Next Article in Journal
Lavandula austroapennina (Lamiaceae): Getting Insights into Bioactive Polyphenols of a Rare Italian Endemic Vascular Plant
Next Article in Special Issue
MIF Increases sFLT1 Expression in Early Uncomplicated Pregnancy and Preeclampsia
Previous Article in Journal
Vitamin D Modulates the Response of Patient-Derived Metastatic Melanoma Cells to Anticancer Drugs
Previous Article in Special Issue
Biomarkers for Pregnancy Latency Prediction after Preterm Premature Rupture of Membranes–A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Apelinergic System in Pregnancy

by
Océane Pécheux
1,*,
Ana Correia-Branco
2,
Marie Cohen
2 and
Begoῆa Martinez de Tejada
1,2,*
1
Obstetrics Division, Department of Woman, Child and Adolescent, Geneva University Hospitals, 1205 Geneva, Switzerland
2
Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8014; https://doi.org/10.3390/ijms24098014
Submission received: 11 March 2023 / Revised: 24 April 2023 / Accepted: 25 April 2023 / Published: 28 April 2023
(This article belongs to the Special Issue Placental Related Disorders of Pregnancy 2.0.)

Abstract

:
The apelinergic system is a highly conserved pleiotropic system. It comprises the apelin receptor apelin peptide jejunum (APJ) and its two peptide ligands, Elabela/Toddler (ELA) and apelin, which have different spatiotemporal localizations. This system has been implicated in the regulation of the adipoinsular axis, in cardiovascular and central nervous systems, in carcinogenesis, and in pregnancy in humans. During pregnancy, the apelinergic system is essential for embryo cardiogenesis and vasculogenesis and for placental development and function. It may also play a role in the initiation of labor. The apelinergic system seems to be involved in the development of placenta-related pregnancy complications, such as preeclampsia (PE) and intrauterine growth restriction, but an improvement in PE-like symptoms and birth weight has been described in murine models after the exogenous administration of apelin or ELA. Although the expression of ELA, apelin, and APJ is altered in human PE placenta, data related to their circulating levels are inconsistent. This article reviews current knowledge about the roles of the apelinergic system in pregnancy and its pathophysiological roles in placenta-related complications in pregnancy. We also discuss the challenges in translating the actors of the apelinergic system into a marker or target for therapeutic interventions in obstetrics.

1. Overview of the Apelinergic System

The apelinergic system is composed of a group of three actors, namely, a receptor named apelin peptide jejunum (APJ) and its two peptide ligands, Elabela/Toddler (ELA) and apelin [1]. The APJ gene, APLNR, was discovered in 1993 and showed homology with the angiotensin II type 1 receptor [1,2]. However, APJ, a seven-transmembrane G protein-coupled receptor (GPCR), did not bind to angiotensin II [2] and was initially considered as an orphan GPCR [1,2]. Its first endogenous ligand, the peptide hormone apelin, was discovered several years later in 1998 by Tatemoto et al. by means of monitoring APJ activity from bovine stomach extracts [3].
APJ and the preproapelin, consisting of 77 amino acid residues, are expressed in embryo and adult human tissues, including heart, vasculature (particularly in endothelial cells), and lung tissue; white adipose tissue; the gastrointestinal tract and the liver; several regions of the central nervous system; retinas; limbs; the skin; kidneys; mammary glands; and placental tissue [1,4,5,6,7,8,9,10,11,12,13,14]. The preproapelin can be cleaved from its C-terminal domain to produce several apelin peptides with different polypeptide chain lengths (apelin-36, apelin-17, and apelin-13). Research has shown that the longer chains of this protein are characterized by lower biological activity, which is why they are converted into short-chain forms [15]. Apelin-36 predominates in rat lung, testis, and uterus [16] and in bovine colostrum [3]. Its concentration is much lower in rat brain as well as in rat and human plasma, where the most abundant forms of apelin are apelin-17 and pyroglutamate-apelin-13 [17,18]. The naturally pyroglutamated apelin-13 form is structurally more resistant to aminopeptidases and is also the most active isoform. It is located in the mammary gland and hypothalamus [16], but also in the heart, where it is the most abundant form [19].
A second endogenous ligand, ELA, was identified in 2013 in zebrafish embryos [20,21] by Chng et al. While seeking to identify the first hormonal peptide implicated in the ability of naive blastomeres to differentiate into one of the three embryonic germ layers, they isolated a human gene named ‘APELA’ (apelin early endogenous ligand), annotated until then as a noncoding transcript. APELA was predicted to encode a hormone with a signal peptide, ELA [20]. Concurrently, Pauli et al. also identified the same gene and named it ‘TODDLER’ [21]. Thus, even if they both bind to APJ, ELA and apelin differ not only in their structure [22] but also by their encoding genes, which is rather unusual for peptide ligands of the same GPCR. ELA is the early ligand in humans, but it remains present in blood during adulthood by means of its expression in the prostate, the kidney, the cardiac endothelium, blood vessels, and the placenta [20,23,24,25,26,27]. Its crucial role in early human development will be further reviewed in Section 2.2.
ELA is a 54-amino acid preprotein processed in different isoform lengths: ELA-32, ELA-22, ELA-11 and, probably, ELA-14 and ELA-21. More precisely, as a result of proteolysis, the ELA sequence is cleaved by furin, generating ELA-11 and ELA-21 [20]. However, cleavage of the signal peptide in the N-terminus produces a 32-amino acid proprotein. ELA-32 is a mature form that becomes a biologically active molecule upon binding to APJ, similar to other isoforms [20]. Although putative furin cleavage sites were predicted to generate the other shorter peptides previously cited [27,28], the detection of a small number of them still needs to be proven in vivo.
Further research is still necessary to identify preponderant ELA and apelin isoforms and the mechanisms regulating their production, especially during physiological and pathological pregnancy. However, the high conservation of APJ, apelin, and ELA suggests that the apelinergic system is a key regulator of essential physiological functions [20,29].

2. The Apelinergic System in the Reproductive System—Pregnancy and Postpartum

2.1. Reproductive System

The topographical distribution of apelinergic-synthesizing neurons in rats [30] and the hypothalamic localization of apelin fibers and receptors [31] have suggested an implication of the apelinergic axis in behavior control and pituitary hormone release [32]. Its implication in reproductive regulation was further supported by the findings of Pope et al., who reported high levels of APJ mRNA and apelin binding sites in the mouse uterine endometrium and ovary [33]. In addition, the corpus luteum presented a high level of APJ expression. These observations suggest that the intraovarian apelinergic system may have an autocrine role [33].
Apelin and APJ are also present in bovine granulosa and oocytes. Apelin increases the secretion of basal and insulin-like growth factor 1 (IGF-1)-induced progesterone in bovine luteinizing granulosa cells, whereas it inhibits oocyte maturation and progesterone secretion from cumulus cells in vitro [34]. Accordingly, in a porcine model, apelin also increased the secretion of basal and IGF1- and FSH-induced progesterone and estradiol secretion, with an increased expression of both apelin and APJ with follicular growth [35]. In the human ovary, the apelinergic axis is localized through different developmental stages, including luteinized human granulosa cells, theca, oocytes, and the corona cumulus complex [36]. In cultured human luteinized granulosa cells, IGF-1 increased APJ expression, and recombinant human apelin stimulated the secretion of both basal and IGF1-induced progesterone and estradiol secretion [36]. The coherence of former data suggests that the apelinergic system, more specifically apelin, plays several roles in the hypothalamus–pituitary–gonadal axis and in the female reproductive organs, thus highlighting a crucial involvement in steroidogenesis [37].

2.2. Development of the Embryo

In human embryonic stem cells (hESC), ELA can potentiate the TGF-β pathway to prime hESCs toward the endoderm lineage [38]. It is abundantly secreted by undifferentiated hESCs, which do not express APJ [38], thus implying that ELA might use a secondary receptor [39]. ELA also appears to be an important endogenous growth factor in human embryos with a crucial role in maintaining the growth and self-renewal of human and mouse ESCs [38], which have a key function in maintaining genome stability. ELA facilitates hESC cell-cycle progression, as well as protein translation, and suppresses stress-induced apoptosis [38]. Accordingly, the inhibition of ELA causes decreased cell growth, cell death, and loss of pluripotency in hESC [38].
The apelinergic system has a complex spatiotemporal regulation in embryology, which needs to be fully elucidated and appears to be species-specific, making it difficult to extrapolate from animal models to human physiology. For example, Freyer et al. observed that, in contrast to zebrafish, ELA is not the first apelinergic ligand to be expressed in mice [40]. In fact, in mice, apelin is first expressed in extraembryonic visceral endoderm and the primitive streak at embryonic day (E) 6.5, whereas APELA expression is detected at E7.0 in the distal epiblast and shortly thereafter in the definitive endoderm [41]. At E8.25, APELA is expressed in extraembryonic tissues and in the chorion and at E9.5 in peripheric trophoblast cells [40]. These authors also observed APJ in the allantois and the vasculature invading the placenta at E9.5, which suggests that apelinergic signaling may function in extraembryonic and embryonic tissues, with an impact on the formation of mesoderm derivatives, such as yolk sac vasculature, hematopoietic progenitors, the chorion, and the allantois [40]. Interestingly, apelin knockout mice do not exhibit the endoderm defects [20] found in zebrafish [20,21], thus suggesting non-conserved roles in vertebrate gastrulation, which could be due to species-specific mechanisms of mesendoderm migration [40]. Apelin was also observed at the end of gastrulation during zebrafish heart development [20,42,43].
ELA is also a key factor in the process of gastrulation. Notably, knockdown of APELA in zebrafishes resulted in the reduced movement of ventral and lateral mesendodermal cells during gastrulation [21]. Indeed, during gastrulation, ELA increases cell velocity in a nondirectional manner toward progress in mesendoderm internalization [21]. Moreover, in zebrafish, it is also involved in guided cell migration by driving angioblast migration to the midline in dorsal aorta formation [44]. In embryo development, the ELA/APJ pathway is also implicated in skeletal development, bone formation, and bone homeostasis [45].
By contrast, ELA is essential for the proper differentiation of endodermal precursors that are known to be crucial for guiding the overlying cardiac progenitors to the heart-forming region [20]. The presence in zebrafish embryos of the grinch mutation, localized in the APLNR zebrafish ortholog, often results in the complete absence of cardiomyocytes, thus highlighting the critical role played by APLNR in myocardial development [46]. Indeed, APLNR knockdown 1-cell embryos and APLNR-deficient mice also show higher lethality due to cardiovascular abnormalities [4,5,43,46,47]. Moreover, later cardiovascular defects in adulthood were observed in most surviving mice embryos [4,5].
Paskaradevan et al. [47] demonstrated a novel mechanism for APLNR signaling in the establishment of a niche required for the proper development of zebrafish myocardial progenitor cells via the activation of Gata5/Smarcd3. However, despite the fact that apelin−/− mutants exhibited cardiac developmental defects, apelin-/- zebrafish [43] and mice [4,48] remained viable and fertile, suggesting that another APJ ligand other than apelin could be involved in embryonic development, i.e., ELA. The implication of ELA was later confirmed as its loss of function in zebrafish [20], and the mouse [24] model produced similar results to APJ deletion, i.e., partial embryonic lethality and cardiovascular defects. Indeed, Chng et al. observed that the loss of ELA in zebrafish embryos caused the development of a rudimentary heart or no heart at all [20]. The authors proposed a zebrafish model in which APLNR is required to fine-tune nodal output, acting as a specific rheostat for the Nodal/TGFβ pathway during the earliest stages of cardiogenesis [49]. In mice, ELA deficiency inhibits embryo blood vessel remodeling and suppresses the angiogenic sprouting of vitelline vessels, dorsal aorta, and outflow-tract and inter-somitic vessels [50]. Moreover, ELA deficiency further causes angiogenesis defects in the mouse embryo through the promotion of the expression of the endothelial cell-specific molecule 1 (ESM1) gene [50]. Likewise, in human embryos, ELA has been proposed as an endogenous secreted growth factor for hESCs that activates the TGF-β pathway to promote vasculogenesis [51].
Globally, the Elabela/APJ axis induces cardiogenesis, vasculogenesis, and bone formation during embryonic development. Furthermore, in adults, it also enhances cardiac contractility, promotes vasodilatory effects, mediates fluid homeostasis, and reduces food intake. In addition, the apelin/APJ axis is involved in embryonic vascular, ocular, and heart development [52]. Apelin has actions on blood pressure [53,54] and vasodilatation, and it has a stimulatory effect on endothelial cell proliferation that may be involved in blood vessel diameter during angiogenesis [55,56]. Of note, these cardiovascular effects of the apelinergic system in adults have not yet been studied during pregnancy.

