics13061134

PubMed Abstract | Crossref Full Text | Google Scholar

132. Ha CT, Wu JA, Irmak S, Lisboa FA, Dizon AM, Warren JW, et al. Human pregnancy specific beta-1-glycoprotein 1 (PSG1) has a potential role in placental vascular morphogenesis1. Biol Reprod. (2010) 83:27–35. doi: 10.1095/biolreprod.109.082412

PubMed Abstract | Crossref Full Text | Google Scholar

133. Turbeville HR and Sasser JM. Preeclampsia beyond pregnancy: long-term consequences for mother and child. Am J Physiol Renal Physiol. (2020) 318:F1315–f1326. doi: 10.1152/ajprenal.00071.2020

PubMed Abstract | Crossref Full Text | Google Scholar

134. Roberts JM, Rich-Edwards JW, McElrath TF, Garmire L, and Myatt L. Subtypes of preeclampsia: recognition and determining clinical usefulness. Hypertension. (2021) 77:1430–41. doi: 10.1161/HYPERTENSIONAHA.120.14781

PubMed Abstract | Crossref Full Text | Google Scholar

135. Brouwers L, de Gier S, Vogelvang TE, Veerbeek JHW, Franx A, van Rijn BB, et al. Prevalence of placental bed spiral artery pathology in preeclampsia and fetal growth restriction: A prospective cohort study. Placenta. (2024) 156:1–9. doi: 10.1016/j.placenta.2024.08.010

PubMed Abstract | Crossref Full Text | Google Scholar

136. Veerbeek JH, Brouwers L, Koster MPH, Koenen SV, van Vliet EOG, Nikkels PGJ, et al. Spiral artery remodeling and maternal cardiovascular risk: the spiral artery remodeling (SPAR) study. J Hypertens. (2016) 34:1570–7. doi: 10.1097/HJH.0000000000000964

PubMed Abstract | Crossref Full Text | Google Scholar

137. Shahid N, Masood M, Bano Z, Naz U, Hussain SF, Anwar A, et al. Role of uterine artery doppler ultrasound in predicting pre-eclampsia in high-risk women. Cureus. (2021) 13:e16276. doi: 10.7759/cureus.16276

PubMed Abstract | Crossref Full Text | Google Scholar

138. Litwinska M, Litwinska E, Lisnere K, Syngelaki A, Wright A, and Nicolaides KH. Stratification of pregnancy care based on risk of pre-eclampsia derived from uterine artery Doppler at 19–24 weeks’ gestation. Ultrasound Obstet Gynecol. (2021) 58:67–76. doi: 10.1002/uog.23623

PubMed Abstract | Crossref Full Text | Google Scholar

139. Lloyd-Davies C, Collins SL, and Burton GJ. Understanding the uterine artery Doppler waveform and its relationship to. spiral artery remodelling. Placenta. (2021) 105:78–84. doi: 10.1016/j.placenta.2021.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

140. Baltajian K, Hecht JL, Wenger JB, Salahuddin S, Verlohren S, Perschel FH, et al. Placental lesions of vascular insufficiency are associated with anti-angiogenic state in women with preeclampsia. Hypertens Pregnancy. (2014) 33:427–39. doi: 10.3109/10641955.2014.926914

PubMed Abstract | Crossref Full Text | Google Scholar

141. Makris A, Yeung KR, Lim SM, Sunderland N, Heffernan S, Thompson JF, et al. Placental growth factor reduces blood pressure in a uteroplacental ischemia model of preeclampsia in nonhuman primates. Hypertension. (2016) 67:1263–72. doi: 10.1161/HYPERTENSIONAHA.116.07286

PubMed Abstract | Crossref Full Text | Google Scholar

142. Parchem JG, Kanasaki K, Kanasaki M, Sugimoto H, Xie L, Hamano Y, et al. Loss of placental growth factor ameliorates maternal hypertension and preeclampsia in mice. J Clin Invest. (2018) 128:5008–17. doi: 10.1172/JCI99026

PubMed Abstract | Crossref Full Text | Google Scholar

143. Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. (2006) 12:642–9. doi: 10.1038/nm1429

PubMed Abstract | Crossref Full Text | Google Scholar

144. Walshe TE, Saint-Geniez M, Maharaj AS, Sekiyama E, Maldonado AE, and D’Amore PA. TGF-beta is required for vascular barrier function, endothelial survival and homeostasis of the adult microvasculature. PloS One. (2009) 4:e5149. doi: 10.1371/journal.pone.0005149

PubMed Abstract | Crossref Full Text | Google Scholar

145. Arroyo JA and Winn VD. Vasculogenesis and angiogenesis in the IUGR placenta. Semin Perinatol. (2008) 32:172–7. doi: 10.1053/j.semperi.2008.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

146. Pardi G, Marconi AM, and Cetin I. Pathophysiology of intrauterine growth retardation: role of the placenta. Acta Paediatr Suppl. (1997) 423:170–2. doi: 10.1111/j.1651-2227.1997.tb18405.x

PubMed Abstract | Crossref Full Text | Google Scholar

147. Srinivas SK, Edlow AG, Neff PM, Sammel MD, Andrela CM, and Elovitz MA. Rethinking IUGR in preeclampsia: dependent or independent of maternal hypertension? J Perinatol. (2009) 29:680–4. doi: 10.1038/jp.2009.83

PubMed Abstract | Crossref Full Text | Google Scholar

148. Chen CP, Bajoria R, and Aplin JD. Decreased vascularization and cell proliferation in placentas of intrauterine growth-restricted fetuses with abnormal umbilical artery flow velocity waveforms. Am J Obstet Gynecol. (2002) 187:764–9. doi: 10.1067/mob.2002.125243

PubMed Abstract | Crossref Full Text | Google Scholar

149. Krebs C, Macara LM, Leiser R, Bowman AW, Greer IA, and Kingdom JCP. Intrauterine growth restriction with absent end-diastolic flow velocity in the umbilical artery is associated with maldevelopment of the placental terminal villous tree. Am J Obstetrics Gynecology. (1996) 175:1534–42. doi: 10.1016/S0002-9378(96)70103-5

PubMed Abstract | Crossref Full Text | Google Scholar

150. Barut F, Barut A, Dogan Gun B, Onak Kandemir N, Ibrahim Harma M, Harma M, et al. Intrauterine growth restriction and placental angiogenesis. Diagn Pathol. (2010) 5:24. doi: 10.1186/1746-1596-5-24

PubMed Abstract | Crossref Full Text | Google Scholar

151. Lyall F, Young A, Boswell F, Kingdom JCP, and Greer IA. Placental expression of vascular endothelial growth factor in placentae from pregnancies complicated by pre-eclampsia and intrauterine growth restriction does not support placental hypoxia at delivery. Placenta. (1997) 18:269–76. doi: 10.1016/S0143-4004(97)80061-6

PubMed Abstract | Crossref Full Text | Google Scholar

152. Joó JG, Rigó J Jr, Börzsönyi B, Demendi C, and Kornya L. Placental gene expression of the placental growth factor (PlGF) in intrauterine growth restriction. J Maternal-Fetal Neonatal Med. (2017) 30:1471–5. doi: 10.1080/14767058.2016.1219993

PubMed Abstract | Crossref Full Text | Google Scholar

153. Regnault TR, Orbus RJ, de Vrijer B, Davidsen ML, Galan HL, Wilkening RB, et al. Placental expression of VEGF, PlGF and their receptors in a model of placental insufficiency-intrauterine growth restriction (PI-IUGR). Placenta. (2002) 23:132–44. doi: 10.1053/plac.2001.0757