2.3. The Apelinergic System in Placenta

In zebrafish, APELA is first expressed in trophoblasts and is robustly upregulated after allantoic fusion, which occurs at an early phase of placental vascular development [24]. After E10.5, ELA becomes restricted to the syncytiotrophoblasts (STBs) juxtaposed to APJ-expressing fetal endothelial cells, suggesting a paracrine mode of action [24].
Georgiadou et al. observed that, in first trimester human placentas, both ELA and apelin were expressed in villous cytotrophoblasts (CTBs), in STBs, and in distal column extravillous CTBs (EVTs), while APJ was expressed in villous CTBs and distal column EVTs, but not in STBs [57]. In addition, they observed strong ELA expression in stromal cells of term placentas [57] in some samples, indicating that trophoblasts are not the only source of placental ELA and that stromal cells might also play a functional role. However, in contradiction to this study, Inusuka et al. observed strong APJ expression on the cellular membranes of first trimester STB, whereas weaker expression was detected in villous CTBs and EVTs [58]. In addition, APJ location varies throughout human gestation. At the beginning of pregnancy, it is mainly located on the cellular membranes of STB and EVTs, and in the second and third trimesters, its presence is more pronounced in the cytoplasm of STBs [58].
The expression of apelin was also observed in the cytoplasm of the blood capillaries, the endothelium, and the placental arteries in term placentas [59]. The apelinergic system might therefore play a role in placental development, such as cell differentiation, proliferation, apoptosis, and invasion (Figure 1).

2.3.1. Differentiation

The development of the placenta depends on the coordination of the proliferation and differentiation of trophoblast cells [60,61]. Each differentiation stage may be related to impaired placental development and cause placental-related pregnancy complications, highlighting the central role of differentiation in their pathogenesis [62]. ELA plays a key role in the regulation of the differentiation stage of human EVTs, including transition from a proliferative to an invasive phenotype [57]. Abnormal EVT differentiation leads to impaired invasion into the decidua by interstitial EVTs and the altered remodeling of spiral arteries by endovascular EVTs. The failure of the physiological transformation of spiral arteries has a role in preeclampsia (PE).

2.3.2. Cell Cycle and Proliferation

The human trophoblast cells must exit the cell cycle in order to differentiate and fuse to form multinucleate STBs [63]. Studies have shown that the depletion of a cell cycle inhibitor (p21) could lead to the reduced expression of fusion-related genes, which adversely affects the fusion capability of trophoblastic cells [64]. Increasing evidence emphasizes the major roles of cell cycle regulators in trophoblast cell division and differentiation [65]. Several cell cycle regulators are expressed in human placenta, with distinct and dynamic expression levels [66]. Apelin-13 treatment alters cyclin expression by particularly stimulating the expression of cyclins D and E and thus the cell cycle progression in both JEG-3 and BeWo cells [59]. It has also been demonstrated that apelin-13 promotes JEG-3 proliferation via APJ and the extracellular signal-regulated kinases (ERK)1 and 2, the signal transducer and activator of transcription 3 (STAT3), and the adenosine monophosphate-activated protein kinase alpha (AMPKα) signaling pathways [59] (Figure 2). Similarly, Ma et al. recently found that ELA promoted the proliferation of BeWo cells [67].

2.3.3. Cell Survival

ELA and apelin can also exert anti-apoptotic effects on BeWo cells by the activation of the PI3K-Akt pathway [67,69] (Figure 2). The apelin/APJ system increases the expression of pro-survival and decreased proapoptotic factors on mRNA and protein levels in both BeWo cells and villous explants [69]. Ferroptosis, a programmed cell death caused by iron-dependent peroxidation of lipids, might be rescued by ELAs by disrupting ferritinophagy and increasing ferritin heavy chain (FTH1) in HTR-8/SVneo cells. Interestingly, some authors report an increased grade of ferroptosis accompanied by a downregulation of the expression of ELA in PE placentas and further confirm an increased grade of ferroptosis together with a downregulation of ELA in PE-like model mouse placentas, thus providing new insights into the mechanism and therapeutic targets of PE [70].

2.3.4. Trophoblastic Invasion

Abnormal EVT invasion into the decidua led to an alteration of spiral artery remodeling by endovascular EVTs and, ultimately, to utero–placental insufficiency. The addition of ELA in the culture medium of the choriocarcinoma cell line JAR was reported to increase their invasiveness in transwell invasion assays [24]. It has also been shown that the treatment of HTR-8/SVneo with apelin or ELA also increased their invasiveness [57] and is dependent on APJ [71]. In addition, ELA induces the invasion and migration of HTR-8/SVneo cells through the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathway [72] (Figure 2).

2.3.5. Placental Hormone Secretion

The apelinergic system might be implicated in the production and secretion of placental hormones [73], which is probably the reason why they vary through pregnancy [74]. Apelin could decrease the secretion of protein hormones through the protein kinase A (PKA) and extracellular signal-regulated kinases (ERK1/2) signaling pathways (Figure 2) [68].

2.4. Labor

Apelin has been shown to inhibit human uterine contractility in vitro [75], suggesting its potential role in parturition. In rats, apelin levels were increased at the end of pregnancy and induced myometrium contractions, with their frequency and amplitude depending on its concentration. This effect does not occur with the PKC inhibitor, indicating that the PKC pathway might be implicated in its mechanism of action [76]. By contrast, an in vitro study showed that apelin suppresses both spontaneous and oxytocin-induced contractions in human myometrial fibers [75]. These contradictory results may be explained by the intracellular balance between vascular dilatation and the smooth-muscle contraction mechanisms of the apelinergic system, as well as the impact of species diversity and reagent concentrations [37].
Higher concentrations of apelin have been found in pregnant women with obesity during pregnancy, which could explain their decreased myometrial contractility, potentially due to the inhibition of the myometrial RhoA/ROCK (RhoA kinase) pathway [77]. Women with obesity have a higher frequency of cesarean sections compared to non-obese women, which is associated with an altered myometrial function that leads to a lower frequency and potency of contractions. The association of apelin and lower uterine contractility in pregnant women with obesity deserves further evaluation. Regarding ELA, neither its expression in the uterus nor its role in myometrium contractility has yet been reported.

2.5. The Apelinergic System and Postpartum/Breastfeeding

Apelin is abundant in breastmilk [78,79] and its level increases with long- and short-term overnutrition, possibly via maternal hyperinsulinemia and the transcriptional upregulation of apelin expression in the myoepithelial cells of the mammary gland [80]. Interestingly, the apelin level is lower in the breast milk of lactating women who have gestational diabetes [79]. At present, little is known regarding the mRNA or protein expression of APELA and ELA in the mammary gland in any mammalian species.

3. Placenta-Related Complications

The apelinergic system has a central role in early placentation. Early placentation dysfunction is a known trigger mechanism for placenta-related pregnancy complications. We review here current knowledge on the possible implication of the apelinergic system in several complications.

3.1. Preeclampsia (PE)

PE is a hypertensive disorder with multiple organ involvement. It affects 5% to 8% of all pregnancies [81] and remains the leading cause of fetal and maternal morbidity and mortality. PE and related disorders cause 14% of maternal deaths each year globally [82]. However, authors suggest that the addition of angiogenic markers to the conventional diagnostic criteria would improve the detection rate of both maternal and perinatal adverse outcomes [83]. In mice, ELA deficiency leads to hallmarks of PE such as hypertension, proteinuria, glomerular endothelial cell hyperplasia, and low birthweight (i.e., intrauterine growth restriction [IUGR]) [24], making ELA-deficient animals a suitable model for the study of PE, as well as the involvement of ELA in the pathogenesis of PE [84].
ELA deficiency in mice causes placental dysfunction characterized by a thin labyrinth, poor angiogenesis, increased apoptosis, decreased proliferation, and delayed STB differentiation [24]. In addition, circulating ELA levels correlate with the severity of maternal proteinuria and kidney damage. Interestingly, the infusion of exogenous ELA normalizes hypertension and proteinuria in ELA-deficient pregnant mice [24], suggesting that circulating ELA participates in maternal cardiovascular and renal adaptations to pregnancy independently of other well-known PE angiogenic factors (soluble fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor [sFlt1/PlGF]) [24]. Moreover, Ma et al. showed that ELA significantly reversed NG-nitro-l-arginine methyl ester (L-NAME)-induced hypertension in mice, reversed the condition of maternal blood sinuses narrowing (in the placental labyrinth zone), and regulated the expression of mouse placental apoptosis factors [67]. L-NAME is a nitric oxide synthase inhibitor that disrupts uterine spiral artery remodeling in pregnant animals and increases placental vasoconstriction and vascular reactivity, and it thus decreases blood flow, leading to placental ischemia [85,86,87]. Treating pregnant rodents in their second and third trimesters with L-NAME results in hypertension, proteinuria, renal damage, IUGR, and thrombocytopenia [88,89,90].
In humans, ELA data are highly contradictory. At the protein level, translational studies do not support the hypothesis that human PE is characterized by an early deficiency in circulating ELA levels. There is no association between circulating ELA-32 in maternal blood and preterm PE [91,92]. By contrast, placental and circulating ELA-32 have been found to be elevated in two studies including women with late-onset PE [92,93] and decreased in another study by Zhou et al. [71]. In addition, Georgiadou et al. found significantly lower levels of circulating ELA in women with a normal body mass index (BMI) who later developed late-onset PE compared to women with uncomplicated pregnancies, while levels in early-onset PE did not reach statistical significance [57]. The authors suggested that ELA could not be used as a first trimester PE screening biomarker due to the large variability and dependence of ELA levels on BMI. Indeed, the study by Zhou et al. included women with a mean BMI < 25, while the study by Panaitescu et al. included a majority of women with a BMI > 25.
Another study reporting on the screening of APELA variants in PE women versus controls concluded that two rare variants were found only in PE cases, suggesting that women who express these rare variants might have a reduced transcription of the protein, which could result in an increased risk of PE [94]. Thus, the apelinergic system could be impaired in a very specific subset of women with PE, and future research should focus on their identification. More studies are also needed to identify whether specific ELA isoforms are dysregulated before the diagnosis of PE [95], but specific enzyme-linked immunosorbent assay (ELISA) tests would be required. To date, only two different ELISA tests are available for this purpose [96]. Pritchard et al. [31] and Panaitescu et al. [32] used the most frequently used one, the kit from Peninsula Laboratories (Peninsula Laboratories International, Inc., BMA Biomedicals, Augst, Switzerland), which is said to react for ELA-32, whereas Villie et al. [53] chose the kit from Creative Diagnostics (Shirley, NY, USA), which cross-reacts with human ELA-21 and ELA-32, and is more expensive.
Regarding apelin, Hamza et al. found that PE-induced rats (L-NAME) showed significantly decreased apelin serum levels [97]. Moreover, they observed significantly increased blood pressure and urine proteins. These parameters negatively correlated with the serum apelin level, and exogenous apelin-13 administration significantly improved them, together with an improvement in the placental histoarchitecture [97]. Accordingly, in reduced uterine perfusion pressure PE-induced rats, Wang et al. observed that apelin-13 treatment significantly improved the symptoms of PE, suggesting that apelin may be a potential target for treating PE [39].
There are also contradictions between the clinical results regarding the expression of apelin and its circulating levels and correlation with PE. Several studies have shown that the serum apelin level was increased in PE [98,98,99], whereas other studies showed decreased apelin mRNA and proteins in PE placentas [58] or maternal blood [100], serum [101,102], or plasma [103] levels compared with normotensive pregnancies [58,71,100,101,103,104]. However, most authors have assessed total apelin levels using the same Apelin-36 ELISA kit from Phoenix Pharmaceuticals (Burlingame, CA, USA) [96], which targets the last 12 amino acids of all isoforms and thus is said to cross-react with apelin-12, apelin-13 and apelin-36. Nevertheless, two teams also studied apelin-13 and also found decreased serum levels [104,105]. On the other hand, a recent review determined that, although there was a high heterogeneity within available studies, there was no difference in circulating maternal apelin levels between the two patient groups [106]. Nevertheless, it was observed that patients with PE had a higher BMI and lower gestational age and birthweight at delivery. When performing a subgroup analysis, PE women with a higher BMI had significantly lower apelin levels, whereas there was no significant apelin difference depending on PE severity.
Data about the apelinergic system levels in newborns are still critically lacking. However, it was demonstrated that ELA and apelin levels were decreased in newborns’ venous-arterial cord blood in women with PE and severe PE compared with healthy pregnant women [100].

3.2. Intrauterine Growth Restriction (IUGR)

IUGR, also called fetal growth restriction, is defined as the failure of the fetus to reach its genetically established growth potential [107,108] and is diagnosed in approximately 10% of pregnancies [109]. Malamitsi-Puchner et al. found the presence of markedly high concentrations of apelin in umbilical plasma samples, which suggests a potential role for this peptide in intrauterine growth [110]. Subsequently, it was observed that apelin levels were decreased in IUGR serum and placenta staining [111] compared to uncomplicated pregnancies or to pregnancies complicated by PE, but the study sample was too small (four cases of IUGR) to reach any conclusion. Apelin is known to stimulate proliferation and inhibit apoptosis in mouse and human osteoblasts [112], which could be a potential mechanism linking apelin and fetal growth.
As mentioned previously, ELA levels were correlated with birthweights in mice [24]. In humans, ELA serum levels have been found to be lower in cases of IUGR in one study [113] but higher in another [114]. These contradictory results might be explained by different IUGR inclusion criteria (estimated fetal weight below the third percentile in the study by Berham et al. and fetal abdomen circumference measurement below the 10th percentile in the study by Yener et al.) and different gestational ages at sample collection (at approximately 30 weeks and at delivery date for Berham, and at approximately 36 weeks for Yener). In addition, Berham et al. excluded hypertensive patients, but Yener et al. did not.

3.3. Gestational Diabetes Mellitus (GDM)

Apelin is known to play a role in blood glucose metabolism [56]. Two studies have shown an increase in the apelin serum level of GDM pregnant women [74,115], whereas other studies reported either decreased concentrations [115,116,117] or an absence of any difference [118,119,120,121]. Other authors studied specifically the second and third trimesters of pregnancy and found that ELA serum levels were decreased in GDM, whereas apelin serum levels increased [74]. Dasgupta et al. reported that apelin expression in GDM placentas was significantly reduced compared with matched controls [122]. Moreover, GDM mice treated with apelin showed a significant improvement in inflammatory cytokines, oxidative stress in the placenta, and glucose and lipid metabolism [123]. This suggests that the apelinergic system pathway is a promising target for the development of prophylactic and therapeutic agents for GDM in the future. However, the data are still inconsistent and more studies are required.

3.4. Miscarriage

Spontaneous abortions are multifactorial, but apart from genetic causes, a placental implication is plausible [124]. Placental histological changes have been reported in this field, but also delays in trophoblast development, impairment in villous vasculogenesis–angiogenesis [125], and insufficient syncytialization [126]. ELA-like APLNR null mice [40] and zebrafish [20] have reduced survival, probably mainly due to heart development and placental defects, but little is known about the direct influence of the apelinergic system on spontaneous abortion. To our knowledge, there is only one publication demonstrating an association of lower maternal ELA levels with spontaneous abortion [127].

4. From Research to the Clinical Setting: Challenges and Limitations

Studies have reported an alteration of circulating and placental ELA and apelin levels in pathologies of pregnancy such as PE and GDM, suggesting that these peptides could be used as biomarkers (Table 1). However, the results of these studies vary significantly (Table 1). First, obtaining specific proper dosages of apelinergic ligands is challenging. Apelin and ELA protein levels have mainly been evaluated using commercial antibody-based immunoassays against various synthetic peptide fragments [73,128,129]. These assays are probably not isoform-specific [130]; this may contribute to the reported large plasma concentrations ranges observed. High-performance liquid chromatography combined with radioimmunoassay detection has also been used, with a confirmed relative specificity for different apelin isoforms [131,132,133], but only a small number of authors have successfully detected apelin in vivo [130,134,135].
Second, the variability of results using the same commercial antibody-based immunoassays suggests critical variances in the specificity of ELA tests from different manufacturers. Indeed, using the same ELISA kit, Pritchard et al. [91] and Panaitescu et al. [92] found different ELA concentrations in samples collected at term (~30 pg/mL and 5 ng/mL, respectively).
A third limitation of the evaluation of the apelinergic system is the lack of guidelines and recommendations for sample handling procedures, such as sample extraction, to ensure the correct use of the ELISA kit. Georgiadou et al. [128] reviewed eight studies reporting data on circulating ELA in human plasma or serum. These studies used ELISA kits from three different companies (Phoenix Pharmaceuticals, Peninsula Laboratories International, and Creative Diagnostics) all recommending sample peptide extraction by means of high performance liquid chromatography. In two studies [25,141], it appears that sample extraction was not performed. In the other studies, the procedures for sample extraction were unclear, making their results difficult to interpret. Interestingly, when the authors indicated that peptide extraction was performed, the results obtained by the commercial kit mirrored the levels obtained by the custom ELISA used by Georgiadou et al., whereas when peptide extraction was not performed, the ELA levels no longer showed their typical large inter-individual variations. In addition, all isoforms have a short half-life, probably of only a few minutes [28], thus implying a rapid and transient response to homeostatic changes and differences in potency [142] and signaling pathways, resulting in difficulties in establishing their correct dosage and in their subsequent use as a therapeutic agent. Thus, further studies are needed to establish guidelines for sample handling and for the measurement and conservation of apelinergic system isoforms.
Another limitation is the variation in the population’s intrinsic characteristics. Apelin varies according to body mass index [143,144], patient age [145], and the presence of diabetes [146,147], or in cases of an inflammatory condition, such as psoriasis [148]. Variations linked to thyroid disorders have also been evaluated [149,150] but are not conclusive. Moreover, apelinemia could decrease during the second half of pregnancy [151,152]. Additionally, as the apelin gene is located on the X chromosome, analyses are sometimes conducted in a sex-specific manner. For example, specific apelin genotypes were associated with lower high-density lipoprotein cholesterol in Iranian women without a metabolic syndrome [153]. Likewise, gender-specific associations between apelin/APJ gene polymorphisms have been highlighted in humans [154]. In the APLNR knockout mouse model, sex-specific effects on conditioned fear responses were observed [155].
ELA levels correlate with age, BMI, heart rate, BNP levels, and left atrial dimension [156]. ELA levels also appears to be higher during pregnancy [24,25] and linked to gestational age [128]. In 50% of women in the resistance to aspirin during and after pregnancy (RADAR) cohort, ELA could not be detected throughout pregnancy and the postpartum period [57]. However, Panaiteiscu et al. were able to detect ELA in the plasma of women during the second and third trimesters [92]. Of note, ELA levels were highly variable between women and did not particularly change during pregnancy, although a slightly higher level was observed in the first trimester [57]. Finally, both the apelin and the ELA peptide levels might be altered in some other pathologies of pregnancy such as GDM, which is an issue that must be considered.
Preclinical studies have also evaluated these peptides for the treatment of PE. Regardless of the rat PE models used, the administration of apelin or ELA peptides significantly improved the symptoms of PE (Table 2). However, these studies were only conducted in rats with PE, and these results should be confirmed in other animal models. Indeed, the spatiotemporal regulation of the apelinergic system in embryology appears to be species-specific [40,41], making it difficult to extrapolate from animal models to human physiology.

5. Conclusions

The apelinergic system in the reproductive field plays a central role in both physiological pregnancy and placenta-related complications in pregnancy. More specifically, apelin has a crucial role in steroidogenesis and in the metabolic regulation of the ovary. ELA also has a potential role in the onset of PE. These data suggest that the apelinergic system is a promising research field with a translational potential related to therapeutic interventions in pregnancy. Further studies investigating the role of the apelinergic system in the development of early pregnancy development and complications remain necessary to fully understand its role and its potential in the development of therapeutic strategies.