PubMed Abstract | Crossref Full Text | Google Scholar

154. Diagnostic criteria and classification of hyperglycaemia first detected in pregnancy: a World Health Organization Guideline. Diabetes Res Clin Pract. (2014) 103:341–63. doi: 10.1016/j.diabres.2013.10.012

PubMed Abstract | Crossref Full Text | Google Scholar

155. Wang H, Li N, Chivesa T, Werfalli M, Sun H, Yuen L, et al. IDF diabetes atlas: estimation of global and regional gestational diabetes mellitus prevalence for 2021 by international association of diabetes in pregnancy study group’s criteria. Diabetes Res Clin Pract. (2022) 183:109050. doi: 10.1016/j.diabres.2021.109050

PubMed Abstract | Crossref Full Text | Google Scholar

156. Mistry SK, Das Gupta R, Alam S, Kaur K, Shamim AA, and Puthussery S. Gestational diabetes mellitus (GDM) and adverse pregnancy outcome in South Asia: A systematic review. Endocrinology Diabetes Metab. (2021) 4:e00285. doi: 10.1002/edm2.285

PubMed Abstract | Crossref Full Text | Google Scholar

157. Venkatesh KK, Lynch CD, Powe CE, Costantine MM, Thung SF, Gabbe SG, et al. Risk of adverse pregnancy outcomes among pregnant individuals with gestational diabetes by race and ethnicity in the United States, 2014-2020. Jama. (2022) 327:1356–67. doi: 10.1001/jama.2022.3189

PubMed Abstract | Crossref Full Text | Google Scholar

158. Carrasco-Wong I, Moller A, Giachini FR, Lima VV, Toledo F, Stojanova J, et al. Placental structure in gestational diabetes mellitus. Biochim Biophys Acta (BBA) - Mol Basis Dis. (2020) 1866:165535. doi: 10.1016/j.bbadis.2019.165535

PubMed Abstract | Crossref Full Text | Google Scholar

159. Higgins M, Felle P, Mooney EE, Bannigan J, and McAuliffe FM. Stereology of the placenta in type 1 and type 2 diabetes. Placenta. (2011) 32:564–9. doi: 10.1016/j.placenta.2011.04.015

PubMed Abstract | Crossref Full Text | Google Scholar

160. Nteeba J, Varberg KM, Scott RL, Simon ME, Iqbal K, and Soares MJ. Poorly controlled diabetes mellitus alters placental structure, efficiency, and plasticity. BMJ Open Diabetes Res Care. (2020) 8:e001243. doi: 10.1136/bmjdrc-2020-001243

PubMed Abstract | Crossref Full Text | Google Scholar

161. Dickinson JE, Meyer BA, Brath PC, Chmielowiec S, Walsh SW, Parisi VM, et al. Placental thromboxane and prostacyclin production in an ovine diabetic model. Am J Obstet Gynecol. (1990) 163:1831–5. doi: 10.1016/0002-9378(90)90759-Z

PubMed Abstract | Crossref Full Text | Google Scholar

162. White V, Jawerbaum A, Sinner D, Pustovrh C, Capobianco E, and González E. Oxidative stress and altered prostanoid production in the placenta of streptozotocin-induced diabetic rats. Reprod Fertil Dev. (2002) 14:117–23. doi: 10.1071/RD01032

PubMed Abstract | Crossref Full Text | Google Scholar

163. Salim MD, Al-Matubsi HY, El-Sharaky AS, Kamel MAN, Oriquat GA, Helmy MH, et al. The levels of vascular endothelial growth factor-A and placental growth factor-2 in embryopathy associated with experimental diabetic gestation. Growth Factors. (2009) 27:32–9. doi: 10.1080/08977190802587049

PubMed Abstract | Crossref Full Text | Google Scholar

164. Koh PO, Sung JH, Won CK, Cho JH, Moon JG, Park OS, et al. Streptozotocin-induced diabetes decreases placenta growth factor (PlGF) levels in rat placenta. J Vet Med Sci. (2007) 69:877–80. doi: 10.1292/jvms.69.877

PubMed Abstract | Crossref Full Text | Google Scholar

165. Jauniaux E, Hussein AM, Einerson BD, and Silver RM. Debunking 20th century myths and legends about the diagnosis of placenta accreta spectrum. Ultrasound Obstet Gynecol. (2022) 59:417–23. doi: 10.1002/uog.24890

PubMed Abstract | Crossref Full Text | Google Scholar

166. Jauniaux E, Grønbeck L, Bunce C, Langhoff-Roos J, and Collins SL. Epidemiology of placenta previa accreta: a systematic review and meta-analysis. BMJ. (2019) 9:. doi: 10.1136/bmjopen-2019-031193

Crossref Full Text | Google Scholar

167. Nisenblat V, Barak S, Griness OB, Degani S, Ohel G, and Gonen R. Maternal complications associated with multiple cesarean deliveries. Obstet Gynecol. (2006) 108:21–6. doi: 10.1097/01.AOG.0000222380.11069.11

PubMed Abstract | Crossref Full Text | Google Scholar

168. Einerson BD, Comstock J, Silver RM, Branch DW, Woodward PJ, and Kennedy A. Placenta accreta spectrum disorder: uterine dehiscence, not placental invasion. Obstet Gynecol. (2020) 135:1104–11. doi: 10.1097/AOG.0000000000003793

PubMed Abstract | Crossref Full Text | Google Scholar

169. Vissers J, Hehenkamp W, Lambalk CB, and Huirne JA. Post-Caesarean section niche-related impaired fertility: hypothetical mechanisms. Hum Reprod. (2020) 35:1484–94. doi: 10.1093/humrep/deaa094

PubMed Abstract | Crossref Full Text | Google Scholar

170. Afshar Y, Yin O, Jeong A, Martinez G, Kim J, Ma F, et al. Placenta accreta spectrum disorder at single-cell resolution: a loss of boundary limits in the decidua and endothelium. Am J Obstet Gynecol. (2024) 230:443.e1–443.e18. doi: 10.1016/j.ajog.2023.10.001

PubMed Abstract | Crossref Full Text | Google Scholar

171. Duzyj C, Buhimschi I, Hardy J, McCarthy M, Zhao G, Cross S, et al. 105: Decreased expression of endostatin (ES) and hypoxia-inducible factor 1&x3b1; (HIF-1&x3b1); is associated with excessive trophoblast invasion and aberrant angiogenesis in placenta accreta. Am J Obstetrics Gynecology. (2013) 208:S58–9. doi: 10.1016/j.ajog.2012.10.270

Crossref Full Text | Google Scholar

172. Duzyj CM, Buhimschi IA, Laky CA, Cozzini G, Zhao G, Wehrum M, et al. Extravillous trophoblast invasion in placenta accreta is associated with differential local expression of angiogenic and growth factors: a cross-sectional study. Bjog. (2018) 125:1441–8. doi: 10.1111/1471-0528.15176

PubMed Abstract | Crossref Full Text | Google Scholar

173. Jauniaux E, Jurkovic D, Hussein AM, and Burton GJ. New insights into the etiopathology of placenta accreta. spectrum. Am J Obstet Gynecol. (2022) 227:384–91. doi: 10.1016/j.ajog.2022.02.038

PubMed Abstract | Crossref Full Text | Google Scholar

174. Pollheimer J, Vondra S, Baltayeva J, Beristain AG, and Knöfler M. Regulation of placental extravillous trophoblasts by the maternal uterine environment. Front Immunol. (2018) 9:2597. doi: 10.3389/fimmu.2018.02597