Author Contributions

Conceptualization, methodology, writing—original draft preparation and review: O.P.; supervision: M.C. and B.M.d.T.; review and editing: all authors. O.P., A.C.-B., M.C. and B.M.d.T.; review and editing: all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. O’Carroll, A.-M.; Lolait, S.J.; Harris, L.E.; Pope, G.R. The apelin receptor APJ: Journey from an orphan to a multifaceted regulator of homeostasis. J. Endocrinol. 2013, 219, R13–R35. [Google Scholar] [CrossRef] [PubMed]
  2. O’Dowd, B.F.; Heiber, M.; Chan, A.; Heng, H.H.Q.; Tsui, L.-C.; Kennedy, J.L.; Shi, X.; Petronis, A.; George, S.R.; Nguyen, T. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene 1993, 136, 355–360. [Google Scholar] [CrossRef]
  3. Tatemoto, K.; Hosoya, M.; Habata, Y.; Fujii, R.; Kakegawa, T.; Zou, M.-X.; Kawamata, Y.; Fukusumi, S.; Hinuma, S.; Kitada, C.; et al. Isolation and Characterization of a Novel Endogenous Peptide Ligand for the Human APJ Receptor. Biochem. Biophys. Res. Commun. 1998, 251, 471–476. [Google Scholar] [CrossRef]
  4. Charo, D.N.; Ho, M.; Fajardo, G.; Kawana, M.; Kundu, R.K.; Sheikh, A.Y.; Finsterbach, T.P.; Leeper, N.J.; Ernst, K.V.; Chen, M.M.; et al. Endogenous regulation of cardiovascular function by apelin-APJ. Am. J. Physiol.-Heart Circ. Physiol. 2009, 297, H1904–H1913. [Google Scholar] [CrossRef]
  5. Kang, Y.; Kim, J.; Anderson, J.P.; Wu, J.; Gleim, S.R.; Kundu, R.K.; McLean, D.L.; Kim, J.; Park, H.; Jin, S.; et al. Apelin-APJ Signaling Is a Critical Regulator of Endothelial MEF2 Activation in Cardiovascular Development. Circ. Res. 2013, 113, 22–31. [Google Scholar] [CrossRef] [PubMed]
  6. Kasai, A.; Shintani, N.; Kato, H.; Matsuda, S.; Gomi, F.; Haba, R.; Hashimoto, H.; Kakuda, M.; Tano, Y.; Baba, A. Retardation of retinal vascular development in apelin-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1717–1722. [Google Scholar] [CrossRef]
  7. Mayeur, S.; Wattez, J.-S.; Lukaszewski, M.-A.; Lecoutre, S.; Butruille, L.; Drougard, A.; Eberlé, D.; Bastide, B.; Laborie, C.; Storme, L.; et al. Apelin Controls Fetal and Neonatal Glucose Homeostasis and Is Altered by Maternal Undernutrition. Diabetes 2015, 65, 554–560. [Google Scholar] [CrossRef] [PubMed]
  8. Hosoya, M.; Kawamata, Y.; Fukusumi, S.; Fujii, R.; Habata, Y.; Hinuma, S.; Kitada, C.; Honda, S.; Kurokawa, T.; Onda, H.; et al. Molecular and functional characteristics of APJ. Tissue distribution of mRNA and interaction with the endogenous ligand apelin. J. Biol. Chem. 2000, 275, 21061–21067. [Google Scholar] [CrossRef] [PubMed]
  9. Kleinz, M.J.; Davenport, A.P. Immunocytochemical localization of the endogenous vasoactive peptide apelin to human vascular and endocardial endothelial cells. Regul. Pept. 2004, 118, 119–125. [Google Scholar] [CrossRef]
  10. Kleinz, M.J.; Skepper, J.N.; Davenport, A.P. Immunocytochemical localisation of the apelin receptor, APJ, to human cardiomyocytes, vascular smooth muscle and endothelial cells. Regul. Pept. 2005, 126, 233–240. [Google Scholar] [CrossRef]
  11. O’Carroll, A.M.; Selby, T.L.; Palkovits, M.; Lolait, S.J. Distribution of mRNA encoding B78/apj, the rat homologue of the human APJ receptor, and its endogenous ligand apelin in brain and peripheral tissues. Biochim. Et Biophys. Acta (BBA)-Gene Struct. Expr. 2000, 1492, 72–80. [Google Scholar] [CrossRef]
  12. Medhurst, A.D.; Jennings, C.A.; Robbins, M.J.; Davis, R.P.; Ellis, C.; Winborn, K.Y.; Lawrie, K.W.M.; Hervieu, G.; Riley, G.; Bolaky, J.E.; et al. Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin. J. Neurochem. 2003, 84, 1162–1172. [Google Scholar] [CrossRef] [PubMed]
  13. Dray, C.; Debard, C.; Jager, J.; Disse, E.; Daviaud, D.; Martin, P.; Attané, C.; Wanecq, E.; Guigné, C.; Bost, F.; et al. Apelin and APJ regulation in adipose tissue and skeletal muscle of type 2 diabetic mice and humans. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E1161–E1169. [Google Scholar] [CrossRef] [PubMed]
  14. He, L.; Xu, J.; Chen, L.; Li, L. Apelin/APJ signaling in hypoxia-related diseases. Clin. Chim. Acta 2015, 451 Pt B, 191–198. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, M.M.; Ashley, E.A.; Deng, D.X.F.; Tsalenko, A.; Deng, A.; Tabibiazar, R.; Ben-Dor, A.; Fenster, B.; Yang, E.; King, J.Y.; et al. Novel role for the potent endogenous inotrope apelin in human cardiac dysfunction. Circulation 2003, 108, 1432–1439. [Google Scholar] [CrossRef] [PubMed]
  16. Kawamata, Y.; Habata, Y.; Fukusumi, S.; Hosoya, M.; Fujii, R.; Hinuma, S.; Nishizawa, N.; Kitada, C.; Onda, H.; Nishimura, O.; et al. Molecular properties of apelin: Tissue distribution and receptor binding. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2001, 1538, 162–171. [Google Scholar] [CrossRef]
  17. Nyimanu, D.; Kay, R.G.; Sulentic, P.; Kuc, R.E.; Ambery, P.; Jermutus, L.; Reimann, F.; Gribble, F.M.; Cheriyan, J.; Maguire, J.J.; et al. Development and validation of an LC-MS/MS method for detection and quantification of in vivo derived metabolites of [Pyr1]apelin-13 in humans. Sci. Rep. 2019, 9, 19934. [Google Scholar] [CrossRef]
  18. Shin, K.; Landsman, M.; Pelletier, S.; Alamri, B.N.; Anini, Y.; Rainey, J.K. Proapelin is processed extracellularly in a cell line-dependent manner with clear modulation by proprotein convertases. Amino Acids 2019, 51, 395–405. [Google Scholar] [CrossRef] [PubMed]
  19. Maguire, J.J.; Kleinz, M.J.; Pitkin, S.L.; Davenport, A.P. [Pyr1]apelin-13 identified as the predominant apelin isoform in the human heart: Vasoactive mechanisms and inotropic action in disease. Hypertens. Dallas Tex 1979 2009, 54, 598–604. [Google Scholar] [CrossRef]
  20. Chng, S.C.; Ho, L.; Tian, J.; Reversade, B. ELABELA: A hormone essential for heart development signals via the apelin receptor. Dev. Cell 2013, 27, 672–680. [Google Scholar] [CrossRef]
  21. Pauli, A.; Norris, M.L.; Valen, E.; Chew, G.-L.; Gagnon, J.A.; Zimmerman, S.; Mitchell, A.; Ma, J.; Dubrulle, J.; Reyon, D.; et al. Toddler: An embryonic signal that promotes cell movement via Apelin receptors. Science 2014, 343, 1248636. [Google Scholar] [CrossRef]
  22. Couvineau, P.; Llorens-Cortes, C.; Iturrioz, X. Elabela/Toddler and apelin bind differently to the apelin receptor. FASEB J. 2020, 34, 7989–8000. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, Z.; Yu, D.; Wang, M.; Wang, Q.; Kouznetsova, J.; Yang, R.; Qian, K.; Wu, W.; Shuldiner, A.; Sztalryd, C.; et al. Elabela-apelin receptor signaling pathway is functional in mammalian systems. Sci. Rep. 2015, 5, 8170. [Google Scholar] [CrossRef] [PubMed]
  24. Ho, L.; van Dijk, M.; Chye, S.T.J.; Messerschmidt, D.M.; Chng, S.C.; Ong, S.; Yi, L.K.; Boussata, S.; Goh, G.H.-Y.; Afink, G.B.; et al. ELABELA deficiency promotes preeclampsia and cardiovascular malformations in mice. Science 2017, 357, 707–713. [Google Scholar] [CrossRef] [PubMed]
  25. Yang, P.; Read, C.; Kuc, R.E.; Buonincontri, G.; Southwood, M.; Torella, R.; Upton, P.D.; Crosby, A.; Sawiak, S.J.; Carpenter, T.A.; et al. Elabela/Toddler Is an Endogenous Agonist of the Apelin APJ Receptor in the Adult Cardiovascular System, and Exogenous Administration of the Peptide Compensates for the Downregulation of Its Expression in Pulmonary Arterial Hypertension. Circulation 2017, 135, 1160–1173. [Google Scholar] [CrossRef]
  26. Flahault, A.; Couvineau, P.; Alvear-Perez, R.; Iturrioz, X.; Llorens-Cortes, C. Role of the Vasopressin/Apelin Balance and Potential Use of Metabolically Stable Apelin Analogs in Water Metabolism Disorders. Front. Endocrinol. 2017, 8, 120. [Google Scholar] [CrossRef]
  27. Ma, Y.; Yue, Y.; Ma, Y.; Zhang, Q.; Zhou, Q.; Song, Y.; Shen, Y.; Li, X.; Ma, X.; Li, C.; et al. Structural Basis for Apelin Control of the Human Apelin Receptor. Structure 1993 2017, 25, 858–866.e4. [Google Scholar] [CrossRef]
  28. Murza, A.; Sainsily, X.; Coquerel, D.; Côté, J.; Marx, P.; Besserer-Offroy, É.; Longpré, J.-M.; Lainé, J.; Reversade, B.; Salvail, D.; et al. Discovery and Structure-Activity Relationship of a Bioactive Fragment of ELABELA that Modulates Vascular and Cardiac Functions. J. Med. Chem. 2016, 59, 2962–2972. [Google Scholar] [CrossRef]
  29. Pitkin, S.L.; Maguire, J.J.; Bonner, T.I.; Davenport, A.P. International Union of Basic and Clinical Pharmacology. LXXIV. Apelin receptor nomenclature, distribution, pharmacology, and function. Pharmacol. Rev. 2010, 62, 331–342. [Google Scholar] [CrossRef]
  30. Reaux, A.; Gallatz, K.; Palkovits, M.; Llorens-Cortes, C. Distribution of apelin-synthesizing neurons in the adult rat brain. Neuroscience 2002, 113, 653–662. [Google Scholar] [CrossRef]
  31. Newson, M.J.F.; Roberts, E.M.; Pope, G.R.; Lolait, S.J.; O’Carroll, A.-M. The effects of apelin on hypothalamic-pituitary-adrenal axis neuroendocrine function are mediated through corticotrophin-releasing factor- and vasopressin-dependent mechanisms. J. Endocrinol. 2009, 202, 123–129. [Google Scholar] [CrossRef] [PubMed]
  32. Kurowska, P.; Barbe, A.; Różycka, M.; Chmielińska, J.; Dupont, J.; Rak, A. Apelin in Reproductive Physiology and Pathology of Different Species: A Critical Review. Int. J. Endocrinol. 2018, 2018, 9170480. [Google Scholar] [CrossRef] [PubMed]
  33. Pope, G.R.; Roberts, E.M.; Lolait, S.J.; O’Carroll, A.-M. Central and peripheral apelin receptor distribution in the mouse: Species differences with rat. Peptides 2012, 33, 139–148. [Google Scholar] [CrossRef]
  34. Roche, J.; Ramé, C.; Reverchon, M.; Mellouk, N.; Rak, A.; Froment, P.; Dupont, J. Apelin (APLN) regulates progesterone secretion and oocyte maturation in bovine ovarian cells. Reprod. Camb. Engl. 2017, 153, 589–603. [Google Scholar] [CrossRef] [PubMed]
  35. Rak, A.; Drwal, E.; Rame, C.; Knapczyk-Stwora, K.; Słomczyńska, M.; Dupont, J.; Gregoraszczuk, E.L. Expression of apelin and apelin receptor (APJ) in porcine ovarian follicles and in vitro effect of apelin on steroidogenesis and proliferation through APJ activation and different signaling pathways. Theriogenology 2017, 96, 126–135. [Google Scholar] [CrossRef] [PubMed]
  36. Roche, J.; Ramé, C.; Reverchon, M.; Mellouk, N.; Cornuau, M.; Guerif, F.; Froment, P.; Dupont, J. Apelin (APLN) and Apelin Receptor (APLNR) in Human Ovary: Expression, Signaling, and Regulation of Steroidogenesis in Primary Human Luteinized Granulosa Cells. Biol. Reprod. 2016, 95, 104. [Google Scholar] [CrossRef]
  37. Wang, X.; Liu, X.; Song, Z.; Shen, X.; Lu, S.; Ling, Y.; Kuang, H. Emerging roles of APLN and APELA in the physiology and pathology of the female reproductive system. PeerJ 2020, 8, e10245. [Google Scholar] [CrossRef] [PubMed]
  38. Ho, L.; Tan, S.Y.X.; Wee, S.; Wu, Y.; Tan, S.J.C.; Ramakrishna, N.B.; Chng, S.C.; Nama, S.; Szczerbinska, I.; Sczerbinska, I.; et al. ELABELA Is an Endogenous Growth Factor that Sustains hESC Self-Renewal via the PI3K/AKT Pathway. Cell Stem Cell 2015, 17, 435–447. [Google Scholar] [CrossRef]
  39. Wang, C.; Liu, X.; Kong, D.; Qin, X.; Li, Y.; Teng, X.; Huang, X. Apelin as a novel drug for treating preeclampsia. Exp. Ther. Med. 2017, 14, 5917–5923. [Google Scholar] [CrossRef]
  40. Freyer, L.; Hsu, C.-W.; Nowotschin, S.; Pauli, A.; Ishida, J.; Kuba, K.; Fukamizu, A.; Schier, A.F.; Hoodless, P.A.; Dickinson, M.E.; et al. Loss of Apela Peptide in Mice Causes Low Penetrance Embryonic Lethality and Defects in Early Mesodermal Derivatives. Cell Rep. 2017, 20, 2116–2130. [Google Scholar] [CrossRef]
  41. Hassan, A.S.; Hou, J.; Wei, W.; Hoodless, P.A. Expression of two novel transcripts in the mouse definitive endoderm. Gene Expr. Patterns GEP 2010, 10, 127–134. [Google Scholar] [CrossRef] [PubMed]
  42. Devic, E.; Paquereau, L.; Vernier, P.; Knibiehler, B.; Audigier, Y. Expression of a new G protein-coupled receptor X-msr is associated with an endothelial lineage in Xenopus laevis. Mech. Dev. 1996, 59, 129–140. [Google Scholar] [CrossRef] [PubMed]
  43. Zeng, X.-X.I.; Wilm, T.P.; Sepich, D.S.; Solnica-Krezel, L. Apelin and Its Receptor Control Heart Field Formation during Zebrafish Gastrulation. Dev. Cell 2007, 12, 391–402. [Google Scholar] [CrossRef]
  44. Helker, C.S.M.; Schuermann, A.; Pollmann, C.; Chng, S.C.; Kiefer, F.; Reversade, B.; Herzog, W. The hormonal peptide Elabela guides angioblasts to the midline during vasculogenesis. Elife 2015, 4, e06726. [Google Scholar] [CrossRef]
  45. Lu, L.; Cao, J.; Li, L.; Chen, L. Elabela, a new endogenous ligand of APJ, functions in embryos and adults organisms. Acta Biochim. Biophys. Sin. 2017, 49, 378–381. [Google Scholar] [CrossRef] [PubMed]
  46. Scott, I.C.; Masri, B.; D’Amico, L.A.; Jin, S.-W.; Jungblut, B.; Wehman, A.M.; Baier, H.; Audigier, Y.; Stainier, D.Y.R. The G Protein-Coupled Receptor Agtrl1b Regulates Early Development of Myocardial Progenitors. Dev. Cell 2007, 12, 403–413. [Google Scholar] [CrossRef] [PubMed]
  47. Paskaradevan, S.; Scott, I.C. The Aplnr GPCR regulates myocardial progenitor development via a novel cell-non-autonomous, Gαi/o protein-independent pathway. Biol. Open 2012, 1, 275–285. [Google Scholar] [CrossRef]
  48. Kuba, K.; Zhang, L.; Imai, Y.; Arab, S.; Chen, M.; Maekawa, Y.; Leschnik, M.; Leibbrandt, A.; Markovic, M.; Schwaighofer, J.; et al. Impaired heart contractility in Apelin gene-deficient mice associated with aging and pressure overload. Circ. Res. 2007, 101, e32–e42. [Google Scholar] [CrossRef]
  49. Deshwar, A.R.; Chng, S.C.; Ho, L.; Reversade, B.; Scott, I.C. The Apelin receptor enhances Nodal/TGFβ signaling to ensure proper cardiac development. Elife 2016, 5, e13758. [Google Scholar] [CrossRef]
  50. Zhou, Q.; Zhang, K.; Guo, Y.; Chen, L.; Li, L. Elabela-APJ axis contributes to embryonic development and prevents pre-eclampsia in pregnancy. Acta Biochim. Biophys. Sin. 2018, 50, 319–321. [Google Scholar] [CrossRef]
  51. Liu, W.; Yan, J.; Pan, W.; Tang, M. Apelin/Elabela-APJ: A novel therapeutic target in the cardiovascular system. Ann. Transl. Med. 2020, 8, 243. [Google Scholar] [CrossRef] [PubMed]
  52. Piairo, P.; Moura, R.S.; Nogueira-Silva, C.; Correia-Pinto, J. The apelinergic system in the developing lung: Expression and signaling. Peptides 2011, 32, 2474–2483. [Google Scholar] [CrossRef]
  53. Tatemoto, K.; Takayama, K.; Zou, M.X.; Kumaki, I.; Zhang, W.; Kumano, K.; Fujimiya, M. The novel peptide apelin lowers blood pressure via a nitric oxide-dependent mechanism. Regul. Pept. 2001, 99, 87–92. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, Q.; Yao, F.; Raizada, M.K.; O’Rourke, S.T.; Sun, C. Apelin gene transfer into the rostral ventrolateral medulla induces chronic blood pressure elevation in normotensive rats. Circ. Res. 2009, 104, 1421–1428. [Google Scholar] [CrossRef]
  55. Kidoya, H.; Ueno, M.; Yamada, Y.; Mochizuki, N.; Nakata, M.; Yano, T.; Fujii, R.; Takakura, N. Spatial and temporal role of the apelin/APJ system in the caliber size regulation of blood vessels during angiogenesis. EMBO J. 2008, 27, 522–534. [Google Scholar] [CrossRef] [PubMed]
  56. Antushevich, H.; Wójcik, M. Review: Apelin in disease. Clin. Chim. Acta Int. J. Clin. Chem. 2018, 483, 241–248. [Google Scholar] [CrossRef]
  57. Georgiadou, D.; Boussata, S.; Ranzijn, W.H.M.; Root, L.E.A.; Hillenius, S.; de Weg, J.M.b.; Abheiden, C.N.H.; de Boer, M.A.; de Vries, J.I.P.; Vrijkotte, T.G.M.; et al. Peptide hormone ELABELA enhances extravillous trophoblast differentiation, but placenta is not the major source of circulating ELABELA in pregnancy. Sci. Rep. 2019, 9, 19077. [Google Scholar] [CrossRef]
  58. Inuzuka, H.; Nishizawa, H.; Inagaki, A.; Suzuki, M.; Ota, S.; Miyamura, H.; Miyazaki, J.; Sekiya, T.; Kurahashi, H.; Udagawa, Y. Decreased expression of apelin in placentas from severe pre-eclampsia patients. Hypertens. Pregnancy 2013, 32, 410–421. [Google Scholar] [CrossRef]
  59. Mlyczyńska, E.; Kurowska, P.; Drwal, E.; Opydo-Chanek, M.; Tworzydło, W.; Kotula-Balak, M.; Rak, A. Apelin and apelin receptor in human placenta: Expression, signalling pathway and regulation of trophoblast JEG-3 and BeWo cells proliferation and cell cycle. Int. J. Mol. Med. 2020, 45, 691–702. [Google Scholar] [CrossRef]
  60. Baczyk, D.; Drewlo, S.; Proctor, L.; Dunk, C.; Lye, S.; Kingdom, J. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death Differ. 2009, 16, 719–727. [Google Scholar] [CrossRef]
  61. Renaud, S.J.; Kubota, K.; Rumi, M.A.K.; Soares, M.J. The FOS transcription factor family differentially controls trophoblast migration and invasion. J. Biol. Chem. 2014, 289, 5025–5039. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, Y.; Damsky, C.H.; Chiu, K.; Roberts, J.M.; Fisher, S.J. Preeclampsia is associated with abnormal expression of adhesion molecules by invasive cytotrophoblasts. J. Clin. Investig. 1993, 91, 950–960. [Google Scholar] [CrossRef] [PubMed]
  63. Lu, X.; Wang, R.; Zhu, C.; Wang, H.; Lin, H.-Y.; Gu, Y.; Cross, J.C.; Wang, H. Fine-Tuned and Cell-Cycle-Restricted Expression of Fusogenic Protein Syncytin-2 Maintains Functional Placental Syncytia. Cell Rep. 2018, 23, 3979. [Google Scholar] [CrossRef] [PubMed]
  64. Kreis, N.-N.; Friemel, A.; Jennewein, L.; Hoock, S.C.; Hentrich, A.E.; Nowak, T.; Louwen, F.; Yuan, J. Functional Analysis of p21Cip1/CDKN1A and Its Family Members in Trophoblastic Cells of the Placenta and Its Roles in Preeclampsia. Cells 2021, 10, 2214. [Google Scholar] [CrossRef]
  65. Xiao, Z.; Yan, L.; Liang, X.; Wang, H. Progress in deciphering trophoblast cell differentiation during human placentation. Curr. Opin. Cell Biol. 2020, 67, 86–91. [Google Scholar] [CrossRef]
  66. DeLoia, J.A.; Burlingame, J.M.; Krasnow, J.S. Differential expression of g1, cyclins during human placentogenesis. Placenta 1997, 18, 9–16. [Google Scholar] [CrossRef] [PubMed]
  67. Ma, J.; Hu, H.; Lin, M.; Chen, L.; Liu, M.; Li, H.; Quan, S. ELABELA alleviates syncytiotrophoblast hypoxia/reoxygenation injury and preeclampsia-like symptoms in mice by reducing apoptosis. Placenta 2021, 106, 30–39. [Google Scholar] [CrossRef]
  68. Dawid, M.; Mlyczynska, E.; Kurowska, P.; Sierpowski, M.; Rak, A. Apelin decreased placental hormone secretion by human trophoblast BeWo cells via apelin receptor, protein kinase A and extracellular signal-regulated kinases 1/2 activation. J. Physiol. Pharmacol. 2019, 70, 895–907. [Google Scholar] [CrossRef]
  69. Mlyczyńska, E.; Myszka, M.; Kurowska, P.; Dawid, M.; Milewicz, T.; Bałajewicz-Nowak, M.; Kowalczyk, P.; Rak, A. Anti-Apoptotic Effect of Apelin in Human Placenta: Studies on BeWo Cells and Villous Explants from Third-Trimester Human Pregnancy. Int. J. Mol. Sci. 2021, 22, 2760. [Google Scholar] [CrossRef]
  70. Yang, H.; Zhang, X.; Ding, Y.; Xiong, H.; Xiang, S.; Wang, Y.; Li, H.; Liu, Z.; He, J.; Tao, Y.; et al. Elabela: Negative Regulation of Ferroptosis in Trophoblasts via the Ferritinophagy Pathway Implicated in the Pathogenesis of Preeclampsia. Cells 2023, 12, 99. [Google Scholar] [CrossRef]