PubMed Abstract | Crossref Full Text | Google Scholar

175. Jauniaux E, Zosmer N, De Braud LV, Ashoor G, Ross J, and Jurkovic D. Development of the utero-placental circulation in cesarean scar pregnancies: a case-control study. Am J Obstet Gynecol. (2022) 226:399.e1–399.e10. doi: 10.1016/j.ajog.2021.08.056

PubMed Abstract | Crossref Full Text | Google Scholar

176. Schwickert A, Henrich W, Vogel M, Melchior K, Ehrlich L, Ochs M, et al. Placenta percreta presents with neoangiogenesis of arteries with von willebrand factor-negative endothelium. Reprod Sci. (2022) 29:1136–44. doi: 10.1007/s43032-021-00763-4

PubMed Abstract | Crossref Full Text | Google Scholar

177. Tseng JJ and Chou MM. Differential expression of growth-, angiogenesis- and invasion-related factors in the development of placenta accreta. Taiwan J Obstet Gynecol. (2006) 45:100–6. doi: 10.1016/S1028-4559(09)60205-9

PubMed Abstract | Crossref Full Text | Google Scholar

178. Faraji A, Akbarzadeh-Jahromi M, Bahrami S, Gharamani S, Raeisi Shahraki H, Kasraeian M, et al. Predictive value of vascular endothelial growth factor and placenta growth factor for placenta accreta. spectrum. J Obstet Gynaecol. (2022) 42:900–5. doi: 10.1080/01443615.2021.1955337

PubMed Abstract | Crossref Full Text | Google Scholar

179. Goh W, Yamamoto SY, Thompson KS, and Bryant-Greenwood GD. Relaxin, its receptor (RXFP1), and insulin-like peptide 4 expression through gestation and in placenta accreta. Reprod Sci. (2013) 20:968–80. doi: 10.1177/1933719112472735

PubMed Abstract | Crossref Full Text | Google Scholar

180. Ma Y, Hu Y, He J, Wen X, Yang H, and Ma J. Abnormal placental development induced by repeated cesarean sections: Investigating an animal model of placenta accreta. Spectr Disord Placenta. (2024) 158:338–46. doi: 10.1016/j.placenta.2024.11.008

PubMed Abstract | Crossref Full Text | Google Scholar

181. Weiss G, Teichman S, Stewart D, Nader D, Wood S, and Unemori E. A randomized, double-blind, placebo-controlled trial of relaxin for cervical ripening in post-delivery date pregnancies. Ann N Y Acad Sci 2009. (1160) p:385–6. doi: 10.1111/j.1749-6632.2008.03794.x

PubMed Abstract | Crossref Full Text | Google Scholar

182. Teerlink JR, Cotter G, Davison BA, Felker GM, Filippatos G, Greenberg BH, et al. Serelaxin, recombinant human relaxin-2, for treatment of acute heart failure (RELAX-AHF): a randomised, placebo-controlled trial. Lancet. (2013) 381:29–39. doi: 10.1016/S0140-6736(12)61855-8

PubMed Abstract | Crossref Full Text | Google Scholar

183. Teerlink JR, Davison BA, Cotter G, Maggioni AP, Sato N, Chioncel O, et al. Effects of serelaxin in patients admitted for acute heart failure: a meta-analysis. Eur J Heart Fail. (2020) 22:315–29. doi: 10.1002/ejhf.1692

PubMed Abstract | Crossref Full Text | Google Scholar

184. Coomarasamy A, Williams H, Truchanowicz E, Seed PT, Small R, Quenby S, et al. A randomized trial of progesterone in women with recurrent miscarriages. N Engl J Med. (2015) 373:2141–8. doi: 10.1056/NEJMoa1504927

PubMed Abstract | Crossref Full Text | Google Scholar

185. Coomarasamy A, Devall AJ, Cheed V, Harb H, Middleton LJ, Gallos ID, et al. A randomized trial of progesterone in women with bleeding in early pregnancy. N Engl J Med. (2019) 380:1815–24. doi: 10.1056/NEJMoa1813730

PubMed Abstract | Crossref Full Text | Google Scholar

186. Melo P, Devall A, Shennan AH, Vatish M, Becker CM, Granne I, et al. Vaginal micronised progesterone for the prevention of hypertensive disorders of pregnancy: A systematic review and meta-analysis. BJOG: An International Journal of Obstetrics & Gynaecology. (2024) 131:727–39. doi: 10.1111/1471-0528.17705

PubMed Abstract | Crossref Full Text | Google Scholar

187. Quenby S and Farquharson RG. Human chorionic gonadotropin supplementation in recurring pregnancy loss: a controlled trial. Fertility Sterility. (1994) 62:708–10. doi: 10.1016/S0015-0282(16)56992-1

PubMed Abstract | Crossref Full Text | Google Scholar

188. Morley LC, Simpson N, and Tang T. Human chorionic gonadotrophin (hCG) for preventing miscarriage. Cochrane Database Syst Rev. (2013) 2013:Cd008611. doi: 10.1002/14651858.CD008611.pub2

PubMed Abstract | Crossref Full Text | Google Scholar

189. Poliwoda S, Noor N, Downs E, Schaaf A, Cantwell A, Ganti L, et al. Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice. Orthop Rev (Pavia). (2022) 14:37498. doi: 10.52965/001c.37498

PubMed Abstract | Crossref Full Text | Google Scholar

190. Du WJ, Chi Y, Yang ZX, Li ZJ, Cui JJ, Song BQ, et al. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther. (2016) 7:163. doi: 10.1186/s13287-016-0418-9

PubMed Abstract | Crossref Full Text | Google Scholar

191. Shi H, Yang Z, Cui J, Tao H, Ma R, and Zhao Y. Mesenchymal stem cell-derived exosomes: a promising alternative in the therapy of preeclampsia. Stem Cell Res Ther. (2024) 15:30. doi: 10.1186/s13287-024-03652-0

PubMed Abstract | Crossref Full Text | Google Scholar

192. Wu D, Liu Y, Liu XX, Liu WF, Shi HR, Zhang Y, et al. Heme oxygenase-1 gene modified human placental mesenchymal stem cells promote placental angiogenesis and spiral artery remodeling by improving the balance of angiogenic factors in vitro. Placenta. (2020) 99:70–7. doi: 10.1016/j.placenta.2020.07.007

PubMed Abstract | Crossref Full Text | Google Scholar

193. Liu Y, Shi HR, Wu D, Xu GX, Ma RL, Liu XX, et al. The protective benefit of heme oxygenase-1 gene-modified human placenta-derived mesenchymal stem cells in a N-nitro-L-arginine methyl ester-induced preeclampsia-like rat model: possible implications for placental angiogenesis. Stem Cells Dev. (2021) 30:991–1002. doi: 10.1089/scd.2021.0174

PubMed Abstract | Crossref Full Text | Google Scholar

194. Liu H, Wang F, Zhang Y, Xing Y, and Wang Q. Exosomal microRNA-139-5p from mesenchymal stem cells accelerates trophoblast cell invasion and migration by motivation of the ERK/MMP-2 pathway via downregulation of protein tyrosine phosphatase. J Obstet Gynaecol Res. (2020) 46:2561–72. doi: 10.1111/jog.14495

PubMed Abstract | Crossref Full Text | Google Scholar

195. Xiong ZH, Wei J, Lu MQ, Jin MY, and Geng HL. Protective effect of human umbilical cord mesenchymal stem cell exosomes on preserving the morphology and angiogenesis of placenta in rats with preeclampsia. BioMed Pharmacother. (2018) 105:1240–7. doi: 10.1016/j.biopha.2018.06.032