  71. Zhou, L.; Sun, H.; Cheng, R.; Fan, X.; Lai, S.; Deng, C. ELABELA, as a potential diagnostic biomarker of preeclampsia, regulates abnormally shallow placentation via APJ. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E773–E781. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, L.; Zhang, Y.; Qu, H.; Xu, F.; Hu, H.; Zhang, Q.; Ye, Y. Reduced ELABELA expression attenuates trophoblast invasion through the PI3K/AKT/mTOR pathway in early onset preeclampsia. Placenta 2019, 87, 38–45. [Google Scholar] [CrossRef] [PubMed]
  73. Dawid, M.; Mlyczyńska, E.; Jurek, M.; Respekta, N.; Pich, K.; Kurowska, P.; Gieras, W.; Milewicz, T.; Kotula-Balak, M.; Rak, A. Apelin, APJ, and ELABELA: Role in Placental Function, Pregnancy, and Foetal Development—An Overview. Cells 2021, 11, 99. [Google Scholar] [CrossRef]
  74. Guo, Y.-Y.; Li, T.; Liu, H.; Tang, L.; Li, Y.-C.; Hu, H.-T.; Su, Y.-F.; Lin, Y.; Wang, Y.-Y.; Li, C.; et al. Circulating levels of Elabela and Apelin in the second and third trimesters of pregnancies with gestational diabetes mellitus. Gynecol. Endocrinol. 2020, 36, 890–894. [Google Scholar] [CrossRef]
  75. Hehir, M.P.; Morrison, J.J. The adipokine apelin and human uterine contractility. Am. J. Obstet. Gynecol. 2012, 206, 359.e1–359.e5. [Google Scholar] [CrossRef]
  76. Kacar, E.; Ercan, Z.; Serhatlioglu, I.; Sumer, A.; Kelestimur, H.; Kutlu, S. The effects of apelin on myometrium contractions in pregnant rats. Cell. Mol. Biol. Noisy-Gd. Fr. 2018, 64, 74–79. [Google Scholar] [CrossRef]
  77. Carvajal, J.A.; Oporto, J.I. The Myometrium in Pregnant Women with Obesity. Curr. Vasc. Pharmacol. 2021, 19, 193–200. [Google Scholar] [CrossRef] [PubMed]
  78. Habata, Y.; Fujii, R.; Hosoya, M.; Fukusumi, S.; Kawamata, Y.; Hinuma, S.; Kitada, C.; Nishizawa, N.; Murosaki, S.; Kurokawa, T.; et al. Apelin, the natural ligand of the orphan receptor APJ, is abundantly secreted in the colostrum. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 1999, 1452, 25–35. [Google Scholar] [CrossRef]
  79. Aydin, S. The presence of the peptides apelin, ghrelin and nesfatin-1 in the human breast milk, and the lowering of their levels in patients with gestational diabetes mellitus. Peptides 2010, 31, 2236–2240. [Google Scholar] [CrossRef]
  80. Marousez, L.; Hanssens, S.; Butruille, L.; Petit, C.; Pourpe, C.; Besengez, C.; Rakza, T.; Storme, L.; Deruelle, P.; Lesage, J.; et al. Breast milk apelin level increases with maternal obesity and high-fat feeding during lactation. Int. J. Obes. 2005 2021, 45, 1052–1060. [Google Scholar] [CrossRef]
  81. Khan, K.S.; Wojdyla, D.; Say, L.; Gülmezoglu, A.M.; Van Look, P.F. WHO analysis of causes of maternal death: A systematic review. Lancet 2006, 367, 1066–1074. [Google Scholar] [CrossRef] [PubMed]
  82. Say, L.; Chou, D.; Gemmill, A.; Tunçalp, Ö.; Moller, A.-B.; Daniels, J.; Gülmezoglu, A.M.; Temmerman, M.; Alkema, L. Global causes of maternal death: A WHO systematic analysis. Lancet Glob. Health 2014, 2, e323–e333. [Google Scholar] [CrossRef] [PubMed]
  83. Binder, J.; Kalafat, E.; Palmrich, P.; Pateisky, P.; Khalil, A. Should angiogenic markers be included in diagnostic criteria of superimposed pre-eclampsia in women with chronic hypertension? Ultrasound Obstet. Gynecol. 2022, 59, 192–201. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, Y.; Wang, L.; Shi, H. The biological function of ELABELA and APJ signaling in the cardiovascular system and pre-eclampsia. Hypertens. Res. 2019, 42, 928–934. [Google Scholar] [CrossRef] [PubMed]
  85. Yallampalli, C.; Garfield, R.E. Inhibition of nitric oxide synthesis in rats during pregnancy produces signs similar to those of preeclampsia. Am. J. Obstet. Gynecol. 1993, 169, 1316–1320. [Google Scholar] [CrossRef]
  86. Khalil, R.A.; Crews, J.K.; Novak, J.; Kassab, S.; Granger, J.P. Enhanced vascular reactivity during inhibition of nitric oxide synthesis in pregnant rats. Hypertension 1979 1998, 31, 1065–1069. [Google Scholar] [CrossRef]
  87. Osol, G.; Barron, C.; Gokina, N.; Mandala, M. Inhibition of nitric oxide synthases abrogates pregnancy-induced uterine vascular expansive remodeling. J. Vasc. Res. 2009, 46, 478–486. [Google Scholar] [CrossRef]
  88. Zuo, J.; Jiang, Z. Melatonin attenuates hypertension and oxidative stress in a rat model of L-NAME-induced gestational hypertension. Vasc. Med. 2020, 25, 295–301. [Google Scholar] [CrossRef]
  89. Motta, C.; Grosso, C.; Zanuzzi, C.; Molinero, D.; Picco, N.; Bellingeri, R.; Alustiza, F.; Barbeito, C.; Vivas, A.; Romanini, M. Effect of Sildenafil on Pre-Eclampsia-Like Mouse Model Induced By L-Name. Reprod. Domest. Anim. 2015, 50, 611–616. [Google Scholar] [CrossRef]
  90. de Souza, C.O.; Peraçoli, M.T.S.; Weel, I.C.; Bannwart, C.F.; Romão, M.; Nakaira-Takahagi, E.; de Medeiros, L.T.L.; Silva, M.G.d.; Peraçoli, J.C. Hepatoprotective and anti-inflammatory effects of silibinin on experimental preeclampsia induced by L-NAME in rats. Life Sci. 2012, 91, 159–165. [Google Scholar] [CrossRef]
  91. Pritchard, N.; Kaitu’u-Lino, T.J.; Gong, S.; Dopierala, J.; Smith, G.C.S.; Charnock-Jones, D.S.; Tong, S. ELABELA/APELA levels are not decreased in the maternal circulation or placenta among women with preeclampsia. Am. J. Pathol. 2018, 188, 1749–1753. [Google Scholar] [CrossRef] [PubMed]
  92. Panaitescu, B.; Romero, R.; Gomez-Lopez, N.; Pacora, P.; Erez, O.; Vadillo-Ortega, F.; Yeo, L.; Hassan, S.S.; Hsu, C.-D. ELABELA Plasma Concentrations are Increased in Women with Late-Onset Preeclampsia. J. Matern.-Fetal Neonatal Med. 2020, 33, 5–15. [Google Scholar] [CrossRef] [PubMed]
  93. Para, R.; Romero, R.; Gomez-Lopez, N.; Tarca, A.L.; Panaitescu, B.; Done, B.; Hsu, R.; Pacora, P.; Hsu, C.-D. Maternal circulating concentrations of soluble Fas and Elabela in early- and late-onset preeclampsia. J. Matern.-Fetal Neonatal Med. 2022, 35, 316–329. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, X.; Bian, Y.; Wan, J.; Li, L.; Yang, P.; Zhao, S.; Zhao, H. Variants in the 5’-UTR of APELA gene in women with preeclampsia. Prenat. Diagn. 2019, 39, 308–313. [Google Scholar] [CrossRef] [PubMed]
  95. Villie, P.; Lefevre, G.; Arrestier, R.; Rousseau, A.; Berkane, N.; Hertig, A. ELABELA concentration is not decreased in maternal plasma before the onset of preeclampsia. Am. J. Obstet. Gynecol. 2019, 220, 284–285. [Google Scholar] [CrossRef]
  96. Eberlé, D.; Marousez, L.; Hanssens, S.; Knauf, C.; Breton, C.; Deruelle, P.; Lesage, J. Elabela and Apelin actions in healthy and pathological pregnancies. Cytokine Growth Factor Rev. 2019, 46, 45–53. [Google Scholar] [CrossRef]
  97. Hamza, R.Z.; Diab, A.A.A.; Zahra, M.H.; Asalah, A.K.; Moursi, S.M.M.; Al-Baqami, N.M.; Al-Salmi, F.A.; Attia, M.S. Correlation between Apelin and Some Angiogenic Factors in the Pathogenesis of Preeclampsia: Apelin-13 as Novel Drug for Treating Preeclampsia and Its Physiological Effects on Placenta. Int. J. Endocrinol. 2021, 2021, 5017362. [Google Scholar] [CrossRef]
  98. Simsek, Y.; Celik, O.; Yilmaz, E.; Karaer, A.; Dogan, C.; Aydin, S.; Ozer, A. Serum levels of apelin, salusin-alpha and salusin-beta in normal pregnancy and preeclampsia. J. Matern.-Fetal Neonatal Med. 2012, 25, 1705–1708. [Google Scholar] [CrossRef]
  99. Kucur, M.; Tuten, A.; Oncul, M.; Acikgoz, A.S.; Yuksel, M.A.; Imamoglu, M.; Balci Ekmekci, O.; Yilmaz, N.; Madazli, R. Maternal serum apelin and YKL-40 levels in early and late-onset pre-eclampsia. Hypertens. Pregnancy 2014, 33, 467–475. [Google Scholar] [CrossRef]
  100. Deniz, R.; Baykus, Y.; Ustebay, S.; Ugur, K.; Yavuzkir, Ş.; Aydin, S. Evaluation of elabela, apelin and nitric oxide findings in maternal blood of normal pregnant women, pregnant women with pre-eclampsia, severe pre-eclampsia and umbilical arteries and venules of newborns. J. Obstet. Gynaecol. 2019, 39, 907–912. [Google Scholar] [CrossRef]
  101. Temur, M.; Yilmaz, Ö.; Taşgöz, F.N.; Kume, T. The evaluation of serum apelin levels in patients complicated with preeclampsia. J. Matern.-Fetal Neonatal Med. 2022, 35, 1848–1852. [Google Scholar] [CrossRef]
  102. Sattar Taha, A.; Zahraei, Z.; Al-Hakeim, H.K. Serum apelin and galectin-3 in preeclampsia in Iraq. Hypertens. Pregnancy 2020, 39, 379–386. [Google Scholar] [CrossRef]
  103. Bortoff, K.D.; Qiu, C.; Runyon, S.; Williams, M.A.; Maitra, R. Decreased Maternal Plasma Apelin Concentrations in Preeclampsia. Hypertens. Pregnancy 2012, 31, 398–404. [Google Scholar] [CrossRef]
  104. Gandham, R.; Dayanand, C.D.; Sheela, S.R.; Kiranmayee, P. Maternal serum Apelin 13 and APLN gene promoter variant -1860T > C in preeclampsia. J. Matern.-Fetal Neonatal Med. 2022, 35, 5008–5016. [Google Scholar] [CrossRef] [PubMed]
  105. Gürlek, B.; Yılmaz, A.; Durakoğlugil, M.E.; Karakaş, S.; Kazaz, İ.M.; Önal, Ö.; Şatıroğlu, Ö. Evaluation of serum apelin-13 and apelin-36 concentrations in preeclamptic pregnancies. J. Obstet. Gynaecol. Res. 2020, 46, 58–65. [Google Scholar] [CrossRef] [PubMed]
  106. Fuentes-Carrasco, M.; Ruíz-Román, R.; Savirón-Cornudella, R.; Pérez-Roncero, G.; López-Baena, M.T.; Pérez-López, F.R. Systematic review and meta-analysis regarding maternal apelin in pregnant women with and without preeclampsia. Gynecol. Endocrinol. 2022, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  107. Burton, G.J.; Jauniaux, E. Pathophysiology of placental-derived fetal growth restriction. Am. J. Obstet. Gynecol. 2018, 218, S745–S761. [Google Scholar] [CrossRef] [PubMed]
  108. Resnik, R. Intrauterine growth restriction. Obstet. Gynecol. 2002, 99, 490–496. [Google Scholar] [CrossRef] [PubMed]
  109. Gordijn, S.J.; Beune, I.M.; Ganzevoort, W. Building consensus and standards in fetal growth restriction studies. Best Pract. Res. Clin. Obs. Gynaecol. 2018, 49, 117–126. [Google Scholar] [CrossRef]
  110. Malamitsi-Puchner, A.; Gourgiotis, D.; Boutsikou, M.; Baka, S.; Hassiakos, D.; Briana, D.D. Circulating apelin concentrations in mother/infant pairs at term. Acta Paediatr. 2007, 96, 1751–1754. [Google Scholar] [CrossRef] [PubMed]
  111. Van Mieghem, T.; Doherty, A.; Baczyk, D.; Drewlo, S.; Baud, D.; Carvalho, J.; Kingdom, J. Apelin in Normal Pregnancy and Pregnancies Complicated by Placental Insufficiency. Reprod. Sci. 2016, 23, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  112. Tang, S.-Y.; Xie, H.; Yuan, L.-Q.; Luo, X.-H.; Huang, J.; Cui, R.-R.; Zhou, H.-D.; Wu, X.-P.; Liao, E.-Y. Apelin stimulates proliferation and suppresses apoptosis of mouse osteoblastic cell line MC3T3-E1 via JNK and PI3-K/Akt signaling pathways. Peptides 2007, 28, 708–718. [Google Scholar] [CrossRef] [PubMed]
  113. Behram, M.; Oğlak, S.C.; Dağ, İ. Circulating levels of Elabela in pregnant women complicated with intrauterine growth restriction. J. Gynecol. Obstet. Hum. Reprod. 2021, 50, 102127. [Google Scholar] [CrossRef] [PubMed]
  114. Yener, G.; Kavak, S.B.; Gul, Y.; Celik Kavak, E.; Gulcu Bulmus, F.; Sanli, C.; Batmaz, I.; Bulu, G. Elabela levels in pregnancies with intrauterine growth retardation. Ginekol. Pol. 2022, 94, 113–118. [Google Scholar] [CrossRef] [PubMed]
  115. Aslan, M.; Celik, O.; Celik, N.; Turkcuoglu, I.; Yilmaz, E.; Karaer, A.; Simsek, Y.; Celik, E.; Aydin, S. Cord blood nesfatin-1 and apelin-36 levels in gestational diabetes mellitus. Endocrine 2012, 41, 424–429. [Google Scholar] [CrossRef]
  116. Akinci, B.; Celtik, A.; Tunali, S.; Genc, S.; Yuksel, F.; Secil, M.; Ozcan, M.A.; Bayraktar, F. Circulating apelin levels are associated with cardiometabolic risk factors in women with previous gestational diabetes. Arch. Gynecol. Obstet. 2014, 289, 787–793. [Google Scholar] [CrossRef] [PubMed]
  117. Boyadzhieva, M.; Atanasova, I.; Zacharieva, S.; Kedikova, S. Adipocytokines during pregnancy and postpartum in women with gestational diabetes and healthy controls. J. Endocrinol. Invest. 2013, 36, 944–949. [Google Scholar] [CrossRef]
  118. Mierzyński, R.; Poniedziałek-Czajkowska, E.; Dłuski, D.; Kamiński, M.; Mierzyńska, A.; Leszczyńska-Gorzelak, B. The Potential Role of Chemerin, Lipocalin 2, and Apelin in the Diagnosis and Pathophysiology of Gestational Diabetes Mellitus. J. Diabetes Res. 2021, 2021, 5547228. [Google Scholar] [CrossRef] [PubMed]
  119. Oncul, M.; Tuten, A.; Erman, H.; Gelisgen, R.; Benian, A.; Uzun, H. Maternal and cord blood apelin, resistin and visfatin levels in gestational diabetes mellitus. Minerva Med. 2013, 104, 527–535. [Google Scholar]
  120. Telejko, B.; Kuzmicki, M.; Zonenberg, A.; Modzelewska, A.; Niedziolko-Bagniuk, K.; Ponurkiewicz, A.; Wawrusiewicz-Kurylonek, N.; Nikolajuk, A.; Szamatowicz, J.; Laudanski, P.; et al. Ghrelin in gestational diabetes: Serum level and mRNA expression in fat and placental tissue. Exp. Clin. Endocrinol. Diabetes 2010, 118, 87–92. [Google Scholar] [CrossRef]
  121. Sun, J.; Ren, J.; Zuo, C.; Deng, D.; Pan, F.; Chen, R.; Zhu, J.; Chen, C.; Ye, S. Circulating apelin, chemerin and omentin levels in patients with gestational diabetes mellitus: A systematic review and meta-analysis. Lipids Health Dis. 2020, 19, 26. [Google Scholar] [CrossRef]
  122. Dasgupta, S.; Banerjee, U.; Mukhopadhyay, P.; Maity, P.; Saha, S.; Das, B. Clinicopathological study and immunohistochemical analysis of expression of annexin A5 and apelin in human placentae of gestational diabetes mellitus. Diabetes Metab. Syndr. 2022, 16, 102435. [Google Scholar] [CrossRef] [PubMed]
  123. Zheng, X.-D.; Huang, Y.; Li, H. Regulatory role of Apelin-13-mediated PI3K/AKT signaling pathway in the glucose and lipid metabolism of mouse with gestational diabetes mellitus. Immunobiology 2021, 226, 152135. [Google Scholar] [CrossRef] [PubMed]
  124. Romero, R.; Kusanovic, J.P.; Chaiworapongsa, T.; Hassan, S.S. Placental bed disorders in preterm labor, preterm PROM, spontaneous abortion and abruptio placentae. Best Pract. Res. Clin. Obstet. Gynaecol. 2011, 25, 313–327. [Google Scholar] [CrossRef] [PubMed]
  125. Manolea, M.M.; Dijmărescu, A.L.; Popescu, F.C.; Novac, M.B.; DiŢescu, D. Evaluation of the implantation site morphology in spontaneous abortion. Romanian J. Morphol. Embryol. Rev. Roum. Morphol. Embryol. 2015, 56, 125–131. [Google Scholar]
  126. Tug, E.; Yirmibes Karaoguz, M.; Nas, T. Expression of the syncytin-1 and syncytin-2 genes in the trophoblastic tissue of the early pregnancy losses with normal and abnormal karyotypes. Gene 2020, 741, 144533. [Google Scholar] [CrossRef]
  127. Qi, Y.; Hou, Y.; Ma, M.; Li, X.; Wu, J. Circulating levels of Elabela in pregnant women with missed abortion. Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol. 2022, 38, 693–696. [Google Scholar] [CrossRef]
  128. Georgiadou, D.; Boussata, S.; van Dijk, M. ELABELA measurements by commercial ELISA kits require sample extraction. Am. J. Physiol.-Endocrinol. Metab. 2019, 317, E1218–E1219. [Google Scholar] [CrossRef]
  129. Marsault, E.; Llorens-Cortes, C.; Iturrioz, X.; Chun, H.J.; Lesur, O.; Oudit, G.Y.; Auger-Messier, M. The apelinergic system: A perspective on challenges and opportunities in cardiovascular and metabolic disorders. Ann. N. Y. Acad. Sci. 2019, 1455, 12–33. [Google Scholar] [CrossRef]
  130. Zhen, E.Y.; Higgs, R.E.; Gutierrez, J.A. Pyroglutamyl apelin-13 identified as the major apelin isoform in human plasma. Anal. Biochem. 2013, 442, 1–9. [Google Scholar] [CrossRef]
  131. De Mota, N.; Reaux-Le Goazigo, A.; El Messari, S.; Chartrel, N.; Roesch, D.; Dujardin, C.; Kordon, C.; Vaudry, H.; Moos, F.; Llorens-Cortes, C. Apelin, a potent diuretic neuropeptide counteracting vasopressin actions through inhibition of vasopressin neuron activity and vasopressin release. Proc. Natl. Acad. Sci. USA 2004, 101, 10464–10469. [Google Scholar] [CrossRef] [PubMed]
  132. Reaux, A.; De Mota, N.; Skultetyova, I.; Lenkei, Z.; El Messari, S.; Gallatz, K.; Corvol, P.; Palkovits, M.; Llorens-Cortès, C. Physiological role of a novel neuropeptide, apelin, and its receptor in the rat brain. J. Neurochem. 2001, 77, 1085–1096. [Google Scholar] [CrossRef] [PubMed]
  133. Azizi, M.; Iturrioz, X.; Blanchard, A.; Peyrard, S.; De Mota, N.; Chartrel, N.; Vaudry, H.; Corvol, P.; Llorens-Cortes, C. Reciprocal Regulation of Plasma Apelin and Vasopressin by Osmotic Stimuli. J. Am. Soc. Nephrol. 2008, 19, 1015–1024. [Google Scholar] [CrossRef]
  134. Brash, L.; Barnes, G.D.; Brewis, M.J.; Church, A.C.; Gibbs, S.J.; Howard, L.S.G.E.; Jayasekera, G.; Johnson, M.K.; McGlinchey, N.; Onorato, J.; et al. Short-Term Hemodynamic Effects of Apelin in Patients With Pulmonary Arterial Hypertension. JACC Basic Transl. Sci. 2018, 3, 176–186. [Google Scholar] [CrossRef] [PubMed]
  135. Mesmin, C.; Dubois, M.; Becher, F.; Fenaille, F.; Ezan, E. Liquid chromatography/tandem mass spectrometry assay for the absolute quantification of the expected circulating apelin peptides in human plasma. Rapid Commun. Mass Spectrom. 2010, 24, 2875–2884. [Google Scholar] [CrossRef]
  136. Cobellis, L.; De Falco, M.; Mastrogiacomo, A.; Giraldi, D.; Dattilo, D.; Scaffa, C.; Colacurci, N.; De Luca, A. Modulation of apelin and APJ receptor in normal and preeclampsia-complicated placentas. Histol. Histopathol. 2007, 22, 1–8. [Google Scholar]
  137. Hanssens, S.; Marousez, L.; Pécheux, O.; Besengez, C.; Storme, L.; Deruelle, P.; Eberlé, D.; Lesage, J. Maternal obesity reduces apelin level in cord blood without altering the placental apelin/elabela-APJ system. Placenta 2022, 128, 112–115. [Google Scholar] [CrossRef]
  138. Telejko, B.; Kuzmicki, M.; Wawrusiewicz-Kurylonek, N.; Szamatowicz, J.; Nikolajuk, A.; Zonenberg, A.; Zwierz-Gugala, D.; Jelski, W.; Laudański, P.; Wilczynski, J.; et al. Plasma apelin levels and apelin/APJ mRNA expression in patients with gestational diabetes mellitus. Diabetes Res. Clin. Pract. 2010, 87, 176–183. [Google Scholar] [CrossRef]
  139. Karagoz, Z.K.; Aydin, S.; Ugur, K.; Tigli, A.; Deniz, R.; Baykus, Y.; Sahin, I.; Yalcin, M.H.; Yavuz, A.; Aksoy, A.; et al. Molecular communication between Apelin-13, Apelin-36, Elabela, and nitric oxide in gestational diabetes mellitus. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 3289–3300. [Google Scholar]
  140. Yamaleyeva, L.M.; Chappell, M.C.; Brosnihan, K.B.; Anton, L.; Caudell, D.L.; Shi, S.; McGee, C.; Pirro, N.; Gallagher, P.E.; Taylor, R.N.; et al. Downregulation of apelin in the human placental chorionic villi from preeclamptic pregnancies. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E852–E860. [Google Scholar] [CrossRef]
  141. Li, Y.; Yang, X.; Ouyang, S.; He, J.; Yu, B.; Lin, X.; Zhang, Q.; Tao, J. Declined circulating Elabela levels in patients with essential hypertension and its association with impaired vascular function: A preliminary study. Clin. Exp. Hypertens. 2020, 42, 239–243. [Google Scholar] [CrossRef] [PubMed]
  142. Chapman, F.A.; Nyimanu, D.; Maguire, J.J.; Davenport, A.P.; Newby, D.E.; Dhaun, N. The therapeutic potential of apelin in kidney disease. Nat. Rev. Nephrol. 2021, 17, 840–853. [Google Scholar] [CrossRef] [PubMed]
  143. Ba, H.-J.; Chen, H.-S.; Su, Z.; Du, M.-L.; Chen, Q.-L.; Li, Y.-H.; Ma, H.-M. Associations between serum apelin-12 levels and obesity-related markers in Chinese children. PLoS ONE 2014, 9, e86577. [Google Scholar] [CrossRef] [PubMed]