PubMed Abstract | Crossref Full Text | Google Scholar

196. Chang X, He Q, Wei M, Jia L, Wei Y, Bian Y, et al. Human umbilical cord mesenchymal stem cell derived exosomes (HUCMSC-exos) recovery soluble fms-like tyrosine kinase-1 (sFlt-1)-induced endothelial dysfunction in preeclampsia. Eur J Med Res. (2023) 28:277. doi: 10.1186/s40001-023-01182-8

PubMed Abstract | Crossref Full Text | Google Scholar

197. Xu L, Wang X, Liu Y, Yang G, Falconer RJ, and Zhao CX. Exposure to maternal depression alters the amygdala transcriptome and microglia morphology in the developing offspring. Lipid Nanoparticles Drug Delivery. (2022) 2:2100109. doi: 10.1002/anbr.202100109

Crossref Full Text | Google Scholar

198. Zhou Y, Ge Q, Wang X, Wang Y, Sun Q, Wang J, et al. Advances in lipid nanoparticle-based disease treatment. ChemMedChem. (2025) 20:. doi: 10.1002/cmdc.202400938

PubMed Abstract | Crossref Full Text | Google Scholar

199. Chaudhary N, Newby AN, Arral ML, Yerneni SS, LoPresti ST, Doerfler R, et al. Lipid nanoparticle structure and delivery route during pregnancy dictate mRNA potency, immunogenicity, and maternal and fetal outcomes. Proc. Natl. Acad. Sci. U.S.A.. (2024) 121:. doi: 10.1073/pnas.2307810121

PubMed Abstract | Crossref Full Text | Google Scholar

200. Swingle KL, Safford HC, Geisler HC, Hamilton AG, Thatte AS, Billingsley MM, et al. Ionizable Lipid Nanoparticles for In Vivo mRNA Delivery to the Placenta during Pregnancy. J Am Chem Soc. (2023) 145:4691–706. doi: 10.1021/jacs.2c12893

PubMed Abstract | Crossref Full Text | Google Scholar

201. Swingle KL, Hamilton AG, Safford HC, Geisler HC, Thatte AS, Palanki R, et al. Placenta-tropic VEGF mRNA lipid nanoparticles ameliorate murine pre-eclampsia. Nature. (2025) 637:412–21. doi: 10.1038/s41586-024-08291-2

PubMed Abstract | Crossref Full Text | Google Scholar

202. Young RE, Nelson KM, Hofbauer SI, Vijayakumar T, Alameh MG, Weissman D, et al. Systematic development of ionizable lipid nanoparticles for placental mRNA delivery using a design of experiments approach. Bioactive Materials. (2024) 34:125–37. doi: 10.1016/j.bioactmat.2023.11.014

PubMed Abstract | Crossref Full Text | Google Scholar

203. Katayama K, Furuki R, Yokoyama H, Kaneko M, Tachibana M, Yoshida I, et al. Enhanced in vivo gene transfer into the placenta using RGD fiber-mutant adenovirus vector. Biomaterials. (2011) 32:4185–93. doi: 10.1016/j.biomaterials.2011.02.038

PubMed Abstract | Crossref Full Text | Google Scholar

204. Carr DJ, Wallace JM, Aitken RP, Milne JS, Mehta V, Martin JF, et al. Uteroplacental adenovirus vascular endothelial growth factor gene therapy increases fetal growth velocity in growth-restricted sheep pregnancies. Hum Gene Ther. (2014) 25:375–84. doi: 10.1089/hum.2013.214

PubMed Abstract | Crossref Full Text | Google Scholar

205. Vaughan OR, Rossi CA, Ginsberg Y, White A, Hristova M, Sebire NJ, et al. Perinatal and long-term effects of maternal uterine artery adenoviral VEGF-A165 gene therapy in the growth-restricted Guinea pig fetus. Am J Physiol Regul Integr Comp Physiol. (2018) 315:R344–r353. doi: 10.1152/ajpregu.00210.2017

PubMed Abstract | Crossref Full Text | Google Scholar

206. Abd Ellah N, Taylor L, Troja W, Owens K, Ayres N, Pauletti G, et al. Development of non-viral, trophoblast-specific gene delivery for placental therapy. PloS One. (2015) 10:e0140879. doi: 10.1371/journal.pone.0140879

PubMed Abstract | Crossref Full Text | Google Scholar

207. Davenport BN, Jones HN, and Wilson RL. Placental treatment with insulin-like growth factor 1 via nanoparticle differentially impacts vascular remodeling factors in Guinea pig sub-placenta/decidua. Front Physiol. (2022) 13:1055234. doi: 10.3389/fphys.2022.1055234

PubMed Abstract | Crossref Full Text | Google Scholar

208. Wilson RL, Lampe K, Gupta MK, Duvall CL, and Jones HN. Nanoparticle-mediated transgene expression of insulin-like growth factor 1 in the growth restricted Guinea pig placenta increases placenta nutrient transporter expression and fetal glucose concentrations. Molecular Reproduction and Development. (2022) 89:540–53. doi: 10.1002/mrd.23644

PubMed Abstract | Crossref Full Text | Google Scholar

209. Wilson RL, Owens K, Sumser EK, Fry MV, Stephens KK, Chuecos M, et al. Nanoparticle mediated increased insulin-like growth factor 1 expression enhances human placenta syncytium function. Placenta. (2020) 93:1–7. doi: 10.1016/j.placenta.2020.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

210. Yu J, Jia J, Guo X, Chen R, and Feng L. Modulating circulating sFlt1 in an animal model of preeclampsia using PAMAM nanoparticles for siRNA delivery. Placenta. (2017) 58:1–8. doi: 10.1016/j.placenta.2017.07.360

PubMed Abstract | Crossref Full Text | Google Scholar

211. Turanov AA, Lo A, Hassler MR, Makris A, Ashar-Patel A, Alterman JF, et al. RNAi modulation of placental sFLT1 for the treatment of preeclampsia. Nat Biotechnol. (2018) 36:1164–73. doi: 10.1038/nbt.4297

PubMed Abstract | Crossref Full Text | Google Scholar

212. Luewan S, Teja-intr M, Sirichotiyakul S, and Tongsong T. Low maternal serum pregnancy-associated plasma protein-A as a risk factor of preeclampsia. Singapore Med J. (2018) 59:55–9. doi: 10.11622/smedj.2017034

PubMed Abstract | Crossref Full Text | Google Scholar

213. Ong CY, Liao AW, Spencer K, Munim S, and Nicolaides KH. First trimester maternal serum free beta human chorionic gonadotrophin and pregnancy associated plasma protein A as predictors of pregnancy complications. Bjog. (2000) 107:1265–70. doi: 10.1111/j.1471-0528.2000.tb11618.x

PubMed Abstract | Crossref Full Text | Google Scholar

214. Keikkala E, Vuorela P, Laivuori H, Romppanen J, Heinonen S, et al. First trimester hyperglycosylated human chorionic gonadotrophin in serum - a marker of early-onset preeclampsia. Placenta. (2013) 34:1059–65. doi: 10.1016/j.placenta.2013.08.006

PubMed Abstract | Crossref Full Text | Google Scholar

215. Leung TY, Sahota DS, Chan LW, Law LW, Fung TY, Leung TN, et al. Prediction of birth weight by fetal crown-rump length and maternal serum levels of pregnancy-associated plasma protein-A in the first trimester. Ultrasound Obstet Gynecol. (2008) 31:10–4. doi: 10.1002/uog.5206

PubMed Abstract | Crossref Full Text | Google Scholar

216. Spencer K, Yu CKH, Cowans NJ, Otigbah C, and Nicolaides KH. Prediction of pregnancy complications by first-trimester maternal serum PAPP-A and free beta-hCG and with second-trimester uterine artery Doppler. Prenat Diagn. (2005) 25:949–53. doi: 10.1002/pd.1251

PubMed Abstract | Crossref Full Text | Google Scholar

217. Panwar M, Kumari A, Anand HP, Arora R, Singh V, and Bansiwal R. Raised neutrophil lymphocyte ratio and serum beta hCG level in early second trimester of pregnancy as predictors for development and severity of preeclampsia. Drug Discov Ther. (2019) 13:34–7. doi: 10.5582/ddt.2019.01006

PubMed Abstract | Crossref Full Text | Google Scholar

218. Dugoff L, Hobbins JC, Malone FD, Porter TF, Luthy D, Comstock CH, et al. First-trimester maternal serum PAPP-A and free-beta subunit human chorionic gonadotropin concentrations and nuchal translucency are associated with obstetric complications: a population-based screening study (the FASTER Trial). Am J Obstet Gynecol. (2004) 191:1446–51. doi: 10.1016/j.ajog.2004.06.052

PubMed Abstract | Crossref Full Text | Google Scholar

219. Zhang X, Huangfu Z, Shi F, and Xiao Z. Predictive performance of serum β-hCG moM levels for preeclampsia screening: A meta-analysis. Front Endocrinol (Lausanne). (2021) 12:619530. doi: 10.3389/fendo.2021.619530

PubMed Abstract | Crossref Full Text | Google Scholar

220. Cowans NJ and Spencer K. First-trimester ADAM12 and PAPP-A as markers for intrauterine fetal growth restriction through their roles in the insulin-like growth factor system. Prenat Diagn. (2007) 27:264–71. doi: 10.1002/pd.1665

PubMed Abstract | Crossref Full Text | Google Scholar

221. Poon LC, Chelemen T, Granvillano O, Pandeva I, and Nicolaides KH. First-trimester maternal serum a disintegrin and metalloprotease 12 (ADAM12) and adverse pregnancy outcome. Obstet Gynecol. (2008) 112:1082–90. doi: 10.1097/AOG.0b013e318188d6f9

PubMed Abstract | Crossref Full Text | Google Scholar

222. Matwejew E, Cowans NJ, Stamatopoulou A, Spencer K, and von Kaisenberg CS. Maternal serum ADAM-12 as a potential marker for different adverse pregnancy outcomes. Fetal Diagn Ther. (2010) 27:32–9. doi: 10.1159/000275669

PubMed Abstract | Crossref Full Text | Google Scholar

223. Romero R, Kusanovic JP, Than NG, Erez O, Gotsch F, Espinoza J, et al. First-trimester maternal serum PP13 in the risk assessment for preeclampsia. Am J Obstet Gynecol. (2008) 199:122.e1–122.e11. doi: 10.1016/j.ajog.2008.01.013

PubMed Abstract | Crossref Full Text | Google Scholar

224. El Sherbiny WS, Soliman A, and Nasr AS. Placental protein 13 as an early predictor in Egyptian patients with preeclampsia, correlation to risk, and association with outcome. J Investig Med. (2012) 60:818–22. doi: 10.2310/JIM.0b013e31824e9a68

PubMed Abstract | Crossref Full Text | Google Scholar

225. Wu Y, Liu Y, and Ding Y. Predictive performance of placental protein 13 for screening preeclampsia in the first trimester: A systematic review and meta-analysis. Front Med (Lausanne). (2021) 8:756383. doi: 10.3389/fmed.2021.756383

PubMed Abstract | Crossref Full Text | Google Scholar

226. Kazatsker MM, Sharabi-Nov A, Meiri H, Sammour R, and Sammar M. Augmented placental protein 13 in placental-associated extracellular vesicles in term and preterm preeclampsia is further elevated by corticosteroids. Int J Mol Sci. (2023) 24:12051. doi: 10.3390/ijms241512051

PubMed Abstract | Crossref Full Text | Google Scholar

227. De Villiers CP, Hedley PL, Placing S, Wøjdemann KR, Shalmi AC, Carlsen AL, et al. Placental protein-13 (PP13) in combination with PAPP-A and free leptin index (fLI) in first trimester maternal serum screening for severe and early preeclampsia. Clin Chem Lab Med. (2017) 56:65–74. doi: 10.1515/cclm-2017-0356

PubMed Abstract | Crossref Full Text | Google Scholar

228. Keikkala E, Forstén J, Ritvos O, Stenman U-H, Kajantie E, Hämäläinen E, et al. Serum Inhibin-A and PAPP-A2 in the prediction of pre-eclampsia during the first and second trimesters in high-risk women. Pregnancy Hypertension. (2021) 25:116–22. doi: 10.1016/j.preghy.2021.05.024

PubMed Abstract | Crossref Full Text | Google Scholar

229. Broumand F, Salari Lak S, Nemati F, and Mazidi A. A study of the diagnostic value of Inhibin A Tests for occurrence of preeclampsia in pregnant women. Electron Physician. (2018) 10:6186–92. doi: 10.19082/6186

PubMed Abstract | Crossref Full Text | Google Scholar

230. Rehfeldt M, Eklund E, Struck J, Sparwasser A, O'Brien B, Palomaki GE, et al. Relaxin-2 connecting peptide (pro-RLX2) levels in second trimester serum samples to predict preeclampsia. Pregnancy Hypertens. (2018) 11:124–8. doi: 10.1016/j.preghy.2017.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

231. Post Uiterweer ED, Koster MPH, Jeyabalan A, Kuc S, Siljee JE, Stewart DR, et al. Circulating pregnancy hormone relaxin as a first trimester biomarker for preeclampsia. Pregnancy Hypertension. (2020) 22:47–53. doi: 10.1016/j.preghy.2020.07.008

PubMed Abstract | Crossref Full Text | Google Scholar

232. Ugwu A, Okunade KS, Oluwole AA, Olabode SAJ, Ani-Ugwu NK, Makwe CC, et al. Serum relaxin in preeclamptic and normotensive pregnant women at the Lagos University Teaching Hospital. Hellenic J Obstetrics Gynecology. (2022) 21:93–101. doi: 10.33574/hjog.0504

Crossref Full Text | Google Scholar

233. Yadav S, Yadav YS, Goel MM, Singh U, Natu SM, and Negi MPS. Calcitonin gene- and parathyroid hormone-related peptides in normotensive and preeclamptic pregnancies: a nested case–control study. Arch Gynecology Obstetrics. (2014) 290:897–903. doi: 10.1007/s00404-014-3303-8

PubMed Abstract | Crossref Full Text | Google Scholar

234. Wan J, Hu Z, Zeng K, Yin Y, Zhao M, Chen M, et al. The reduction in circulating levels of estrogen and progesterone in women with preeclampsia. Pregnancy Hypertens. (2018) 11:18–25. doi: 10.1016/j.preghy.2017.12.003

PubMed Abstract | Crossref Full Text | Google Scholar

235. Efanga S, Akintomide OA, Udofia AT, Okon OA, and Okpara HC. Serum human placental lactogen assays in ultrasound evaluated pregnancy-induced hypertension: A marker of placental function in pregnancy. Niger J Physiol Sci. (2023) 38:7–12. doi: 10.54548/njps.v38i1.2

PubMed Abstract | Crossref Full Text | Google Scholar

236. Taylor BD, Ness RB, Olsen J, Hougaard DM, Skogstrand K, Roberts JM, et al. Serum leptin measured in early pregnancy is higher in women with preeclampsia compared with normotensive pregnant women. Hypertension. (2015) 65:594–9. doi: 10.1161/HYPERTENSIONAHA.114.03979

PubMed Abstract | Crossref Full Text | Google Scholar

237. Toplu M, Aktürk E, Cıngıllıoğlu B, Uzun HC, Kaya YDY, and Genç S. Serum leptin as a diagnostic biomarker in preeclampsia: A prospective controlled study. Bagcilar Med Bull. (2025) 12:1–19. doi: 10.4274/BMB.galenos.2025.03521

Crossref Full Text | Google Scholar

238. Florio P, Imperatore A, Sanseverino F, Torricelli M, Reis FM, Lowry PJ, et al. The measurement of maternal plasma corticotropin-releasing factor (CRF) and CRF-binding protein improves the early prediction of preeclampsia. J Clin Endocrinol Metab. (2004) 89:4673–7. doi: 10.1210/jc.2004-0186

PubMed Abstract | Crossref Full Text | Google Scholar

239. Laatikainen T, Virtanen T, Kaaja R, and Salminen-Lappalainen K. Corticotropin-releasing hormone in maternal and cord plasma in pre-eclampsia. Eur J Obstet Gynecol Reprod Biol. (1991) 39:19–24. doi: 10.1016/0028-2243(91)90136-9

PubMed Abstract | Crossref Full Text | Google Scholar

240. Paiva SPC, Veloso CA, Campos FFC, Carneiro MM, Tilan JU, Wang H, et al. Elevated levels of neuropeptide Y in preeclampsia: A pilot study implicating a role for stress in pathogenesis of the disease. Neuropeptides. (2016) 55:127–35. doi: 10.1016/j.npep.2015.09.006

PubMed Abstract | Crossref Full Text | Google Scholar

241. Blackburn CA, Keelan JA, Taylor RS, and North RA. Maternal serum activin A is not elevated before preeclampsia in women who are at high risk. Am J Obstet Gynecol. (2003) 188:807–11. doi: 10.1067/mob.2003.173

PubMed Abstract | Crossref Full Text | Google Scholar

242. Spencer K, Cowans NJ, and Nicolaides KH. Maternal serum inhibin-A and activin-A levels in the first trimester of pregnancies developing pre-eclampsia. Ultrasound Obstet Gynecol. (2008) 32:622–6. doi: 10.1002/uog.6212

PubMed Abstract | Crossref Full Text | Google Scholar

243. Su J, Huang X, Meng S, and Wang S. Investigating serum and placental levels of IGF-1 and IGF-1R in preeclampsia patients and their clinical implications. Int J Womens Health. (2025) 17:729–38. doi: 10.2147/IJWH.S512910

PubMed Abstract | Crossref Full Text | Google Scholar

244. Sibiude J, Guibourdenche J, Dionne M-D, Le Ray C, Anselem O, Serreau R, et al. Placental growth factor for the prediction of adverse outcomes in patients with suspected preeclampsia or intrauterine growth restriction. PloS One. (2012) 7:e50208. doi: 10.1371/journal.pone.0050208

PubMed Abstract | Crossref Full Text | Google Scholar

245. Polliotti BM, Fry AG, Saller DN, Mooney RA, Cox C, and Miller RK. Second-trimester maternal serum placental growth factor and vascular endothelial growth factor for predicting severe, early-onset preeclampsia. Obstet Gynecol. (2003) 101:1266–74. doi: 10.1016/s0029-7844(03)00338-7

PubMed Abstract | Crossref Full Text | Google Scholar

246. Kim S-Y, Ryu H-M, Yang J-H, Kim M-Y, Han J-Y, and Kim J-O. Increased sFlt-1 to PlGF ratio in women who subsequently develop preeclampsia. J Korean Med Sci. (2007) 22:873–7. doi: 10.3346/jkms.2007.22.5.873

PubMed Abstract | Crossref Full Text | Google Scholar

247. Tarasevičienė V, Grybauskienė R, and Mačiulevičienė R. sFlt-1, PlGF, sFlt-1/PlGF ratio and uterine artery Doppler for preeclampsia diagnostics. Medicina (Kaunas). (2016) 52:349–53. doi: 10.1016/j.medici.2016.11.008

PubMed Abstract | Crossref Full Text | Google Scholar

248. Papamichail M, Fasoulakis Z, Daskalakis G, Theodora M, Rodolakis A, and Antsaklis P. Importance of low pregnancy associated plasma protein-A (PAPP-A) levels during the first trimester as a predicting factor for adverse pregnancy outcomes: A prospective cohort study of 2636 pregnant women. Cureus. (2022) 14:e31256. doi: 10.7759/cureus.31256

PubMed Abstract | Crossref Full Text | Google Scholar

249. Springer S, Worda K, Franz M, Karner E, Krampl-Bettelheim E, and Worda C. Fetal growth restriction is associated with pregnancy associated plasma protein A and uterine artery doppler in first trimester. J Clin Med. (2023) 12:2502. doi: 10.3390/jcm12072502

PubMed Abstract | Crossref Full Text | Google Scholar

250. Huang J, Liu Y, Yang H, Xu Y, and Lv W. The effect of serum β-human chorionic gonadotropin on pregnancy complications and adverse pregnancy outcomes: A systematic review and meta-analysis. Comput Math Methods Med 2022. (2022) p:8315519. doi: 10.1155/2022/8315519

PubMed Abstract | Crossref Full Text | Google Scholar

251. Chafetz I, Kuhnreich I, Sammar M, Tal Y, Gibor Y, Meiri H, et al. First-trimester placental protein 13 screening for preeclampsia and intrauterine growth restriction. Am J Obstet Gynecol. (2007) 197:35.e1–7. doi: 10.1016/j.ajog.2007.02.025

PubMed Abstract | Crossref Full Text | Google Scholar

252. Ron G, Chefetz I, Grimpel YI, Sammar M, and Meiri H. Assessment of first trimester maternal serum placental protein 13 for the prediction of IUGR with and without preeclampsia. Am J Obstetrics Gynecology - AMER J OBSTET GYNECOL. (2006) 195:41–9. doi: 10.1016/j.ajog.2006.10.533

Crossref Full Text | Google Scholar

253. Cowans NJ, Spencer K, and Meiri H. First-trimester maternal placental protein 13 levels in pregnancies resulting in adverse outcomes. Prenatal Diagnosis. (2008) 28:121–5. doi: 10.1002/pd.1921

PubMed Abstract | Crossref Full Text | Google Scholar

254. Efanga S, Akintomide AO, Obiora CI, Okpara HC, and Efanga I. Correlation of sonographic fetal growth evaluation with maternal serum Human Placental Lactogen levels. IOSR J Dental Med Sci. (2021) 20:43–8. doi: 10.9790/0853-2006124348

Crossref Full Text | Google Scholar

255. Stefaniak M and Dmoch-Gajzlerska E. Maternal serum and cord blood leptin concentrations at delivery in normal pregnancies and in pregnancies complicated by intrauterine growth restriction. Obes Facts. (2022) 15:62–9. doi: 10.1159/000519609

PubMed Abstract | Crossref Full Text | Google Scholar

256. Wadhwa PD, Garite TJ, Porto M, Glynn L, Chicz-DeMet A, Dunkel-Schetter C, et al. Placental corticotropin-releasing hormone (CRH), spontaneous preterm birth, and fetal growth restriction: a prospective investigation. Am J Obstet Gynecol. (2004) 191:1063–9. doi: 10.1016/j.ajog.2004.06.070

PubMed Abstract | Crossref Full Text | Google Scholar

257. Whitehead CL, Walker SP, Ye L, Mendis S, Kaitu'u-Lino TJ, Lappas M, et al. Placental specific mRNA in the maternal circulation are globally dysregulated in pregnancies complicated by fetal growth restriction. J Clin Endocrinol Metab. (2013) 98:E429–36. doi: 10.1210/jc.2012-2468

PubMed Abstract | Crossref Full Text | Google Scholar

258. Morpurgo PS, Cetin I, Borgato S, Cortelazzi D, Nobile-DeSantis MS, Vaghi I, et al. Circulating levels of inhibin A, inhibin B and activin A in normal and intrauterine growth restricted (IUGR) fetuses. Eur J Obstet Gynecol Reprod Biol. (2004) 117:38–44. doi: 10.1016/j.ejogrb.2004.02.030

PubMed Abstract | Crossref Full Text | Google Scholar

259. Tuzluoğlu S, Üstünyurt E, Karaşin SS, and Karaşin ZT. Investigation of serum pregnancy-specific beta-1-glycoprotein and relationship with fetal growth restriction. JBRA Assist Reprod. (2022) 26:267–73. doi: 10.5935/1518-0557.20210068

PubMed Abstract | Crossref Full Text | Google Scholar

260. Giorgione V, Ramnarine S, Malik A, and Bhide A. The value of angiogenetic biomarkers in the detection of early onset fetal growth restriction. Eur J Obstetrics Gynecology Reprod Biol. (2024) 299:91–5. doi: 10.1016/j.ejogrb.2024.05.036

PubMed Abstract | Crossref Full Text | Google Scholar

261. Chen W, Wei Q, Liang Q, Song S, and Li J. Diagnostic capacity of sFlt-1/PlGF ratio in fetal growth restriction: A systematic review and meta-analysis. Placenta. (2022) 127:37–42. doi: 10.1016/j.placenta.2022.07.020

PubMed Abstract | Crossref Full Text | Google Scholar

262. Iannaccone A, Reisch B, Mavarani L, Darkwah Oppong M, Kimmig R, Mach P, et al. OP02.07: sEng versus sFlt-1/PlGF ratio: prediction of PE, HELLP syndrome and IUGR. Ultrasound Obstetrics Gynecology. (2021) 58:63–3. doi: 10.1002/uog.23932

Crossref Full Text | Google Scholar

263. Stepan H, Krämer T, and Faber R. Maternal plasma concentrations of soluble endoglin in pregnancies with intrauterine growth restriction. J Clin Endocrinol Metab. (2007) 92:2831–4. doi: 10.1210/jc.2006-2774

PubMed Abstract | Crossref Full Text | Google Scholar

264. Ravikumar G, Mukhopadhyay A, Mani C, Kocchar P, Crasta J, Thomas T, et al. Placental expression of angiogenesis-related genes and their receptors in IUGR pregnancies: correlation with fetoplacental and maternal parameters. J Maternal-Fetal Neonatal Med. (2020) 33:3954–61. doi: 10.1080/14767058.2019.1593362

PubMed Abstract | Crossref Full Text | Google Scholar

265. Al-Ofi E, Alrafiah A, Maidi S, Almaghrabi S, and Hakami N. Altered expression of angiogenic biomarkers in pregnancy associated with gestational diabetes. Int J Gen Med. (2021) 14:3367–75. doi: 10.2147/IJGM.S316670

PubMed Abstract | Crossref Full Text | Google Scholar

266. Laskowska M, Laskowska K, and Oleszczuk J. Endoglin in pregnancy complicated by fetal intrauterine growth restriction in normotensive and preeclamptic pregnant women: a comparison between preeclamptic patients with appropriate-for-gestational-age weight infants and healthy pregnant women. J Matern Fetal Neonatal Med. (2012) 25:806–11. doi: 10.3109/14767058.2011.595852

PubMed Abstract | Crossref Full Text | Google Scholar

267. Conde-Agudelo A, Papageorghiou AT, Kennedy SH, and Villar J. Novel biomarkers for predicting intrauterine growth restriction: a systematic review and meta-analysis. Bjog. (2013) 120:681–94. doi: 10.1111/1471-0528.12172

PubMed Abstract | Crossref Full Text | Google Scholar

268. Alonso Lopez Y, Dereke J, Landin-Olsson M, Strevens H, Nilsson C, and Hillman M. Plasma levels of relaxin-2 are higher and correlated to C-peptide levels in early gestational diabetes mellitus. Endocrine. (2017) 57:545–7. doi: 10.1007/s12020-017-1354-x

PubMed Abstract | Crossref Full Text | Google Scholar

269. Zaman I, Swaminathan R, Brackenridge A, Sankaralingam A, and McGowan B. Assessment of relaxin levels in pregnant women with gestational diabetes mellitus. Endocrine Abstracts. (2014) 17:652–60. doi: 10.1530/endoabs.35.P377

Crossref Full Text | Google Scholar

270. Merviel P, Müller F, Guibourdenche J, Berkane N, Gaudet R, Bréart G, et al. Correlations between serum assays of human chorionic gonadotrophin (hCG) and human placental lactogen (hPL) and pre-eclampsia or intrauterine growth restriction (IUGR) among nulliparas younger than 38 years. Eur J Obstet Gynecol Reprod Biol. (2001) 95:59–67. doi: 10.1016/S0301-2115(00)00370-5

PubMed Abstract | Crossref Full Text | Google Scholar

271. Liu Y, Guo F, Maraka S, Zhang Y, Zhang C, Tim IM, et al. Associations between human chorionic gonadotropin, maternal free thyroxine, and gestational diabetes mellitus. Thyroid. (2021) 31:1282–8. doi: 10.1089/thy.2020.0920

PubMed Abstract | Crossref Full Text | Google Scholar

272. Zorlu U, Elmas B, Ergün GT, Sucu ST, Ozan E, Aydoǧdu E, et al. Determining the risk of gestational diabetes using machine learning: A study on first-trimester PAPP-A and β-hCG data. J Gynaecol Obstet. (2025) 113:11296–301. doi: 10.1002/ijgo.70264

PubMed Abstract | Crossref Full Text | Google Scholar

273. Cheuk QK, Lo TK, Wong SF, and Lee CP. Association between pregnancy-associated plasma protein-A levels in the first trimester and gestational diabetes mellitus in Chinese women. Hong Kong Med J. (2016) 22:30–8. doi: 10.12809/hkmj144470

PubMed Abstract | Crossref Full Text | Google Scholar

274. Balkaş G and Çelen Ş. Early prediction of gestational diabetes mellitus and insulin therapy requirement using first-trimester PAPP-A and free β-hCG moMs levels: A retrospective case–control study. J Clin Med. (2024) 13:7725. doi: 10.3390/jcm13247725

PubMed Abstract | Crossref Full Text | Google Scholar

275. Zhao B, et al. Early second trimester maternal serum markers in the prediction of gestational diabetes mellitus. J Diabetes Investig. (2018) 9:967–74. doi: 10.1111/jdi.12798

PubMed Abstract | Crossref Full Text | Google Scholar

276. Rassie K, Giri R, Joham AE, Teede H, and Mousa A. Human placental lactogen in relation to maternal metabolic health and fetal outcomes: A systematic review and meta-analysis. Int J Mol Sci. (2022) 23:15621. doi: 10.3390/ijms232415621

PubMed Abstract | Crossref Full Text | Google Scholar

277. Kautzky-Willer A, Pacini G, Tura A, Bieglmayer C, Schneider B, Ludvik B, et al. Increased plasma leptin in gestational diabetes. Diabetologia. (2001) 44:164–72. doi: 10.1007/s001250051595

PubMed Abstract | Crossref Full Text | Google Scholar

278. Soheilykhah S, Mojibian M, Rahimi-Saghand S, Rashidi M, and Hadinedoushan H. Maternal serum leptin concentration in gestational diabetes. Taiwanese J Obstetrics Gynecology. (2011) 50:149–53. doi: 10.1016/j.tjog.2011.01.034

PubMed Abstract | Crossref Full Text | Google Scholar

279. Roca-Rodríguez MDM, Ramos-García P, López-Tinoco C, and Aguilar-Diosdado M. Significance of serum-plasma leptin profile during pregnancy in gestational diabetes mellitus: A systematic review and meta-analysis. J Clin Med. (2022) 11:2433. doi: 10.3390/jcm11092433

PubMed Abstract | Crossref Full Text | Google Scholar

280. Eleftheriades M, Papastefanoud I, Lambrinoudaki I, Kappou D, Lavranos D, Akalestos A, et al. Elevated placental growth factor concentrations at 11–14 weeks of gestation to predict gestational diabetes mellitus. Metabolism. (2014) 63:1419–25. doi: 10.1016/j.metabol.2014.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

281. Mosimann B, Amylidi S, Risch L, Wiedemann U, Surbek D, Baumann M, et al. First-trimester placental growth factor in screening for gestational diabetes. Fetal Diagnosis Ther. (2015) 39:287–91. doi: 10.1159/000441027

PubMed Abstract | Crossref Full Text | Google Scholar

282. Madiwale S, Kasture V, Sundrani D, Krishnaveni GV, Gupte S, and Joshi S. Angiogenic markers in gestational diabetes and their association with placental dimensions. Mol Cell Biochem. (2025) 480:3637–46. doi: 10.1007/s11010-024-05189-5

PubMed Abstract | Crossref Full Text | Google Scholar

283. Kapustin RV, Kopteeva EV, Alekseenkova EN, Korenevsky AV, Smirnov IV, and Arzhanova ON. Prediction of preeclampsia based on maternal serum endoglin level in women with pregestational diabetes mellitus. Hypertens Pregnancy. (2022) 41:173–80. doi: 10.1080/10641955.2022.2068574

PubMed Abstract | Crossref Full Text | Google Scholar

284. Balkaş G and Çaglar T. Elevated first-trimester PAPP-A is a marker in high-risk pregnancies with an increased risk of placenta accreta in predicting adverse outcomes. Eur Rev Med Pharmacol Sci. (2023) 27:9955–61. doi: 10.26355/eurrev_202310_34174

PubMed Abstract | Crossref Full Text | Google Scholar

285. Li Y, Meng Y, Chi Y, Li P, and He J. Meta-analysis for the relationship between circulating pregnancy-associated plasma protein A and placenta accreta. spectrum. Med (Baltimore). (2023) 102:e34473. doi: 10.1097/MD.0000000000034473

PubMed Abstract | Crossref Full Text | Google Scholar

286. Belousova V, Ignatko I, Bogomazova I, Zarova E, Pesegova S, Samusevich A, et al. Combined first-trimester PAPP-A and free β-hCG levels for the early diagnosis of placenta accreta spectrum and placenta previa: A case-control study. Int J Mol Sci. (2025) 26:6187. doi: 10.3390/ijms26136187

PubMed Abstract | Crossref Full Text | Google Scholar

287. Einerson BD, Straubhar A, Soisson S, Szczotka K, Dodson MK, Silver RM, et al. Hyperglycosylated hCG and placenta accreta spectrum. Am J Perinatol. (2019) 36:22–6. doi: 10.1055/s-0038-1636501

PubMed Abstract | Crossref Full Text | Google Scholar

288. Kawashima A, Sekizawa A, Ventura W, Koide K, Hori KY, Okai T, et al. Increased levels of cell-free human placental lactogen mRNA at 28–32 gestational weeks in plasma of pregnant women with placenta previa and invasive placenta. Reprod Sci. (2014) 21:215–20. doi: 10.1177/1933719113492209

PubMed Abstract | Crossref Full Text | Google Scholar

289. Li J, Zhang N, Zhang Y, Hu X, Gao G, Ye Y, et al. Human placental lactogen mRNA in maternal plasma play a role in prenatal diagnosis of abnormally invasive placenta: yes or no? Gynecol Endocrinol. (2019) 35:631–4. doi: 10.1080/09513590.2019.1576607

PubMed Abstract | Crossref Full Text | Google Scholar

290. Berezowsky A, Pardo J, Ben-Zion M, Wiznitzer A, and Aviram A. Second trimester biochemical markers as possible predictors of pathological placentation: A retrospective case-control study. Fetal Diagn Ther. (2019) 46:187–92. doi: 10.1159/000492829

PubMed Abstract | Crossref Full Text | Google Scholar

291. Wang F, Man D, and Liu S. Elevated second trimester alpha-fetoprotein increases the risk of placenta accreta. Clin. Exp. Obstet. Gynecol. (2023) 50:232. doi: 10.31083/j.ceog5011232

Crossref Full Text | Google Scholar

292. Zhang F, Xia L, Zeng L, You H, Liu Q, and Wang Y. Relationship between maternal serum sFlt-1 level and placenta accreta spectrum disorders in the third trimester. Arch Gynecology Obstetrics. (2024) 310:2453–9. doi: 10.1007/s00404-024-07734-5

PubMed Abstract | Crossref Full Text | Google Scholar

293. Alessandrini L, Aryananda R, Ariani G, Agustina B, Aldika Akbar MI, Dachlan EG, et al. The correlation between serum levels and placental tissue expression of PLGF and sFLT-1 and the FIGO grading of the placenta accreta. Spectr Disord J Matern Fetal Neonatal Med. (2023) 36:2183744. doi: 10.1080/14767058.2023.2183744

PubMed Abstract | Crossref Full Text | Google Scholar

294. Chen DB, Bird IM, Zheng J, and Magness RR. Membrane estrogen receptor-dependent extracellular signal-regulated kinase pathway mediates acute activation of endothelial nitric oxide synthase by estrogen in uterine artery endothelial cells. Endocrinology. (2004) 145:113–25. doi: 10.1210/en.2003-0547

PubMed Abstract | Crossref Full Text | Google Scholar

295. Kazemi Aski S, Sharami SH, Tavangar A, Kazemnezhad E, Dalil Heirati SF, and Etezadi A. Comparison of pregnancy-associated plasma protein-A levels in women with and without intrauterine growth restriction. J Obstetrics Gynecology Cancer Res. (2024) 9:14–21. doi: 10.30699/jogcr.9.1.14

Crossref Full Text | Google Scholar

Keywords: uterine vasculature, feto-maternal interface, placental hormones, pregnancy complications, angiogenic factors

Citation: Esposito M, Paulesu L and Mandalà M (2025) The role of placental hormones and metabolites in modulating uterine circulation in physiological and pathological pregnancies. Front. Endocrinol. 16:1637570. doi: 10.3389/fendo.2025.1637570

Received: 29 May 2025; Accepted: 28 July 2025;
Published: 19 August 2025.

Edited by:

Fred Sinowatz, Ludwig Maximilian University of Munich, Germany

Reviewed by:

Arpita Vyas, Washington University in St. Louis, United States
Yongdan Ma, Peking University, China

Copyright © 2025 Esposito, Paulesu and Mandalà. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Maurizio Mandalà, bS5tYW5kYWxhQHVuaWNhbC5pdA==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.