  144. El Wakeel, M.A.; El-Kassas, G.M.; Kamhawy, A.H.; Galal, E.M.; Nassar, M.S.; Hammad, E.M.; El-Zayat, S.R. Serum Apelin and Obesity-Related Complications in Egyptian Children. Open Access Maced. J. Med. Sci. 2018, 6, 1354–1358. [Google Scholar] [CrossRef]
  145. Vinel, C.; Lukjanenko, L.; Batut, A.; Deleruyelle, S.; Pradère, J.-P.; Le Gonidec, S.; Dortignac, A.; Geoffre, N.; Pereira, O.; Karaz, S.; et al. The exerkine apelin reverses age-associated sarcopenia. Nat. Med. 2018, 24, 1360–1371. [Google Scholar] [CrossRef]
  146. Meral, C.; Tascilar, E.; Karademir, F.; Tanju, I.A.; Cekmez, F.; Ipcioglu, O.M.; Ercin, C.N.; Gocmen, I.; Dogru, T. Elevated plasma levels of apelin in children with type 1 diabetes mellitus. J. Pediatr. Endocrinol. Metab. 2010, 23, 497–502. [Google Scholar] [CrossRef]
  147. Castan-Laurell, I.; El Boustany, R.; Pereira, O.; Potier, L.; Marre, M.; Fumeron, F.; Valet, P.; Gourdy, P.; Velho, G.; Roussel, R. Plasma Apelin and Risk of Type 2 Diabetes in a Cohort From the Community. Diabetes Care 2019, 43, e15–e16. [Google Scholar] [CrossRef]
  148. Capo, A.; Di Nicola, M.; Costantini, E.; Reale, M.; Amerio, P. Circulating levels of Apelin-36 in patients with mild to moderate psoriasis. G. Ital. Di Dermatol. E Venereol. Organo Uff. Soc. Ital. Di Dermatol. E Sifilogr. 2020, 155, 646–651. [Google Scholar] [CrossRef]
  149. Zorlu, M.; Kiskac, M.; Karatoprak, C.; Kesgin, S.; Cakirca, M.; Yildiz, K.; Ardic, C.; Cikrikcioglu, M.A.; Erkoc, R. Assessment of serum apelin and lipocalin-2 levels in patients with subclinical hypothyroidism. Ann. Endocrinol. 2014, 75, 10–14. [Google Scholar] [CrossRef]
  150. Gürel, A.; Doğantekin, A.; Özkan, Y.; Aydın, S. Serum apelin levels in patients with thyroid dysfunction. Int. J. Clin. Exp. Med. 2015, 8, 16394–16398. [Google Scholar]
  151. Kourtis, A.; Gkiomisi, A.; Mouzaki, M.; Makedou, K.; Anastasilakis, A.D.; Toulis, K.A.; Gerou, S.; Gavana, E.; Agorastos, T. Apelin levels in normal pregnancy. Clin. Endocrinol. 2011, 75, 367–371. [Google Scholar] [CrossRef] [PubMed]
  152. Van Mieghem, T.; van Bree, R.; Van Herck, E.; Pijnenborg, R.; Deprest, J.; Verhaeghe, J. Maternal apelin physiology during rat pregnancy: The role of the placenta. Placenta 2010, 31, 725–730. [Google Scholar] [CrossRef] [PubMed]
  153. Hashemi, M.; Rezaei, H.; Eskandari-Nasab, E.; Kaykhaei, M.A.; Taheri, M. Association between the apelin rs2235306 gene polymorphism and metabolic syndrome. Turk. J. Med. Sci. 2014, 44, 775–780. [Google Scholar] [CrossRef]
  154. Wu, X.-D.; Zhang, N.; Liang, M.; Liu, W.-L.; Lin, B.-B.; Xiao, Y.-R.; Li, Y.-Z.; Zeng, K.; Lin, C.-Z. Gender-specific association between Apelin/APJ gene polymorphisms and hypertension risk in Southeast China. Gene 2018, 669, 63–68. [Google Scholar] [CrossRef]
  155. Laudermilk, L.T.; Harper, K.M.; Moy, S.S.; Runyon, S.; Zhou, B.; Koller, B.; Maitra, R. Aplnr knockout mice display sex-specific changes in conditioned fear. Behav. Brain Res. 2021, 400, 113059. [Google Scholar] [CrossRef] [PubMed]
  156. Ma, Z.; Zhao, L.; Zhang, Y.-P.; Zhong, J.-C.; Yang, X.-C. Declined ELABELA plasma levels in hypertension patients with atrial fibrillation: A case control study. BMC Cardiovasc. Disord. 2021, 21, 390. [Google Scholar] [CrossRef] [PubMed]
  157. Chae, S.A.; Son, J.S.; Du, M. Prenatal exercise in fetal development: A placental perspective. FEBS J. 2022, 289, 3058–3071. [Google Scholar] [CrossRef]
  158. Ma, Z.; Zhao, L.; Martin, S.; Zhang, Y.; Dong, Y.; Zhong, J.-C.; Yang, X.-C. Lower Plasma Elabela Levels in Hypertensive Patients With Heart Failure Predict the Occurrence of Major Adverse Cardiac Events: A Preliminary Study. Front. Cardiovasc. Med. 2021, 8, 638468. [Google Scholar] [CrossRef]
Figure 1. Apelinergic system expression and roles in placenta. ELA: Elabela; EVT: extravillous trophoblast.
Figure 1. Apelinergic system expression and roles in placenta. ELA: Elabela; EVT: extravillous trophoblast.
Ijms 24 08014 g001
Figure 2. Activation of different signaling pathways through ELA (framed in green), apelin (framed in green), or both (framed in cyan) in the binding of APJ in human trophoblast [59,68,69]. ELA: Elabela; APJ: apelin peptide jejunum; AMPK: adenosine monophosphate-activated protein kinase; ERK1/2: extracellular signal-activated kinase 1/2; PKA: protein kinase A; PI3K: phosphatidylinositol 3-kinases.
Figure 2. Activation of different signaling pathways through ELA (framed in green), apelin (framed in green), or both (framed in cyan) in the binding of APJ in human trophoblast [59,68,69]. ELA: Elabela; APJ: apelin peptide jejunum; AMPK: adenosine monophosphate-activated protein kinase; ERK1/2: extracellular signal-activated kinase 1/2; PKA: protein kinase A; PI3K: phosphatidylinositol 3-kinases.
Ijms 24 08014 g002
Table 1. Altered APJ, apelin, and ELA levels in human pregnancy disorders compared to a physiological pregnancy during pregnancy and in the postpartum period.
Table 1. Altered APJ, apelin, and ELA levels in human pregnancy disorders compared to a physiological pregnancy during pregnancy and in the postpartum period.
VariableTissue/FluidExpressionMethodsPathologiesReference
Pregnancy
Molecule: APLNR
deliveryplacentamRNA, proteinReal-time PCR, IHCLate onset PE (↘)[71]
deliveryplacentaproteinIHCPE (≈)[58]
deliveryplacentaproteinIHCPE (↗)[136]
deliveryplacentamRNART-qPCRMaternal obesity (≈)[137]
deliveryplacenta, adipose tissuemRNAReal-time PCRGDM (≈)[138]
Molecule: APLN
deliveryserum, cord bloodproteinELISAGDM (≈in cord blood, ↗ in serum)[115]
24–28 WGserumproteinELISA, EIAGDM (↘)[117]
2nd and 3rd trimesterserumproteinELISAGDM (↗only in the 2nd trimester )[74]
deliveryserumproteinELISAGDM (≈)[118]
deliverymaternal and cord bloodproteinELISAGDM (↘ in cord blood, ≈ in maternal blood )[119]
24–32 WG and delivery plasma, adipose tissue, placentaproteinELISA, Real-time PCRGDM (≈mRNA and circulating level)[138]
deliveryplacentaproteinIHCGDM (↘)[122]
2nd trimesterserumproteinELISAGDM (↗)[139]
20–34 WG and deliveryserum, placentaprotein, mRNAELISA, IHC, RT-PCRPreterm ≈, IUGR and PE (↘ prot, ≈mRNA)[111]
deliveryplacentaproteinRIAPE (↘)[140]
deliveryplasmaproteinELISAPE (↘)[103]
time of diagnosisserumproteinEIAPE (↗)[98]
time of diagnosisserumproteinEIAPE (↘)[101]
deliverymaternal and cord bloodproteinELISAPE (↘ in both maternal and cord blood)[100]
deliveryplacentaproteinIHCPE (↘)[122]
deliveryserumproteinELISAPE (↘)[104]
deliveryplacenta, serumprotein, mRNAELISA, IHC, WB, RT-qPCRPE (↘ in placenta, ↗ in maternal circulation)[58]
deliveryserumproteinELISAPE (↘) [105]
deliveryplacentaproteinIHCPE (↗)[136]
time of diagnosisserumproteinELISAPE (↗)[99]
deliveryplasma, cord blood, placentaprotein, mRNAELISA, RT-qPCRMaternal obesity (≈mRNA and plasma, ↘ in cord blood)[137]
Molecule: ELA
time of diagnosisserumproteinELISAIUGR (↘) [113]
deliveryserumproteinELISAIUGR (↗) [114]
deliverymaternal and cord bloodproteinELISAPE (↘ in both maternal and cord blood)[100]
deliveryplasmaproteinELISAEarly-onset PE (≈), late-onset PE (↗)[92]
deliveryserum, urine, placentaprotein, mRNAELISA, IHC, Real-time PCRLate-onset PE (circulating and placental level ↘)[71]
delivery plasma, placentaprotein, mRNAELISA, RNA sequencingPE (≈ placental mRNA and circulating protein)[91]
1st trimester serumproteinELISAGH/PE (≈)[67]
2nd trimesterserumproteinELISAGDM (↗)[139]
2nd and 3rd trimesterserumproteinELISAGDM (↘ during second trimester)[74]
deliveryplasma, cord blood, placentaprotein, mRNAELISA, RT-qPCRMaternal obesity (≈mRNA and protein)[137]
time of diagnosisserumproteinELISA MA (↘) [127]
Post-partum
Molecule: APLN
serum, colostrum and mature milkproteinELISAGDM (↘ in colostrum and milk)[79]
plasmaproteinELISAGDM (↘) [116]
plasma, breast milkproteinELISAObesity (↗with BMI)[80]
PCR: polymerase chain reaction; IHC: immunohistochemistry; PE: preeclampsia; GDM: gestational diabetes mellitus; IUGR: intrauterine growth restriction; EIA: enzyme immunoassay; ELISA: enzyme-linked immunosorbent assay; MA: missed abortion.
Table 2. Effect of administration of apelin and ELA peptides during pregnancy pathologies [39,97,140,157,158].
Table 2. Effect of administration of apelin and ELA peptides during pregnancy pathologies [39,97,140,157,158].
ModelPeptideOutputReference
Obesogenic diet miceApelinImprovement in placental function of obese dams[157]
L-NAME-induced PE in ratsApelin-13Reno-protective effects[97]
PE rat model (TGA-PE)Pyr-
Apelin-13
Improvement in hemodynamic response and renal injury
without fetal toxicity
[140]
PE rat model by reduced uterine perfusion
pressure
ApelinAmelioration of PE symptoms[39]
L-NAME-induced PE in ratsELAReversion of the phenotypes of L-NAME-induced PE[158]
PE: preeclampsia; L-NAME: L-nitro-arginine methyl ester; ELA: Elabela. Many technical challenges need to be overcome before these can be used in clinical settings.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pécheux, O.; Correia-Branco, A.; Cohen, M.; Martinez de Tejada, B. The Apelinergic System in Pregnancy. Int. J. Mol. Sci. 2023, 24, 8014. https://doi.org/10.3390/ijms24098014

AMA Style

Pécheux O, Correia-Branco A, Cohen M, Martinez de Tejada B. The Apelinergic System in Pregnancy. International Journal of Molecular Sciences. 2023; 24(9):8014. https://doi.org/10.3390/ijms24098014

Chicago/Turabian Style

Pécheux, Océane, Ana Correia-Branco, Marie Cohen, and Begoῆa Martinez de Tejada. 2023. "The Apelinergic System in Pregnancy" International Journal of Molecular Sciences 24, no. 9: 8014. https://doi.org/10.3390/ijms24098014

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop