Next Article in Journal
Differential Physio-Biochemical and Metabolic Responses of Peanut (Arachis hypogaea L.) under Multiple Abiotic Stress Conditions
Next Article in Special Issue
Effect of Placenta-Derived Mesenchymal Stromal Cells Conditioned Media on an LPS-Induced Mouse Model of Preeclampsia
Previous Article in Journal
Label-Free Study of the Global Cell Behavior during Exposure to Environmental Radiofrequency Fields in the Presence or Absence of Pro-Apoptotic or Pro-Autophagic Treatments
Previous Article in Special Issue
Protease Inhibitor Anti-HIV, Lopinavir, Impairs Placental Endocrine Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Placental Dysfunction in Assisted Reproductive Pregnancies: Perinatal, Neonatal and Adult Life Outcomes

by
Claudio Manna
1,2,
Valentina Lacconi
1,3,
Giuseppe Rizzo
1,4,
Antonino De Lorenzo
1 and
Micol Massimiani
1,3,*
1
Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
2
Biofertility Center of Reproductive Medicine, Viale degli Eroi di Rodi 214, 00128 Rome, Italy
3
Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
4
Department of Obstetrics & Gynecology Moscow Russian Federation, The First I.M. Sechenov Moscow State Medical University, Trubetskaya Street, House 8, Building 2, 119991 Moscow, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(2), 659; https://doi.org/10.3390/ijms23020659
Submission received: 23 November 2021 / Revised: 4 January 2022 / Accepted: 5 January 2022 / Published: 8 January 2022
(This article belongs to the Special Issue Embryo Implantation and Placental Development)

Abstract

:
Obstetric and newborn outcomes of assisted reproductive technology (ART) pregnancies are associated with significative prevalence of maternal and neonatal adverse health conditions, such as cardiovascular and metabolic diseases. These data are interpreted as anomalies in placentation involving a dysregulation of several molecular factors and pathways. It is not clear which extent of the observed placental alterations are the result of ART and which originate from infertility itself. These two aspects probably act synergically for the final obstetric risk. Data show that mechanisms of inappropriate trophoblast invasion and consequent altered vascular remodeling sustain several clinical conditions, leading to obstetric and perinatal risks often found in ART pregnancies, such as preeclampsia, fetal growth restriction and placenta previa or accreta. The roles of factors such as VEGF, GATA3, PIGF, sFLT-1, sEndoglin, EGFL7, melatonin and of ART conditions, such as short or long embryo cultures, trophectoderm biopsy, embryo cryopreservation, and supraphysiologic endometrium preparation, are discussed. Inflammatory local conditions and epigenetic influence on embryos of ART procedures are important research topics since they may have important consequences on obstetric risk. Prevention and treatment of these conditions represent new frontiers for clinicians and biologists involved in ART, and synergic actions with researchers at molecular levels are advocated.

1. Introduction

There is a rapidly increasing interest in placentation in obstetric adverse outcome, neonatal and adult life health after infertility, and its related therapies. Abnormal placentation is a common finding in the infertile population, even among those couples conceiving spontaneously after a period of infertility. A higher risk of preterm birth (PTB) and low birth weight (LBW) has been found in these pregnancies [1]. Moreover, abnormal placentation and obstetric complications such as PTB, preeclampsia (PE), and fetal growth restriction (FGR) have been associated with endometriosis, a common factor of infertility [2]. Another maternal condition at risk for the development of placental anomalies, commonly associated with infertility, is polycystic ovary syndrome (PCOS), in which the prevalence of gestational diabetes mellitus (GDM) is significantly increased [3]. Of note, GDM is an independent risk factor for the onset of placental disorders with altered structure, function and hypertrophic growth of the organ [4].
An important actual focus in reproductive medicine, neonatology and population health is that up to 6% (range between 0.2% and 6.4%) of European births is conceived by assisted reproductive technology (ART) [5] and concerns existing health conditions of individuals born following ART. ART is a group of in vitro techniques used to treat moderate and severe infertility, including in vitro fertilization (IVF), intracytoplasmic sperm injection (ICSI), frozen embryo transfer (FET), oocyte donation (OD), blastocyst culture, intrauterine insemination, and preimplantation genetic testing for aneuploidy (PGT-A). Unfortunately, each of these techniques may represent a possible confounding factor to the identification of a precise relationship between ART procedures and obstetric or neonatal outcomes, including children and their adulthood health. Many studies report an evident increase of obstetrical risk and perinatal complications with ART, especially for hypertensive disorders of pregnancy (HDP), from gestational hypertension to eclampsia [6,7,8,9,10,11,12,13]. A recent meta-analysis of 50 cohort studies including 161,370 ART and 2,280,241 spontaneously conceived singleton pregnancies found increased risks for several obstetric complications. Significant worst outcomes regarded pregnancy-induced hypertension, placenta previa, abruption, antepartum hemorrhage, oligohydramnios, cesarean delivery, PTB, very low birth weight (VLBW), LBW and perinatal mortality and morbidity [14].
The role of the placenta in these conditions has been demonstrated in several studies. It is well documented that ART can be associated with changes in placental morphology and structure, growth dynamics, imprinted and non-imprinted genes, and other aspects regulating placentation [15]. Several studies demonstrate that incidence of placenta previa was significantly higher in ART than in spontaneous conception (OR 3.14, 95% CI [8]; RR 3.71, 95% CI [16]). Moreover, the placentas from ART pregnancies presented a significantly greater weight and higher placental weight-to-birth weight ratio [17,18]. Some observations indicate that placentas of pregnancies obtained by ART have an increased placental thickness and higher incidence of hematomas [19]. It has been also demonstrated that in children conceived by ART the frequency of imprinting disorders is higher than expected in the general population. ART is associated with increased risk of epigenetic alterations influencing gene expression and DNA methylation in early development and in the placenta, eventually triggering diseases and affecting long-term health [20,21,22]. These findings support the hypothesis of a primary ART responsibility for adverse perinatal outcomes with evident suspicions for anomalies of placentation, although fertility treatments themselves are often associated with impaired placentation and related adverse pregnancy outcomes [1]. Infertile couples who did not undergo ART showed an increased risk of adverse obstetric outcomes, such as placental abruption, fetal loss and GDM [23].
Based on these observations, the main question to address is whether placental alterations (from macroscopic to molecular level) observed in treated infertile patients are the result of ART or if they originate from infertility itself. A deeper investigation of the underlying molecular basis of reduced placental functions in infertility condition and especially after ART treatment is then needed.
In this review, we provide a summary of the most recent literature on placental dysfunction associated with obstetric, perinatal outcomes in singletons born after NON-ART and ART treatments. ART and infertility may be the cause of common dysregulated pathways, including changes in trophoblast invasion, environmental conditions, vascular defects, chronic inflammation, and oxidative stress. Each of them can be prevalent or coexist with others with different and common molecular pathways in a grading and timing that can configurate many clinical features, leading often to neonatal–adult and maternal consequences. This review is based on systematic reviews (SRs), large cohort studies, meta-analyses, and genetic, epigenetic and molecular studies.

2. Abnormal Placentation and ART: Molecular Factors and Involved Signaling

Abnormal placentation may present in a variety of phenotypes, severity, clinical conditions and consequences as the result of several types of infertility treatments and techniques used in ART. For these reasons, it becomes difficult to linearly relate placental influence to obstetrical and perinatal (or neonatal) outcomes after ART.
An altered expression of factors and molecules involved in proper placental development, leading to impaired trophoblast invasion and subsequent reduced vascular remodeling and placenta hypoperfusion, sustain several clinical conditions leading to obstetric and perinatal risks often found in ART pregnancies, such as PE, FGR and placenta previa or accrete (Figure 1) [24,25,26,27,28]. Syncytiotrophoblast stress has been associated with a dysregulated expression of placental growth factor (PlGF) and soluble fms-like tyrosine kinase 1 (sFLT-1) [29,30]. Circulating levels of the anti-angiogenic factor sFLT-1 are increased, and those of PlGF are decreased even before the onset of the clinical symptoms of PE and FGR [31,32,33,34,35]. The increased ratio sFLT-1:PlGF is thought to contribute to the systemic endothelial response and correlate with the severity of FGR and PE. A recent study indicates that the release of sFLT-1 from the placenta is regulated by the epidermal growth factor receptor (EGFR) pathway and the mitochondrial electron transport chain and its downstream pathways, both significantly increased in preeclamptic placentas [36]. The inhibition of these signaling pathways significantly reduces sFLT-1 release from primary cytotrophoblast cells [36]. Vrooman demonstrated in the rat model that an embryo culture from the 1-cell to blastocyst stage increased levels of sFLT-1 together with placental overgrowth, reduced fetal weight, and lower placental DNA methylation [37].
Over the last decades, several other factors have been demonstrated to be altered in pregnancy disorders associated with abnormal placentation (e.g., soluble endoglin (sEndoglin), PlGF and epidermal growth factor-like domain 7 (EGFL7)), with the aim to create a panel of markers to allow an earlier and more precise diagnosis of PE. sEndoglin has been shown to be upregulated in abnormal placenta, typical of FGR and PE, and released into the maternal blood, where it acts as antiangiogenic factor by inhibiting transforming growth factor-beta (TGF-β) signaling in the vasculature. sEndoglin markedly increased, beginning 2 to 3 months before clinical manifestations of PE [38,39]. EGFL7, originally discovered as a largely endothelial restricted gene, has been recently demonstrated to be expressed in the placenta and involved in placental angiogenesis and trophoblast migration [40,41,42]. Altered EGFL7 expression is associated with abnormal placentation and systemic maternal endothelial dysfunction, observed in PE [40,43,44]. Maternal treatment with nitric oxide (NO) donors increases placental EGFL7 levels and improves maternal hemodynamic state and perinatal outcome [45,46]. In the maternal circulation, endothelial dysfunction and abnormal hemodynamic state are associated with increased levels of EGFL7, which return to control levels after NO treatment [44]. Moreover, EGFL7 dosage in maternal circulation allows to discriminate between PE and FGR [43]. Although there are no data correlating EGFL7 with ART, dosage of its circulating levels could help identify ART-associated abnormal placentation.
In FGR and placental insufficiency, the levels of the hormone melatonin are significantly reduced, and this decrease is correlated with proinflammatory activities of tumor necrosis factor alpha (TNF-α), interleukine-1beta (IL-1β), and IL-6 [47]. Melatonin is an antioxidant factor and an anti-inflammatory agent [48,49]. Several studies support the production of this hormone in the ovary as a whole [50,51], the granulosa cells, including those making up the cumulus oophorus [52,53], and the oocyte [54]. In women undergoing ART, melatonin significantly increased the implantation rate [55]; a similar result was obtained in women affected by PCOS undergoing intrauterine insemination [56]. Melatonin crosses the cell membrane, thus interacting with intracellular molecules via different signaling pathways and displaying scavenger functions [57]. Melatonin upregulates the primary implantation receptors, ErbB1 and ErbB4, and significantly reduces intracellular ROS in mouse blastocysts (increasing the embryo total antioxidant capacity) and promotes mitosis of the inner cell mass and trophectoderm cells [58]. Recently, it has been demonstrated that the supplementation of culture medium with melatonin (10−9 mol/L) improves the growth of mouse parthenogenetic embryo potentially by promoting cell cycle progression [59]. Despite all these functions, melatonin is not present in culture media used in ART. We could speculate that the addition of melatonin to the ART media may be beneficial.

3. Pathophysiology of the Placenta in Pregnancy Complications and ART Pregnancies

Altered hormonal milieu, epigenetic changes, immune activity dysregulation, dysmetabolism and inflammation are all conditions attributable to ART and associated with an abnormal placentation. This section provides a theoretical basis on placental changes caused by ART that are most strongly associated with an increased risk of fetal, neonatal and long-term diseases.

3.1. Altered Hormonal Milieu Effect on Placental Development

The interpretation of the effects of ARTs on pregnancy and the possible associated pathologies is complicated by the supra-physiological hormonal levels in the recipient, as a consequence of the ovarian stimulation treatment [37]. Bourgain and Devroey in 2003 suggested that increased hormone blood levels might alter the timing of endometrial receptivity, with possible suboptimal embryo implantation and development [60]. It was also suspected, by studies in animal models, that high estrogen levels exert a detrimental effect on spiral artery remodeling by the trophoblast [61]. Moreover, it has been observed that, in FET procedures, levels of progesterone higher than 32.5 ng/mL on the day of embryo transfer, were associated with no pregnancy [62]. This cut off level seems improbable in ovarian controlled stimulation conditions of fresh IVF cycles, where many corpora lutei are functioning and progesterone level, together with estradiol, are much higher. Conversely, the lack of hormones might also affect placental development by creating conditions leading to increased risks for HDP, as it has been suggested for FET following hormonal endometrial preparation [63,64]. Using these protocols, ovulation does not occur, and the corpus luteum, which secretes important protective vasoactive substances, is not formed [9,10,65]. Among these factors, relaxin is a potent vasodilator promoting vascular compliance [66] and facilitating the adaptation of the maternal cardiovascular system to pregnancy [67]. Moreover, in programmed FET cycles, the use of estradiol and progesterone has been recently associated with obstetric adverse outcomes, i.e., PTB, LBW, and PE, which are small for gestational age (SGA) and large for gestational age (LGA) when compared to natural FET cycle [9]. Thyroid hormone (TH) has been also demonstrated as an important player in reproduction; both hypothyroidism and hyperthyroidism have been associated with subfertility, recurrent miscarriages, and adverse pregnancy outcomes [68,69]. It has been recently demonstrated that TH regulates protease expression and activation of Notch signaling in implantation and embryo development [70]. Moreover, levothyroxine (LT4) treatment has a positive effect on conception capacity and reduces miscarriages when administered to euthyroid women with autoimmune thyroid disease affected by recurrent miscarriages [71].
Several common infertility conditions, such as endometriosis and PCOS, often treated with ART, are characterized by a hormonal dysregulation that affects proper placentation. In endometriosis, a defective deep placentation may derive from functional abnormalities of the eutopic endometrium, as well as an imbalance in endocrine and inflammatory markers [72]. PTB, placenta previa, placental abruption, gestational hypertension, PE, LBW, SGA, cesarean delivery, postpartum hemorrhage, and stillbirth have been significantly associated with endometriosis in a systematic review and meta-analyses, including in 39 studies [73]. PCOS is another condition leading to infertility, in which the defective trophoblast invasion and placentation may be caused by mother’s hyperandrogenism [74,75]. Testosterone can act directly on trophoblast invasion, with modifications of placenta morphology and function [76,77]. PCOS is a chronic low-grade inflammation associated with metabolic dysfunction that is enhanced in pregnancy by the induction of an endothelial dysfunction [78]. This condition might in turn prevent normal remodeling of spiral vessels and the physiologic decrease of uterine artery impedance, thus reducing the depth of endovascular trophoblast invasion. As a consequence of these patterns, in patients with PCOS the placental weight, thickness, density and volume was found significantly reduced [76]. In women with PCOS undergoing IVF, the risk of adverse pregnancy outcomes was found to be significantly elevated. In a recent meta-analysis including 29 observational studies, Sha et al. (2019) described an increased risk for GDM, pregnancy-induced hypertension (PIH), LGA, miscarriage and PTB in women with PCOS undergoing ART [79].

3.2. Epigenetic Changes after ART Techniques Are Associated with Altered Gene Expression in the Placenta and Congenital Imprinting Disorders

Epigenetic regulatory mechanisms can originate from several sources: direct DNA methylation, non-coding RNA, imprinting, and post-translational modification of histone proteins and chromatin remodeling [80]. According to some clinical studies, it was speculated that prolonged exposure to extracorporeal environment might predispose human embryos to disorders of genomic imprinting and to epigenetic modification [81]. Each stage of embryo development, from fertilization to blastocyst formation, also in humans, can undergo to epigenetic changes and consequent differential gene expression in the placenta [82,83]. In the pre-implantation period, the embryonic epigenome is entirely reprogrammed by specific methylations [84], leading to an increase in the risk of gene expression alterations under specific circumstances. For example, there is evidence that aberrant fetal DNA methylation might cause abnormal development of the placenta and recurrent spontaneous abortion [85].
We must consider that ART procedures occur simultaneously to this extensive epigenetic reprogramming and we cannot rule out that the stress involved in ART might affect the establishment and/or maintenance of genomic imprinting (Figure 2). Actually, more than 100 imprinted genes have been found to be strictly involved in the growth and development of embryos [15]. In placentas from IVF pregnancies, a different methylation pattern was observed in comparison to non-IVF gestations and environmental conditions were considered responsible for these findings (Figure 2) [82,83]. IVF disturbs the DNA methylation of the imprinting control region of H19/Igf2, Snrpn, Peg3, and Kcnq1ot1 genes, inducing morphological alterations in the placenta and an increased risk of adult metabolic syndrome [86]. Studies using animal models have demonstrated a dysregulation of several maternally and paternally expressed imprinted genes (e.g., Kcnq1ot1 involved in placental growth, Peg10 that is required for the differentiation of placental spongiotrophoblast and labyrinth, Peg11/Rtl1 involved the development of placental labyrinth and nutrient passive transport, Sfmbt2 playing a key role in the maintenance of trophoblast cell types) in the IVF group as compared to the control group [87,88,89]. Most of these genes play an important role in the proper placental morphology and function. Moreover, specific transcription factors, such as GATA binding protein 3 (GATA3), produced by trophectoderm at blastocyst stage, may be responsible for tissue or cell altered invasion and migration under the influence of the local environment [90].
Consequently, particular attention was paid to blastocyst stage transfer and its possible adverse effect on perinatal outcome. In this regard, there are data from animal studies demonstrating that in vitro fertilization and embryo culture can strongly impact the placental transcriptome [91,92]. In a multicenter double-blind randomized controlled trial, including 836 couples, a significative reduction of birthweight (158 g) and an increase of PTB (8.6% vs. 2.2%) was associated with one of the two culture media used for embryo culture [93]. Moreover, it has been demonstrated that in vitro culture conditions may obstacle mitochondrial maturation and oxidative phosphorylation, inducing cells to increase glycolysis with the aim of maintaining energetic homeostasis: this pathway is represented by the so-called “Warburg effect” [94]. This adaptation pathway may induce epigenetic changes with possible adverse consequences on fetal growth (Figure 2) [94]. There is evidence that the longer the in vitro embryo culture lasts (i.e., blastocyst transfer in comparison to cleavage stage), the more epigenetic changes that can occur [81,94], especially in the developing placenta, although this is not surprising. In this respect, we should consider that this organ develops from the outer trophoblastic layer and consequently is more sensible to epigenetic regulatory changes caused by environmental manipulation and influences during ART. Placental overgrowth was demonstrated by Vroman in the rat model, where embryo culture from 1-cell to blastocyst stage showed adverse outcomes. Reduced fetal weight and lower placental DNA methylation, together with the abovementioned increased levels of sFLT-1, were observed [37]. Additional evidence of aberrant DNA methylation following in vitro embryo culture has been obtained using the bovine model [95]. In this study, 2-, 8- and 16-cell stage embryos were flushed from the oviducts and allowed to further develop in vitro up to the blastocyst stage and their methylome investigated. It was clear that each step of the in vitro culture gave a contribution to epigenetic alterations and that the highest levels resulted around the time of early genomic activation of embryo. In this regard, a recent discovery demonstrated that human embryonic genomic activation initiates at one-cell stage [96].
However, it would be important to distinguish the potential contribution of epigenetic problems related to infertility from epigenetic alteration deriving from the culture conditions [97]. Besides the ART procedures themselves, differential methylation has also been identified in placentas of pregnancies conceived from couples with infertility utilizing different fertility treatments [82,98]. Differential DNA methylation in placentas from pregnancies conceived with different fertility treatments, even as early as the late first trimester, has been described [99].
Furthermore, ART can influence epigenetic regulation of placental formation and function by changing the embryonic environment, placental gene expression, and placental adaptive response to embryonic development [100]. In this respect, Zhang et al. [91] analyzed genes in placental samples obtained from the IVF and control groups, identifying seventeen upregulated and nine downregulated genes. Moreover, when compared to normal pregnancies, the transcriptome profile of first-trimester placental villi appeared significantly altered in IVF-ET pregnancies. A total of 3405 differentially regulated genes in the placenta following IVF (>2-fold change; p < 0.05) was identified by Zhao et al. [92], with 1910 upregulated and 1495 downregulated. The study revealed that these genes are involved in more than 50 biological processes and pathways: coagulation cascades, immune responses, transmembrane signaling, metabolism, cell cycle, stress control, invasion, and angiogenesis, all playing an important role in early pregnancy. From a clinical point of view, many studies have provided useful data on placental pathologies linked to the in vitro environment in which embryo develops. A Swedish population-based registry study from 2002 through 2013 showed an increased risk of placenta previa after blastocyst transfer compared to cleavage-stage transfer (OR 2.18; 95% CI 1.79–2.65) and spontaneous conception (OR 6.38; 95% CI 5.31–7.66) [101]. For FET, higher birthweight, and higher risk of LGA and VLGA were found in blastocyst vs. cleavage stage transfer [102,103,104]. Similar data were reported by Huang (2020), comparing results from day 3 vs. day 7 embryo in 4489 infertile women from 2006 to 2017 [105].
An example of the invasiveness of the in vitro reproductive machinery is the introduction in the clinical practice of the ICSI, which overcomes severe male infertility problems [106], injecting a single sperm into the cytoplasm of a mature oocyte. The mechanical introduction of one sperm escaping the natural control of the fertilization process by the oocyte has raised several criticisms. Reasonable concerns were expressed considering that animal studies on the safety of this practice were limited in comparison with those performed by Edwards before Louise Brown was born in 1978. The revision of some large experience and reviews did not report or demonstrated important consequences of this procedure on the newborn [107], and thus far, the cost–benefit ratio seems reasonable. However, abnormal spermatozoa prepared for ICSI showed improper methylation patterns at imprinting regions, leading to conceive embryos affected by genetic imprinting disorders. Not surprisingly, the alterations in the sperm might explain why ICSI is the fertilization procedure that induced the highest number of differentially expressed genes [108].
An increased reported incidence of autism spectrum disorders in babies born from ART might find some possible source in epigenetic alterations [82]. Numerous studies have reported an increased risk of congenital imprinting disorders in children conceived by ART (Figure 2) [109,110]. In 2019, a wide epidemiological study in Japan found 3.44-, 4.46-, and 8.91-fold increased frequencies of Prader–Willi syndrome (PWS), Beckwith–Wiedemann syndrome (BWS), and Silver–Russell syndrome (SRS) after ART, respectively [111]. In particular, BWS, which is a clinically and genetically heterogeneous overgrowth disease, is caused by epigenetic defects at the 11p15 chromosomal region and a four- to six-fold increased risk of its outcome in ART pregnancies has been reported [22].

3.3. Immune Dysregulation at the Maternal-Fetal Interface

Increased immune activity at the maternal–fetal interface and significant histological and immunohistochemical differences were observed in placentas from pregnancies obtained by IVF of heterologous oocytes, compared to those obtained using homologous oocytes [112], and may be the consequence of a host versus graft rejection-like condition. This aspect was further investigated using 3D ultrasound analysis. This technique demonstrated a more marked reduction of first-trimester placental volume in pregnancies obtained using donor oocytes [113]. A recent retrospective cohort study, including 1114 singleton pregnancies, of which 105 conceived with IVF, further supports the involvement of the immune system in pregnancy complications associated with ART [114]. A higher incidence of villitis was observed in placentas from pregnancies obtained following IVF (16.2% vs. 8.3%; p = 0.007).

3.4. Mechanical Stress on Embryo and Placental Development (the Case of PGT-A)

PGT-A is an ART technique that is increasingly performed, considering that it represents 40% of all IVF cycles in the USA [115]. Its aim is to discover if produced embryos are aneuploid and, in this case, they should not be transferred into the uterus, in order to prevent implantation failure and miscarriages. PGT-A preliminarily requires the in vitro development of embryos up to the blastocyst stage, in order to collect 5 to 10 trophoblast cells. Embryos are then frozen while waiting for the genetic analysis results. Embryos with no or few aneuploidies are thawed and transferred (FET) in the uterine cavity after endometrial hormonal preparation using estrogen and progesterone. In this condition, natural ovulation and formation of the corpus luteum are suppressed. Similar to PGT-A, preimplantation genetic test for monogenic diseases (PGT-M) allows to screen for embryos affected by monogenic diseases.
Some authors have expressed concerns on the possibility that trophectoderm biopsy might disturb embryo development, leading to potential adverse consequences on obstetric outcomes [65,116]. It has been hypothesized that removal of cells from the tissue that will become the placenta does not affect embryo implantation rate but may negatively influence later phases of placentation. Placenta-derived anomalies, such as placenta previa and accreta, HDP, and FGR might be consequences of this abnormal placentation [117,118,119,120].
In a comparative study investigating obstetric and neonatal outcome following PGT-A in blastocyst-stage biopsy with frozen embryo transfer and cleavage-stage biopsy with fresh embryo transfer, Jing et al. (2016) found that PGT-A-FET was associated with a higher incidence of gestational hypertension (9.0% vs. 2.3%; p = 0.017) [121]. These data might indicate a higher risk of placental injury as a consequence of PGT-A procedure on the trophectoderm. Similar indications came from the results of a study on 345 singleton and 76 twin deliveries after PGT-M. PGT-M showed an increased risk of obstetric complications, when compared with pregnancies conceived spontaneously or by IVF without PGT [122]. In particular, these data showed a higher rate of HDP (6.9%), compared with spontaneous conception patients (2.3%) and the IVF group (4.7%). It was reported, as well, a higher rate of SGA neonates (12.4%), in comparison with those from spontaneous conception group (3.9%) and the IVF patients (4.5%). Moreover, Makhijani et al. (2021) observed a significantly higher probability of developing HDP in patients who underwent PGT, compared with those who did not (adjusted odds ratio (aOR) 1.943, 95% CI 1.072–3.521; p = 0.029) [123]. In this study, the potential influence of other factors causing HDP, such as the hormonal endometrial preparation was considered. In their binary logistic regression model, they found that, with the adjusted odds of HDP, the risk associated with trophectoderm biopsy remained significantly higher. A recent large single-center study found a two-fold higher risk of pregnancy-related hypertension following 241 blastocyst biopsy and frozen transfer than in 515 non-biopsied controls [123].
Conversely, is it worthwhile to look for embryonic aneuploidies. It was shown that, among embryos with aneuploid cells, 31% were of meiotic origin and 74% related to mitotic aneuploidies, with maximum aneuploidy originating by days 4–5 after fertilization, falling to 5–6% by day 7 [124]. Consequently, we should consider the environment influence on the mitotic origin of aneuploidies in developing embryos. From a strict clinical point of view, PGT-A does not improve IVF outcomes nor does it reduce miscarriage rates [125]. Moreover, healthy babies have been born after the transfer of aneuploid mosaic embryos [126,127,128]. This is not surprising since it is well known the self-correction ability of aneuploid embryos. It has been demonstrated that, in mice, a p-53-dependent autophagy-mediated apoptotic process is able to eliminate aneuploid cells [129]. In humans, this process is promoted by the bone morphogenetic protein-4 (BMP4) [130]. These mechanisms, together with the fact that the results obtained from a trophectoderm biopsy cannot be representative of the whole embryo [131], contradict the assumption that all aneuploid embryos should be eliminated and constitutes a strong criticism for the PGT-A clinical utility [125]. The Scientific and Clinical Advances Advisory Committee of HFEA recently changed the rating of PGT-A to “red”, meaning that there is no evidence to show that the treatment is effective and safe [125].

4. Metabolic and Cardiovascular Consequences of Placental Dysregulation in Mothers Following ART

Studies demonstrated that vascular dysfunctions in IVF pregnancies were similar to those identified when babies were conceived naturally from mothers affected by PE [132,133]. Sundheimer and Pisarska explain that the size of the placental bed and successful trophoblast invasion and spiral artery remodeling determine maternal blood flow. With these premises, abnormal placentation associated with infertility can represent a consequent marker of overall health for both the mother and her offspring [134]. It has been well known for a long time that inadequate trophoblast invasion and poor spiral artery remodeling may cause reduced vascular perfusion in pregnancy [135].
Women of advanced maternal age are generally at increased risk of GDM, HDP, operative and cesarean deliveries [136,137]; however, those who conceive with the use of IVF are at increased risk of retained placenta, suggesting that placentation abnormalities may contribute to maternal morbidity, and this may be more pronounced in women with infertility [138].
Fertility treatments (IVF and NON-IVF combined) have been associated with an increased risk of severe maternal morbidity (SMM), defined as unexpected outcomes of labor and delivery that result in significant short- or long-term consequences to a woman’s health after [139]. In a population-based cohort study of 114,409 singleton pregnancies with conceptions dating from 11 January 2013 to 10 January 2014 in Ontario, IVF was associated with an increased risk of SMM (rate 11.3/1000; aRR 1.89, 95% CI: 1.06–3.39) [140]. In this study, the authors also noted supra-additive effects of high body mass index (BMI) and IVF on the risk of PE and GDM. The link between fertility treatment and SMM was also confirmed in another cohort study conducted from Ontario between 2006 and 2012, demonstrating that women undergoing fertility treatment are at higher risk of SMM with a relative risk of 1.39 (95% confidence interval (CI) 1.23–1.56). Indicators of SMM were considered severe postpartum hemorrhage, maternal admission to an intensive care unit, puerperal sepsis, hysterectomy and cardiac conditions [141]. The American Heart Association considers a history of PE or HDP as a major risk factor in these women for the development of cardiovascular diseases (CVD) [142].
In summary, cardiovascular and metabolic diseases derived from impaired placentation following infertility treatments may have long-term consequences on both the mother and the newborn.

5. Health Risk in Infancy as a Consequence of Placental Dysfunction Following ART

According to the “developmental origins of health and disease theory” [143] and the Barker hypothesis of the developmental origins of chronic adult disease [144], placental dysfunction and abnormal fetal development may have long-term consequences on the neonate and his development, from childhood to adult life. A higher risk of diabetes mellitus and CVD were found in children whose mothers had PE and HDP, by long-term follow up observations [145,146]. A track of this condition is represented by the finding of reduced endothelial function in mothers and children after PE [147].
Evidence suggests, at least in animal studies, that IVF can promote endothelial dysfunction with increased vessel stiffness in offspring. This can induce arterial hypertension in vivo and an epigenetic origin of the condition has been suggested [132]. IVF causality is also suggested by the significantly elevated activity of enzymatic regulators in the cardiovascular system of IVF offspring [148]. Additionally, there is increasing evidence that offspring conceived by IVF displays a level of vascular dysfunction similar to that seen in children spontaneously conceived by mothers with PE [132,133]. Of note, in 2015 in a randomized case-control trial, antioxidant administration to IVF children was able to improve NO bioavailability and responsiveness of both systemic and pulmonary circulation [149], thus indicating that vascular dysfunctions induced by ART are reversible in young people.
Regarding the increased metabolic risk later in adult life, IVF-conceived mice showed decreased glucose tolerance, increased fasting glucose levels, and reduced insulin-stimulated protein kinase B (Akt) phosphorylation in the liver later in their adult life [150]. These findings are similar to those seen in humans. Some individuals conceived by ART have shown LBW, higher weight, height, and BMI in late infancy [151] and fasting insulin levels [152]. Adolescents conceived by ART showed significant differences in growth kinetics, glucose levels, and blood pressure in comparison with those conceived naturally by sub-fertile parents [153,154], suggesting that these alterations may be caused by ART procedures. Development delay at birth (probably caused by epigenetic alterations) and accelerated growth in late infancy, with metabolic alterations in adolescence, may predispose these individuals to chronic diseases such as obesity and type 2 diabetes [155,156]. An epigenetic programming of metabolism during prenatal and postnatal periods as a response to imprinting alterations that occurred during early embryonic development has been hypothesized [156,157].

6. Simultaneous Action of Factors Dysregulating Normal Placentation

Infertility and its treatments, including ART and its various procedures (ICSI, blastocyst culture + FET + PGT-A with ovarian stimulation or hormonal preparation), may individually or synergistically participate in the dysregulation of embryo and placenta development. Consequently, it is probably impossible to separate the relative contribution of the many factors influencing embryo, placental, and newborn health when an infertile woman is treated with the many types of ART techniques. These techniques may represent confounding factors for a full understanding of previous studies and future ones to be designed.
Each technique may lead to important epigenetic changes and differential gene expression in the placenta [82,83] or damage the developing embryos with thermal, oxidative and mechanical stressing actions [82,98,158].
Pivotal reproductive hormones, such as human chorionic gonadotropin, progesterone and estradiol, are found at high concentrations at the maternal–fetal interface during vessel remodeling. This observation allows to hypothesize that such relevant vascular transformation may be under the influence of these hormones, which may control trophoblast migration [90,159]. A potential role in placental function dysregulation has been proposed for estrogens. In this respect, elevated estrogen exposure (as in controlled ovarian hyperstimulation) has been associated with higher rates of LBW and FGR [160].
Are perinatal adverse outcomes in infertile patients the consequence of ART or are they the consequence of the underlying infertility? A possible answer to this basic question may come from infertile couples conceiving spontaneously with no ART treatments; in these couples, a higher risk of PTB and LBW has been reported [1]. However, an increasing number of observations show vascular dysfunctions similar to those observed in PE in IVF offspring [132,133], and a common mechanism of action could be postulated.
The induction of defective methylation and consequent alteration of gene expression, which may impair placentation [85], can be caused by oxidative stress [161,162], as in endometriosis and by the altered hormonal milieu, i.e., supraphysiologic estradiol levels due to ovarian hyperstimulation [97]. Particular negative effects on embryo development may occur in PGT-A procedures, in which the epigenetic risk for long in vitro culture may be added to the trophectoderm mechanical stress. If the patient is also affected by PCOS, this adverse metabolic condition for the placenta would considerably increase the overall obstetric, perinatal and postnatal final risk [76,77]. All these factors could exert additive effects, leading to the pathologic condition.

7. Final Remarks

The huge and increasing number of ART cycles in the world, together with the discovery of epigenetic, obstetric, maternal and newborn associated risk, should raise concerns in the medical and scientific communities. Observing the three main world ART registers (United States of America, Europe, Australia and New Zealand) during the period 1997–2016, we acknowledge that the numbers of recorded ART treatments increased considerably (5.3-fold in Europe, 4.6-fold in the USA, 3.0-fold in Australia and New Zealand [163]. In 2016, the ICSI technique overcome IVF and accounted for 50% to 60% of all cycles of in vitro insemination. A sharp rise in the number of freeze-all cycles has been observed both in Australia and New Zealand (reaching 26.5% of all oocyte retrieval cycles for IVF and ICSI in 2016) and in the USA (19.2% in 2016). It is relevant to highlight that the majority of the freeze-all cycles involve blastocyst stage embryos. Practically, FET has overcome fresh embryo transfer in the USA and Australia. PGT-A in the USA is performed in 40% of IVF cycles with an increasing trend [115] and, in Australia, represents the fourth most used type of ART technique [163]. This context should be considered when trying to investigate related and prospective ART risks in a growing infertile population. From a clinical point of view and according to the presented data, we can imagine an increasing risk with this pattern: ovarian stimulation < IVF < ICSI < Blastocyst culture < PGT-A. On this basis, we could speculate the higher risk in a patient with PCOS or endometriosis that performs PGT-A.
Considering the many factors and metabolic pathways that we are continuously discovering to be involved in reproductive processes, from fertilization to embryo implantation and feto-placental development, we are far from completing the entire and extremely complex picture. Our experimental findings are able to shed light on small and specific tiles of the complex mosaic regarding many reproductive patterns, often not well known or even completely obscure to our understanding and knowledge.
These observations open the question that regard our medical action, that is, the prevention of consequences linked to artificial procedure that move too far away from physiologic patterns. Not completely knowing the interrelated quite complex molecular pathways skipped or artificially modified, we cannot pharmacologically correct them in a more rational and appropriate way.
With these premises, the prevention of impaired placentation might be considered before starting an ART procedure with a metabolic improvement to women (e.g., dietary and pharmacologic tools in case of PCOS and insulin resistance); additionally, a secondary prevention might be represented by the right choice of therapeutic techniques and protocols (e.g., less invasive embryo culture/procedures and mild stimulation) and with a more personalized obstetric control before pathologic conditions of the placenta become clinically evident as hypertensive or metabolic disorders. If pathologic placentation produces detectable effect starting on the fifth or sixth gestational week, we should not wait until the second or third trimester to observe its clinical consequences, and first trimester miscarriages might not be of genetic origin. A higher prevalence of spontaneous abortion with ART should prompt us to investigate both embryo aneuploidies and placental pathology [164].
In conclusion, it is reasonable to continue research on all key points of the reproductive process, and it is logical to try to correct or prevent (using medical procedures) what is becoming pathologic on the basis of our increasing knowledge. However, maybe it is wiser not to stray too far from the physiology of reproduction. Those who are dealing with ART are requested to study with more interest the physiology and the pathology of the placenta, not only from the usual obstetric point of view. Balancing the pro and cons of our reproductive interventions and observing long-term health of the mother and offspring, remain important topics, although much has to be investigated on biological and clinical levels. With this aim, collaborative studies between clinicians and biologists should be strongly encouraged.

Author Contributions

Conceptualization, C.M. and M.M.; writing—original draft preparation, C.M. and M.M.; writing—review and editing, C.M., V.L., G.R. and M.M.; visualization, G.R. and A.D.L.; supervision, A.D.L. and M.M. 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.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PTBpreterm birth
LBWlow birth weight
PEpreeclampsia
FGRfetal growth restriction
PCOSpolycystic ovary syndrome
GDMgestational diabetes mellitus
ARTassisted reproductive technology
IVFin vitro fertilization
ICSIintracytoplasmic sperm injection
FETfrozen embryo transfer
ODoocyte donation
PGT-Apreimplantation genetic testing for aneuploidy
HDPhypertensive disorders of pregnancy
VLBWvery low birth weight
PlGFplacental growth factor
sFLT-1soluble fms-like tyrosine kinase 1
EGFRepidermal growth factor receptor
sENDOGLINsoluble endoglin
EGFL7epidermal growth factor-like domain 7
TGF-βtransforming growth factor-beta
NOnitric oxide
TNF-αtumor necrosis factor-alpha
ILinterleukin
SGAsmall for gestational age
LGAlarge for gestational age
THthyroid hormone
LT4levothyroxine
PIHpregnancy induced hypertension
PGT-Mpreimplantation genetic test for monogenic diseases
BMP-4bone morphogenetic protein-4
CVDcardiovascular disease
PWSPrader–Willi syndrome
ASAngelman syndrome
BWSBeckwith–Wiedemann syndrome
SRSSilver–Russell syndrome
SMMsevere maternal morbidity

References

  1. Messerlian, C.; Maclagan, L.; Basso, O. Infertility and the risk of adverse pregnancy outcomes: A systematic review and meta-analysis. Hum. Reprod. 2013, 28, 125–137. [Google Scholar] [CrossRef] [Green Version]
  2. Healy, D.L.; Breheny, S.; Halliday, J.; Jaques, A.; Rushford, D.; Garrett, C.; Talbot, J.M.; Baker, H.W. Prevalence and risk factors for obstetric haemorrhage in 6730 singleton births after assisted reproductive technology in Victoria Australia. Hum. Reprod. 2010, 25, 265–274. [Google Scholar] [CrossRef] [Green Version]
  3. Palomba, S.; de Wilde, M.A.; Falbo, A.; Koster, M.P.; La Sala, G.B.; Fauser, B.C. Pregnancy complications in women with polycystic ovary syndrome. Hum. Reprod. Update 2015, 21, 575–592. [Google Scholar] [CrossRef] [Green Version]
  4. Vambergue, A.; Fajardy, I. Consequences of gestational and pregestational diabetes on placental function and birth weight. World J. Diabetes 2011, 2, 196–203. [Google Scholar] [CrossRef]
  5. De Geyter, C.; Calhaz-Jorge, C.; Kupka, M.S.; Wyns, C.; Mocanu, E.; Motrenko, T.; Scaravelli, G.; Smeenk, J.; Vidakovic, S.; Goossens, V.; et al. ART in Europe, 2014: Results generated from European registries by ESHRE: The European IVF-monitoring consortium (EIM) for the European society of human reproduction and Embryology (ESHRE). Hum. Reprod. 2018, 33, 1586–1601. [Google Scholar] [CrossRef]
  6. Helmerhorst, F.M.; Perquin, D.A.M.; Donker, D.; Keirse, M.J.N.C. Perinatal outcome of singletons and twins after assisted conception: A systematic review of controlled studies. BMJ 2004, 328, 261–264. [Google Scholar] [CrossRef] [Green Version]
  7. Pandey, S.; Shetty, A.; Hamilton, M.; Bhattacharya, S.; Maheshwari, A. Obstetric and perinatal outcomes in singleton pregnancies resulting from IVF/ICSI: A systematic review and meta-analysis. Hum. Reprod. Update 2012, 18, 485–503. [Google Scholar] [CrossRef]
  8. Luke, B. Pregnancy and birth outcomes in couples with infertility with and without assisted reproductive technology: With an emphasis on US population-based studies. Am. J. Obstet. Gynecol. 2017, 217, 270–281. [Google Scholar] [CrossRef]
  9. Ginström Ernstad, E.; Wennerholm, U.; Khatibi, A.; Petzold, M.; Bergh, C. Neonatal and maternal outcome after frozen embryo transfer; increased risks in programmed cycles. Am. J. Obstet. Gynecol. 2019, 221, 126.e1–126.e18. [Google Scholar] [CrossRef]
  10. Saito, K.; Kuwahara, A.; Ishikawa, T.; Morisaki, N.; Miyado, M.; Miyado, K.; Fukami, M.; Miyasaka, N.; Ishihara, O.; Irahara, M.; et al. Endometrial preparation methods for frozen-thawed embryo transfer are associated with altered risks of hypertensive disorders of pregnancy, placenta accreta, and gestational diabetes mellitus. Hum. Reprod. 2019, 34, 1567–1575. [Google Scholar] [CrossRef]
  11. Sunderam, S.; Kissin, D.M.; Zhang, Y.; Folger, S.G.; Boulet, S.L.; Warner, L.; Callaghan, W.M.; Barfield, W.D. Assisted reproductive technology surveillance—United States, 2016. MMWR Surveill. Summ. 2019, 68, 1–23. [Google Scholar] [CrossRef] [Green Version]
  12. Luke, B.; Brown, M.B.; Eisenberg, M.L.; Callan, C.; Botting, B.J.; Pacey, A.; Alastair, G. In vitro fertilization and risk for hypertensive disorders of pregnancy: Associations with treatment parameters. Am. J. Obstet. Gynecol. 2020, 222, 350.e1–350.e13. [Google Scholar] [CrossRef]
  13. Manna, C. Maternal-Fetal Medicine, Practical Aspects Il; Arduini, D., Palermo, M.S.F., Eds.; AMOLCA Publishing House: Medellin, Colombia, 2021. [Google Scholar]
  14. Qin, J.; Liu, X.; Sheng, X.; Wang, H.; Gao, S. Assisted reproductive technology and the risk of pregnancy-related complications and adverse pregnancy outcomes in singleton pregnancies: A meta- analysis of cohort studies. Fertil. Steril. 2016, 105, 73–85. [Google Scholar] [CrossRef] [Green Version]
  15. Xiang, M.; Chen, S.; Zhang, X.; Ma, Y. Placental diseases associated with assisted reproductive technology. Reprod. Biol. 2021, 21, 100505. [Google Scholar] [CrossRef]
  16. Yang, X.; Li, Y.; Li, C.; Zhang, W. Current overview of pregnancy complications and live-birth outcome of assisted reproductive technology in mainland China. Fertil. Steril. 2014, 101, 385–391. [Google Scholar] [CrossRef] [PubMed]
  17. Haavaldsen, C.; Tanbo, T.; Eskild, A. Placental weight in singleton pregnancies with and without assisted reproductive technology: A population study of 536,567 pregnancies. Hum. Reprod. 2012, 27, 576–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Reig, A.; Seli, E. The association between assisted reproductive technologies and low birth weight. Curr. Opin. Obstet. Gynecol. 2019, 31, 183–187. [Google Scholar] [CrossRef]
  19. Joy, J.; Gannon, C.; McClure, N.; Cooke, I. Is Assisted Reproduction Associated with Abnormal Placentation? Pediatr. Dev. Pathol. 2012, 15, 306–314. [Google Scholar] [CrossRef]
  20. Ochoa, E. Alteration of Genomic Imprinting after Assisted Reproductive Technologies and Long-Term Health. Life 2021, 11, 728. [Google Scholar] [CrossRef] [PubMed]
  21. Kopca, T.; Tulay, P. Association of Assisted Reproductive Technology Treatments with Imprinting Disorders. Glob. Med. Genet. 2021, 8, 1–6. [Google Scholar] [CrossRef]
  22. Fontana, L.; Tabano, S.; Maitz, S.; Colapietro, P.; Garzia, E.; Gerli, A.G.; Sirchia, S.M.; Miozzo, M. Clinical and Molecular Diagnosis of Beckwith-Wiedemann Syndrome with Single- or Multi-Locus Imprinting Disturbance. Int. J. Mol. Sci. 2021, 22, 3445. [Google Scholar] [CrossRef]
  23. Kroener, L.; Wang, E.T.; Pisarska, M.D. Predisposing Factors to Abnormal First Trimester Placentation and the Impact on Fetal Outcomes. Semin. Reprod. Med. 2016, 34, 27–35. [Google Scholar] [CrossRef] [Green Version]
  24. Brosens, I.; Pijnenborg, R.; Vercruysse, L.; Romero, R. The “Great Obstetrical Syndromes” are associated with disorders of deep placentation. Am. J. Obstet. Gynecol. 2011, 204, 193–201. [Google Scholar] [CrossRef] [Green Version]
  25. Fisher, S.J. Why is placentation abnormal in preeclampsia? Am. J. Obstet. Gynecol. 2015, 213 (Suppl. 4), S115–S122. [Google Scholar] [CrossRef] [Green Version]
  26. Burton, G.J.; Yung, H.W.; Cindrova-Davies, T.; Charnock-Jones, D.S. Placental endoplasmic reticulum stress and oxidative stress in the pathophysiology of unexplained intrauterine growth restriction and early onset preeclampsia. Placenta 2009, 30, S43–S48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Jackson, M.R.; Walsh, A.J.; Morrow, R.J.; Mullen, J.B.; Lye, S.J.; Ritchie, J.W. Reduced placental villous tree elaboration in small-for-gestational-age pregnancies: Relationship with umbilical artery Doppler waveforms. Am. J. Obstet. Gynecol. 1995, 172, 518–525. [Google Scholar] [CrossRef]
  28. Redman, C.W.; Sargent, I.L. Latest advances in understanding preeclampsia. Science 2005, 308, 1592–1594. [Google Scholar] [CrossRef] [PubMed]
  29. Benschop, L.; Schalekamp-Timmermans, S.; Broere-Brown, Z.A.; Roeters van Lennep, J.E.; Jaddoe, V.W.V.; Roos-Hesselink, J.W.; Ikram, M.K.; Steegers, E.A.P.; Robert, J.M.; Gandley, R.E. Placental growth factor as an indicator of maternal cardiovascular risk after pregnancy. Circulation 2019, 139, 1698–1709. [Google Scholar] [CrossRef]
  30. Redman, C.W.; Staff, A.C. Preeclampsia, biomarkers, syncytiotrophoblast stress, and placental capacity. Am. J. Obstet. Gynecol. 2015, 213 (Suppl. 4), S9.e1–S9.e4. [Google Scholar] [CrossRef]
  31. Zeisler, H.; Llurba, E.; Chantraine, F.; Vatish, M.; Staff, A.C.; Sennström, M.; Olovsson, M.; Brennecke, S.P.; Stepan, H.; Allegranza, D.; et al. Pre-dictive Value of the sFlt-1:PlGF Ratio in Women with Suspected Preeclampsia. N. Engl. J. Med. 2016, 374, 13–22. [Google Scholar] [CrossRef]
  32. Stepan, H.; Unversucht, A.; Wessel, N.; Faber, R. Predictive value of maternal angiogenic factors in second trimester pregnancies with abnormal uterine perfusion. Hypertension 2007, 49, 818–824. [Google Scholar] [CrossRef] [Green Version]
  33. Hertig, A.; Berkane, N.; Lefevre, G.; Toumi, K.; Marti, H.P.; Capeau, J.; Uzan, S.; Rondeau, E. Maternal serum sFLT-1 concentration is an early and reliable predictive marker of preeclampsia. Clin. Chem. 2004, 50, 1702–1703. [Google Scholar] [CrossRef]
  34. Tidwell, S.C.; Ho, H.N.; Chiu, W.H.; Torry, R.J.; Torry, D.S. Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am. J. Obstet. Gynecol. 2001, 184, 1267–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Thadhani, R.; Mutter, W.P.; Wolf, M.; Levine, R.J.; Taylor, R.N.; Sukhatme, V.P.; Ecker, J.; Karumanchi, S.A. First trimester placental growth factor and solu-ble fms-like tyrosine kinase 1 and risk for preeclampsia. J. Clin. Endocrinol. Metab. 2004, 89, 770–775. [Google Scholar] [CrossRef] [PubMed]
  36. Hastie, R.; Brownfoot, F.C.; Pritchard, N.; Hannan, N.J.; Cannon, P.; Nguyen, V.; Palmer, K.; Beard, S.; Tong, S.; Kaitu’u-Lino, T.J. EGFR (Epidermal Growth Factor Receptor) Signaling and the Mitochondria Regulate sFlt-1 (Soluble FMS-Like Tyrosine Kinase-1) Secretion. Hypertension 2019, 73, 659–670. [Google Scholar] [CrossRef]
  37. Vrooman, L.A.; Rhon-Calderon, E.A.; Chao, O.Y.; Nguyen, D.K.; Narapareddy, L.; Dahiya, A.K.; Putt, M.E.; Schultz, R.M.; Bartolomei, M.S. Assisted reproductive technologies induce temporally specific placental defects and the PE risk marker sFLT-1 in mouse. Development 2020, 29, 147. [Google Scholar] [CrossRef]
  38. Cui, L.; Shu, C.; Liu, Z.; Tong, W.; Cui, M.; Wei, C.; Tang, J.J.; Liu, X.; Hu, J.; Jiang, J.; et al. The expression of serum sEGFR, sFlt-1, sEndoglin and PLGF in preeclampsia. Pregnancy Hypertens. 2018, 13, 127–132. [Google Scholar] [CrossRef]
  39. Levine, R.J.; Lam, C.; Qian, C.; Yu, K.F.; Maynard, S.E.; Sachs, B.P.; Sibai, B.M.; Epstein, F.H.; Romero, R.; Thadhani, R.; et al. Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N. Engl. J. Med. 2006, 355, 992–1005. [Google Scholar] [CrossRef]
  40. Lacko, L.A.; Massimiani, M.; Sones, J.L.; Hurtado, R.; Salvi, S.; Ferrazzani, S.; Davisson, R.L.; Campagnolo, L.; Stuhlmann, H. Novel expression of EGFL7 in placental trophoblast and endothelial cells and its implication in preeclampsia. Mech. Dev. 2014, 133, 163–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Massimiani, M.; Vecchione, L.; Piccirilli, D.; Spitalieri, P.; Amati, F.; Salvi, S.; Ferrazzani, S.; Stuhlmann, H.; Campagnolo, L. Epidermal growth factor-like domain 7 promotes migration and invasion of human trophoblast cells through activation of MAPK, PI3K and NOTCH signaling pathways. Mol. Hum. Reprod. 2015, 21, 435–451. [Google Scholar] [CrossRef] [Green Version]
  42. Lacko, L.A.; Hurtado, R.; Hinds, S.; Poulos, M.G.; Butler, J.M.; Stuhlmann, H. Altered feto-placental vascularization, feto-placental malperfusion and fetal growth restriction in mice with Egfl7 loss of function. Development. 2017, 144, 2469–2479. [Google Scholar] [CrossRef] [Green Version]
  43. Massimiani, M.; Salvi, S.; Tiralongo, G.M.; Moresi, S.; Stuhlmann, H.; Valensise, H.; Lanzone, A.; Campagnolo, L. Circulating EGFL7 distinguishes between FGR and PE: An observational case-control study. Sci. Rep. 2021, 11, 17919. [Google Scholar] [CrossRef]
  44. Massimiani, M.; Lacko, L.A.; Burke Swanson, C.S.; Salvi, S.; Argueta, L.B.; Moresi, S.; Ferrazzani, S.; Gelber, S.E.; Baergen, R.N.; Toschi, N.; et al. Increased circulating levels of Epidermal Growth Factor-like Domain 7 in pregnant women affected by preeclampsia. Transl. Res. 2019, 207, 19–29. [Google Scholar] [CrossRef]
  45. Massimiani, M.; Tiralongo, G.M.; Salvi, S.; Fruci, S.; Lacconi, V.; La Civita, F.; Mancini, M.; Stuhlmann, H.; Valensise, H.; Campagnolo, L. Treatment of pregnancies complicated by intrauterine growth restriction with nitric oxide donors increases placental expression of Epidermal Growth Factor-Like Domain 7 and improves fetal growth: A pilot study. Transl. Res. 2021, 228, 28–41. [Google Scholar] [CrossRef] [PubMed]
  46. Valensise, H.; Vasapollo, B.; Novelli, G.P.; Giorgi, G.; Verallo, P.; Galante, A.; Arduini, D. Maternal and fetal hemodynamic effects induced by nitric oxide donors and plasma volume expansion in pregnancies with gestational hypertension complicated by intrauterine growth restriction with absent end diastolic flow in the umbilical artery. Ultrasound Obstet. Gynecol. 2008, 31, 55–64. [Google Scholar] [CrossRef]
  47. Berbets, A.; Koval, H.; Barbe, A.; Albota, O.; Yuzko, O. Melatonin decreases and cytokines increase in women with placental insufficiency. J. Matern.-Fetal Neonatal Med. 2019, 25, 1–6. [Google Scholar] [CrossRef] [PubMed]
  48. Chuffa, L.G.; Lupi, L.A., Jr.; Seiva, F.R.; Martinez, M.; Domeniconi, R.F.; Pinheiro, P.F.; Dos Santos, L.D.; Martinez, F.E. Quantitative proteomic profiling reveals that diverse metabolic pathways are influenced by melatonin in an in vivo model of ovarian carcinoma. J. Proteome Res. 2016, 15, 3872–3882. [Google Scholar] [CrossRef]
  49. De Almeida Chuffa, L.G.; Seiva, F.R.F.; Cucielo, M.S.; Silveira, H.S.; Reiter, R.J.; Lupi, L.A. Mitochondrial functions and melatonin: A tour of the reproductive cancers. Cell. Mol. Life Sci. 2019, 76, 837–863. [Google Scholar] [CrossRef] [PubMed]
  50. Itoh, M.T.; Ishizuka, B.; Kudo, Y.; Fusama, S.; Amemiya, A.; Sumi, Y. Detection of melatonin and serotonin N-acetyltransferase and hydroxyindole-Omethyltransferase activities in the rat ovary. Mol. Cell Endocrinol. 1997, 136, 7–14. [Google Scholar] [CrossRef]
  51. Itoh, M.T.; Ishizuka, B.; Kuribayashi, Y.; Amemiya, A.; Sumi, Y. Melatonin, its precursors, and synthesizing enzyme activities in the human ovary. Mol. Hum. Reprod. 1999, 5, 402–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. El-Raey, M.; Geshi, M.; Somfai, T.; Kaneda, M.; Hirako, M.; Abdel-Ghaffar, A.E.; Sosa, G.A.; El-Roos, M.E.A.; Nagai, T. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation in vitro in cattle. Mol. Reprod. Dev. 2011, 78, 250–262. [Google Scholar] [CrossRef]
  53. Amireault, P.; Dube, F. Serotonin and its antidepressant-sensitive transport in mouse cumulus-oocyte complexes and early embryos. Biol. Reprod. 2005, 73, 358–365. [Google Scholar] [CrossRef]
  54. Sakaguchi, K.; Itoh, M.T.; Takahaashi, N.; Tarumi, W.; Ishizuka, B. The rat oocyte synthesises melatonin. Reprod. Fertil. Dev. 2013, 25, 674–682. [Google Scholar] [CrossRef]
  55. Tamura, H.; Takasaki, A.; Miwa, I.; Taniguchi, K.; Maekawa, R.; Asada, H.; Taketani, T.; Matsuoka, A.; Yamagata, Y.; Shimamura, K.; et al. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J. Pineal Res. 2008, 44, 280–287. [Google Scholar] [CrossRef] [PubMed]
  56. Zheng, M.; Tong, J.; Li, W.P.; Chen, Z.J.; Zhang, C. Melaton in concentration in follicular fluid is correlated with antral follicle count (AFC) and in vitro fertilization (IVF) outcomes in women undergoing assisted reproductive technology (ART) procedures. Gynecol. Endocrinol. 2018, 34, 446–450. [Google Scholar] [CrossRef] [PubMed]
  57. De Almeida Chuffa, L.G.; Seiva, F.R.F.; Cucielo, M.S.; Silveira, H.S.; Reiter, R.J.; Lupi, L.A. Clock genes and the role of melatonin in cancer cells: An overview. Melatonin Res. 2019, 2, 133–157. [Google Scholar] [CrossRef]
  58. Moshkdanian, G.; Moghani-Ghoroghi, F.; Pasbakhsh, P.; Nematollahi-Mahani, S.N.; Najafi, A.; Kashani, S.R. Melatonin upregulates ErbB1 and ErbB4, two primary implantation receptors, in pre-implantation mouse embryos. Iran. J. Basic Med. Sci. 2017, 20, 655–661. [Google Scholar] [CrossRef]
  59. Pan, B.; Qazi, I.H.; Guo, S.; Yang, J.; Qin, J.; Lv, T.; Zang, S.; Zhang, Y.; Zeng, C.; Meng, Q.; et al. Melatonin improves the first cleavage of parthenogenetic embryos from vitrified-warmed mouse oocytes potentially by promoting cell cycle progression. J. Anim. Sci. Biotechnol. 2021, 12, 84. [Google Scholar] [CrossRef]
  60. Bourgain, C.; Devroey, P. The endometrium in stimulated cycles for IVF. Hum. Reprod. Update 2003, 9, 515–522. [Google Scholar] [CrossRef]
  61. Bonagura, T.W.; Pepe, G.J.; Enders, A.C.; Albrecht, E.D. Suppression of extravillous trophoblast vascular endothelial growth factor expression and uterine spiral artery invasion by estrogen during early baboon pregnancy. Endocrinology 2008, 149, 5078–5087. [Google Scholar] [CrossRef] [Green Version]
  62. Alyasin, A.; Agha-Hosseini, M.; Kabirinasab, M.; Saeidi, H.; Nashtaei, M.S. Serum progesterone levels greater than 32,5 ng/ml on the day of embryo transfer are associated with lower live birth rate after artificial endometrial preparation: A prospective study. Reprod. Biol. Endocrinol. 2021, 19, 24. [Google Scholar] [CrossRef]
  63. von Versen-Höynck, F.; Schaub, A.M.; Chi, Y.Y.; Chiu, K.H.; Liu, J.; Lingis, M.; Stan Williams, R.; Rhoton-Vlasak, A.; Nichols, W.W.; Fleischmann, R.R.; et al. Increased preeclampsia risk and reduced aortic compliance with in vitro fertilization cycles in the absence of a corpus luteum. Hypertension 2019, 73, 640–649. [Google Scholar] [CrossRef]
  64. Makhijani, R.; Bartels, C.; Godiwala, P.; Bartolucci, A.; Nulsen, J.; Grow, D.; Benadiva, C.; Engmann, L. Maternal and perinatal outcomes in programmed versus natural vitrified-warmed blastocyst transfer cycles. Reprod. Biomed. Online 2020, 41, 300–308. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, W.Y.; von Versen-Höynck, F.; Kapphahn, K.I.; Fleischmann, R.R.; Zhao, Q.; Baker, V.L. Maternal and neonatal outcomes associated with trophectoderm biopsy. Fertil. Steril. 2019, 112, 283–290. [Google Scholar] [CrossRef] [PubMed]
  66. Marshall, S.A.; Leo, C.H.; Senadheera, S.N.; Girling, J.E.; Tare, M.; Parry, L.J. Relaxin deficiency attenuates pregnancy-induced adaptation of the mesenteric artery to angiotensin II in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 310, R847–R857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. von Versen-Höynck, F.; Strauch, N.K.; Liu, J.; Chi, Y.Y.; Keller-Woods, M.; Conrad, K.P.; Baker, V.L. Effect of mode of conception on maternal serum relaxin, creatinine, and sodium concentrations in an infertile population. Reprod. Sci. 2019, 26, 412–419. [Google Scholar] [CrossRef]
  68. Obregon, M.J.; Mallol, J.; Pastor, R.; Morreale de Escobar, G.; Escobar del Rey, F. L-thyroxine and 3,5,3’-triiodo-L-thyronine in rat embryos before onset of fetal thyroid function. Endocrinology 1984, 114, 305–307. [Google Scholar] [CrossRef] [Green Version]
  69. van den Boogaard, E.; Vissenberg, R.; Land, J.A.; van Wely, M.; van der Post, J.A.; Goddijn, M.; Bisschop, P.H. Significance of (sub)clinical thyroid dysfunction and thyroid autoimmunity before conception and in early pregnancy: A systematic review. Hum. Reprod. Update 2011, 17, 605–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Piccirilli, D.; Baldini, E.; Massimiani, M.; Camaioni, A.; Salustri, A.; Bernardini, R.; Centanni, M.; Ulisse, S.; Moretti, C.; Campagnolo, L. Thyroid hormone regulates protease expression and activation of Notch signaling in implantation and embryo development. J. Endocrinol. 2018, 236, 1–12. [Google Scholar] [CrossRef]
  71. Dal Lago, A.; Galanti, F.; Miriello, D.; Marcoccia, A.; Massimiani, M.; Campagnolo, L.; Moretti, C.; Rago, R. Positive Impact of Levothyroxine Treatment on Pregnancy Outcome in Euthyroid Women with Thyroid Autoimmunity Affected by Recurrent Miscarriage. J. Clin. Med. 2021, 10, 2105. [Google Scholar] [CrossRef]
  72. Brosens, I.; Brosens, J.J.; Fusi, L.; Al-Sabbagh, M.; Kuroda, K.; Benagiano, G. Risks of adverse pregnancy outcome in endometriosis. Fertil. Steril. 2012, 98, 30–35. [Google Scholar] [CrossRef] [Green Version]
  73. Breintoft, K.; Pinnerup, R.; Henriksen, T.; Rytter, D.; Uldbjerg, N.; Forman, A.; Arendt, L. Endometriosis and Risk of Adverse Pregnancy Outcome: A Systematic Review and Meta-Analysis. J. Clin. Med. 2021, 10, 667. [Google Scholar] [CrossRef]
  74. Jindal, P.; Regan, L.; Fourkala, E.O.; Rai, R.; Moore, G.; Goldin, R.D.; Sebire, N.J. Placental pathology of recurrent spontaneous abortion: The role of histopathological examination of products of conception in routine clinical practice: A mini review. Hum. Reprod. 2007, 22, 313–316. [Google Scholar] [CrossRef] [PubMed]
  75. Longtine, M.S.; Nelson, D.M. Placental dysfunction and fetal programming: The importance of placental size, shape, histopathology, and molecular composition. Semin. Reprod. Med. 2011, 29, 187–196. [Google Scholar] [CrossRef] [Green Version]
  76. Palomba, S.; Russo, T.; Falbo, A.; Di Cello, A.; Tolino, A.; Tucci, L.; La Sala, G.B.; Zullo, F. Macroscopic and microscopic findings of the placenta in women with polycystic ovary syndrome. Hum. Reprod. 2013, 28, 2838–2847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Palomba, S.; Russo, T.; Falbo, A.; Di Cello, A.; Amendola, G.; Mazza, R.; Tolino, A.; Zullo, F.; Tucci, L.; La Sala, G.B. Decidual endovascular trophoblast invasion in women with polycystic ovary syndrome: An experimental case-control study. J. Clin. Endocrinol. Metab. 2012, 97, 2441–2449. [Google Scholar] [CrossRef]
  78. Palomba, S.; Falbo, A.; Russo, T.; Battista, L.; Tolino, A.; Orio, F.; Zullo, F. Uterine blood flow in pregnant patients with polycystic ovary syndrome: Relationships with clinical outcomes. BJOG 2010, 117, 711–721. [Google Scholar] [CrossRef] [PubMed]
  79. Sha, T.; Wang, X.; Cheng, W.; Yan, Y. A meta-analysis of pregnancy-related outcomes and complications in women with polycystic ovary syndrome undergoing IVF. Reprod. Biomed. Online 2019, 39, 281–293. [Google Scholar] [CrossRef] [Green Version]
  80. Maccani, M.A.; Marsit, C.J. Epigenetics in the Placenta. Am. J. Reprod. Immunol. 2009, 62, 78–89. [Google Scholar] [CrossRef] [Green Version]
  81. Maheshwari, A.; Hamilton, M.; Bhattacharya, S. Should we be promoting embryo transfer at blastocyst stage? Reprod. Biomed. Online 2016, 32, 142–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Katari, S.; Turan, N.; Bibikova, M.; Erinle, O.; Chalian, R.; Foster, M.; Gaughan, J.P.; Coutifaris, C.; Sapienza, C. DNA methylation and gene expression differences in children conceived in vitro or in vivo. Hum. Mol. Genet. 2009, 18, 3769–3778. [Google Scholar] [CrossRef]
  83. Nelissen, E.C.; van Montfoort, A.P.; Dumoulin, J.C.; Evers, J.L. Epigenetics and the placenta. Hum. Reprod. Update 2011, 17, 397–417. [Google Scholar] [CrossRef] [Green Version]
  84. Robins, J.C.; Marsit, C.J.; Padbury, J.F.; Sharma, S.S. Endocrine disruptors, environmental oxygen, epigenetics and pregnancy. Front. Biosci. 2011, 3, 690–700. [Google Scholar] [CrossRef] [Green Version]
  85. Novakovic, B.; Rakyan, V.; Ng, H.K.; Manuelpillai, U.; Dewi, C.; Wong, N.C.; Morley, R.; Down, T.; Beck, S.; Craig, J.M.; et al. Specific tumour-associated methylation in normal human term placenta and first-trimester cytotrophoblasts. Hum. Reprod. 2008, 14, 547–554. [Google Scholar] [CrossRef] [Green Version]
  86. de Waal, E.; Mak, W.; Calhoun, S.; Stein, P.; Ord, T.; Krapp, C.; Coutifaris, C.; Schultz, R.M.; Bartolomei, M.S. In vitro culture increases the frequency of stochastic epigenetic errors at imprinted genes in placental tissues from mouse concepti produced through assisted reproductive technologies. Biol. Reprod. 2014, 90, 22. [Google Scholar] [CrossRef] [PubMed]
  87. Li, B.; Chen, S.; Tang, N.; Xiao, X.; Huang, J.; Jiang, F.; Huang, X.; Sun, F.; Wang, X. Assisted reproduction causes reduced fetal growth associated with downregulation of paternally expressed imprinted genes that enhance fetal growth in mice. Biol. Reprod. 2016, 94, 45. [Google Scholar] [CrossRef] [Green Version]
  88. Xiang, M.; Ma, Y.; Lei, H.; Wen, L.; Chen, S.; Wang, X. In vitro fertilization placenta overgrowth in mice is associated with downregulation of the paternal imprinting gene H19. Mol. Reprod. Dev. 2019, 86, 1940–1950. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, S.; Sun, F.Z.; Huang, X.; Wang, X.; Tang, N.; Zhu, B.; Li, B. Assisted reproduction causes placental maldevelopment and dysfunction linked to reduced fetal weight in mice. Sci. Rep. 2015, 5, 10596. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, B.; Kroener, L.L.; Xu, N.; Wang, E.T.; Banks, A.; Williams, J.; Goodarzi, M.O.; Chen, Y.I.; Tang, J.; Wang, Y.; et al. Function and Hormonal Regulation of GATA3 in Human First Trimester Placentation. Biol. Reprod. 2016, 95, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Zhang, Y.; Cui, Y.; Zhou, Z.; Sha, J.; Li, Y.; Liu, J. Altered global gene expressions of human placentae subjected to assisted reproductive technology treatments. Placenta 2010, 31, 251–258. [Google Scholar] [CrossRef]
  92. Zhao, L.; Zheng, X.; Liu, J.; Zheng, R.; Yang, R.; Wang, Y.; Sun, L. The placental transcriptome of the first- trimester placenta is affected by in vitro fertilization and embryo transfer. Reprod. Biol. Endocrinol. 2019, 17, 50. [Google Scholar] [CrossRef]
  93. Kleijkers, S.H.M.; Mantikou, E.; Slappendel, E.; Consten, D.; van Echten-Arends, J.; Wetzels, A.M.; van Wely, M.; Smits, L.J.M.; van Montfoort, A.P.A.; Repping, S.; et al. Influence of embryo culture medium (G5 and HTF) on pregnancy and perinatal outcome after IVF: A multicenter RCT. Hum. Reprod. 2016, 31, 2219–2230. [Google Scholar] [CrossRef]
  94. Cagnone, G.; Sirard, M.A. The embryonic stress response to in vitro culture: Insight from genomic analysis. Reproduction 2016, 152, R247–R261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Salilew-Wondim, D.; Saeed-Zidane, M.; Hoelker, M.; Gebremedhn, S.; Poirier, M.; Pandey, H.O.; Tholen, E.; Neuhoff, C.; Held, E.; Besenfelder, U.; et al. Genome-wide DNA methylation patterns of bovine blastocysts derived from in vivo embryos subjected to in vitro culture before, during or after embryonic genome activation. BMC Genom. 2018, 19, 424. [Google Scholar] [CrossRef] [Green Version]
  96. Asami, M.; Lam, B.; Ma, M.K.; Rainbow, K.; Braun, S.; VerMilyea, M.D.; Yeo, G.; Perry, A. Human embryonic genome activation initiates at the one-cell stage. Cell Stem Cell 2021. [Google Scholar] [CrossRef]
  97. Berntsen, S.; Söderström-Anttila, V.; Wennerholm, U.B.; Laivuori, H.; Loft, A.; Oldereid, N.B.; Romundstad, L.B.; Bergh, C.; Pinborg, A. The health of children conceived by ART: “The chicken or the egg?”. Hum. Reprod. Update 2019, 25, 137–158. [Google Scholar] [CrossRef] [PubMed]
  98. Song, S.; Ghosh, J.; Mainigi, M.; Turan, N.; Weinerman, R.; Truongcao, M.; Coutifaris, C.; Sapienza, C. DNA methylation differences between in vitro- and in vivo-conceived children are associated with ART procedures rather than infertility. Clin. Epigenet. 2015, 7, 41. [Google Scholar] [CrossRef] [Green Version]
  99. Xu, N.; Barlow, G.M.; Cur, J.; Wang, E.T.; Lee, B.; Akhlaghpour, M.; Kroener, L.; Williams, J.; Rotter, J.I.; Chen, Y.I.; et al. Comparison of genome-wide and gene-specific DNA methylation profiling in first trimester chorionic villi from pregnancies conceived with infertility treatments. Reprod. Sci. 2017, 24, 996–1004. [Google Scholar] [CrossRef] [PubMed]
  100. Choux, C.; Carmignac, V.; Bruno, C.; Sagot, P.; Vaiman, D.; Fauque, P. The placenta: Phenotypic and epigenetic modifications induced by Assisted Reproductive Technologies throughout pregnancy. Clin. Epigenet. 2015, 7, 87. [Google Scholar] [CrossRef]
  101. Ginstrom Ernstad, E.; Bergh, C.; Khatibi, A.; Kallen, K.B.; Westlander, G.; Nilsson, S.; Wennerholm, U.B. Neonatal and maternal outcome after blastocyst transfer: A population-based registry study. Am. J. Obstet. Gynecol. 2016, 214, 378.e1–378.e10. [Google Scholar] [CrossRef]
  102. Makinen, S.; Soderstrom-Anttila, V.; Vainio, J.; Suikkari, A.M.; Tuuri, T. Does long in vitro culture promote large for gestational age babies? Hum. Reprod. 2013, 28, 828–834. [Google Scholar] [CrossRef] [Green Version]
  103. Ishihara, O.; Araki, R.; Kuwahara, A.; Itakura, A.; Saito, H.; Adamson, G.D. Impact of frozen-thawed single-blastocyst transfer on maternal and neonatal outcome: An analysis of 277,042 single-embryo transfer cycles from 2008 to 2010 in Japan. Fertil. Steril. 2014, 101, 128–133. [Google Scholar] [CrossRef]
  104. Zhu, J.; Lin, S.; Li, M.; Chen, L.; Lian, Y.; Liu, P.; Qiao, J. Effect of in vitro culture period on birthweight of singleton newborn. Hum. Reprod. 2014, 29, 448–454. [Google Scholar] [CrossRef] [Green Version]
  105. Huang, J.; Yang, X.; Wu, J.; Kuang, Y.; Wang, Y. Impact of Day 7 Blastocyst Transfer on Obstetric and Perinatal Outcome of Singletons Born After Vitrified-Warmed Embryo Transfer. Front. Physiol. 2020, 11, 74. [Google Scholar] [CrossRef] [PubMed]
  106. Palermo, G.; Joris, H.; Devroey, P.; Van Steirteghem, A.C. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992, 340, 17–18. [Google Scholar] [CrossRef]
  107. Pereira, N.; O’Neill, C.; Lu, V.; Rosenwaks, Z.; Palermo, G.D. The safety of intracytoplasmic sperm injection and long-term outcomes. Reproduction 2017, 154, F61–F70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Feuer, S.K.; Liu, X.; Donjacour, A.; Simbulan, R.; Maltepe, E.; Rinaudo, P. Transcriptional Signatures throughout Development: The Effects of Mouse Embryo Manipulation In Vitro. Reproduction 2017, 153, 107–122. [Google Scholar] [CrossRef] [Green Version]
  109. Hiura, H.; Okae, H.; Miyauchi, N.; Sato, F.; Sato, A.; Van De Pette, M.; John, R.M.; Kagami, M.; Nakai, K.; Soejima, H.; et al. Characterization of DNA Methylation Errors in Patients with Imprinting Disorders Conceived by Assisted Reproduction Technologies. Hum. Reprod. 2012, 27, 2541–2548. [Google Scholar] [CrossRef] [Green Version]
  110. Hiura, H.; Okae, H.; Chiba, H.; Miyauchi, N.; Sato, F.; Sato, A.; Arima, T. Imprinting Methylation Errors in ART. Reprod. Med. Biol. 2014, 13, 193–202. [Google Scholar] [CrossRef] [Green Version]
  111. Hattori, H.; Hiura, H.; Kitamura, A.; Miyauchi, N.; Kobayashi, N.; Takahashi, S.; Okae, H.; Kyono, K.; Kagami, M.; Ogata, T.; et al. Association of Four Imprinting Disorders and ART. Clin. Epigenet. 2019, 11, 1–12. [Google Scholar] [CrossRef] [Green Version]
  112. Gundogan, F.; Bianchi, D.W.; Scherjon, S.A.; Roberts, D.J. Placental pathology in egg donor pregnancies. Fertil. Steril. 2010, 93, 397–404. [Google Scholar] [CrossRef] [PubMed]
  113. Rizzo, G.; Aiello, E.; Pietrolucci, M.E.; Arduini, D. Placental volume and uterine artery Doppler evaluation at 11 + 0 to 13 + 6 weeks’ gestation in pregnancies conceived with in-vitro fertilization: Comparison between autologous and donor oocyte recipients. Ultrasound Obstet. Gynecol. 2016, 47, 726–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ganer Herman, G.; Tamayev, L.; Feldstein, O.; Bustan, M.; Rachimiel, Z.; Schreiber, L.; Raziel, A.; Bar, J.; Kovo, M. Placental-related disorders of pregnancy and IVF: Does placental histological examination explain the excess risk? Reprod. Biomed. Online 2020, 41, 81–87. [Google Scholar] [CrossRef]
  115. Munne, S. Status of preimplantation genetic testing and embryo selection. Reprod. Biomed. Online 2018, 37, 393–396. [Google Scholar] [CrossRef] [PubMed]
  116. Paulson, R.J. Outcome of in vitro fertilization cycles with preimplantation genetic testing for aneuploidies: Let’s be honest with one another. Fertil. Steril. 2019, 112, 1013–1014. [Google Scholar] [CrossRef] [Green Version]
  117. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 134: Fetal growth restriction. Obstet. Gynecol. 2013, 121, 1122–1133. [Google Scholar] [CrossRef]
  118. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 743: Low dose aspirin use in pregnancy. Obstet. Gynecol. 2018, 132, e44–e52. [Google Scholar] [CrossRef]
  119. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 202: Gestational hypertension and pre-eclampsia. Obstet. Gynecol. 2019, 133, 211–218. [Google Scholar] [CrossRef]
  120. American College of Obstetricians and Gynecologists and Society for the Maternal-Fetal Medicine. Obstetrics care consensus: Placenta accreta spectrum. Obstet. Gynecol. 2018, 132, 259–275. [Google Scholar] [CrossRef] [PubMed]
  121. Jing, S.; Luo, K.; He, H.; Lu, C.; Zhang, S.; Tan, Y.; Gong, F.; Lu, G.; Lin, G. Obstetric and neonatal outcomes in blastocyst-stage biopsy with frozen embryo transfer and cleavage-stage biopsy with fresh embryo transfer after preimplantation genetic diagnosis/screening. Fertil. Steril. 2016, 106, 105–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Feldman, B.; Orvieto, R.; Weisel, M.; Aizer, A.; Meyer, R.; Haas, J.; Kirshenbaum, M. Obstetric and Perinatal Outcomes in Pregnancies Conceived After Preimplantation Genetic Testing for Monogenetic Diseases. Obstet. Gynecol. 2020, 136, 782–791. [Google Scholar] [CrossRef]
  123. Makhijani, R.; Bartels, C.B.; Godiwala, P.; Bartolucci, A.; DiLuigi, A.; Nulsen, J.; Grow, D.; Benadiva, C.; Engmann, L. Impact of trophectoderm biopsy on obstetric and perinatal outcomes following frozen–thawed embryo transfer cycles. Hum. Reprod. 2021, 36, 340–348. [Google Scholar] [CrossRef]
  124. Starostic, M.R.; Sosina, O.A.; McCoy, R.C. Single-cell analysis of human embryos reveals diverse patterns of aneuploidy and mosaicism. Genome Res. 2020, 30, 814–835. [Google Scholar] [CrossRef] [PubMed]
  125. Gleicher, N.; Pasquale, P.; Brivanlou, A. Preimplantation Genetic Testing for Aneuploidy—A Castle Built on Sand. Trends Mol. Med. 2021, 27, 731–742. [Google Scholar] [CrossRef]
  126. Gleicher, N.; Vidali, A.; Braverman, J.; Kushnir, V.A.; Albertini, D.F.; Barad, D.H. Further evidence against use of PGS in poor prognosis patients: Report of normal births after transfer of embryos reported as aneuploid. Fertil. Steril. 2015, 104, E59. [Google Scholar] [CrossRef]
  127. Greco, E.; Minasi, M.G.; Fiorentino, F. Healthy babies after intrauterine transfer of mosaic aneuploid blastocysts. N. Engl. J. Med. 2015, 373, 2089–2090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Patrizio, P.; Shoham, G.; Shoham, Z.; Leong, M.; Barad, D.H.; Gleicher, N. Worldwide live births following transfer of chromosomally “abnormal” embryos after PGT/A. Results of worldwide web-based survey. J. Assist. Reprod. Genet. 2019, 36, 1599–1607. [Google Scholar] [CrossRef]
  129. Singla, S.; Iwamoto-Stohl, L.K.; Zhu, M.; Zernicka-Goetz, M. Autophagy-mediated apoptosis eliminates aneuploid cells in a mouse model of chromosome mosaicism. Nat. Commun. 2020, 11, 2958. [Google Scholar] [CrossRef]
  130. Yang, M.; Rito, T.; Naftaly, J.; Hu, J.; Albertini, D.F.; Barad, D.H.; Brivanlou, A.H.; Gleicher, N. Self-correction of mosaicism in human embryos and gastruloids. Nat. Cell. Biol. 2020, 114, E14–E15. [Google Scholar] [CrossRef]
  131. Preimplantation Genetic Testing—Good Practice Recommendations of the European Society of Human Reproduction and Embryology (ESHRE). 2020, p. 124. Available online: www.eshre.eu/guidelines (accessed on 23 November 2021).
  132. Rexhaj, E.; Paoloni-Giacobino, A.; Rimoldi, S.F.; Fuster, D.G.; Anderegg, M.; Somm, E.; Bouillet, E.; Allemann, Y.; Sartori, C.; Scherrer, U. Mice generated by in vitro fertilization exhibit vascular dysfunction and shortened life span. J. Clin. Investig. 2013, 123, 5052–5060. [Google Scholar] [CrossRef] [Green Version]
  133. Jayet, P.Y.; Rimoldi, S.F.; Stuber, T.; Salmòn, C.S.; Hutter, D.; Rexhaj, E.; Thalmann, S.; Schwab, M.; Turini, P.; Sartori-Cucchia, C.; et al. Pulmonary and systemic vascular dysfunction in young offspring of mothers with preeclampsia. Circulation. 2010, 122, 488–494. [Google Scholar] [CrossRef] [Green Version]
  134. Sundheimer, L.; Pisarska, M. Abnormal Placentation Associated with Infertility as a Marker of Overall Health. Semin. Reprod. Med. 2017, 35, 205–216. [Google Scholar] [CrossRef] [PubMed]
  135. Brosens, I.A.; Robertson, W.B.; Dixon, H.G. The role of the spiral arteries in the pathogenesis of preeclampsia. Obstet. Gynecol. Annu. 1972, 1, 177–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Cunningham, F.G.; Leveno, K.J. Childbearing among older women—The message is cautiously optimistic. N. Engl. J. Med. 1995, 333, 1002–1004. [Google Scholar] [CrossRef]
  137. Kenny, L.C.; Lavender, T.; McNamee, R.; O’Neill, S.M.; Mills, T.; Khashan, A.S. Advanced maternal age and adverse pregnancy outcome: Evidence from a large contemporary cohort. PLoS ONE 2013, 8, e56583. [Google Scholar] [CrossRef] [PubMed]
  138. Jackson, S.; Hong, C.; Wang, E.T.; Alexander, C.; Gregory, K.D.; Pisarska, M.D. Pregnancy outcomes in very advanced maternal age pregnancies: The impact of assisted reproductive technology. Fertil. Steril. 2015, 103, 76–80. [Google Scholar] [CrossRef]
  139. Wang, E.T.; Ozimek, J.A.; Greene, N.; Ramos, L.; Vyas, N.; Kilpatrick, S.J.; Pisarska, M.D. Impact of fertility treatment on severe maternal morbidity. Fertil. Steril. 2016, 106, 423–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Dayan, N.; Joseph, K.S.; Fell, D.B.; Laskin, C.A.; Basso, O.; Park, A.L.; Luo, J.; Guan, J.; Ray, J.G. Infertility treatment and risk of severe maternal morbidity: A propensity score-matched cohort study. CMAJ 2019, 191, E118–E127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Dayan, N.; Fell, D.B.; Guo, Y.; Wang, H.; Velez, M.P.; Spitzer, K.; Laskin, C.A. Severe maternal morbidity in women with high BMI in IVF and unassisted singleton pregnancies. Hum. Reprod. 2018, 33, 1548–1556. [Google Scholar] [CrossRef] [PubMed]
  142. Mosca, L.; Benjamin, E.J.; Berra, K.; Bezanson, J.L.; Dolor, R.J.; Lloyd-Jones, D.M.; Newby, L.K.; Piña, I.L.; Roger, V.L.; Shaw, L.J.; et al. Effectiveness-based guidelines for the prevention of cardiovascular disease in women—2011 update: A guideline from the American Heart Association. J. Am. Coll. Cardiol. 2011, 57, 1404–1423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Briana, D.D.; Germanou, K.; Boutsikou, M.; Boutsikou, T.; Athanasopoulos, N.; Marmarinos, A.; Gourgiotis, D.; Malamitsi-Puchner, A. Potential prognostic biomarkers of cardiovascular disease in fetal macrosomia: The impact of gestational diabetes. J. Matern.-Fetal Neonatal. Med. 2018, 31, 895–900. [Google Scholar] [CrossRef]
  144. Barker, D.J. The developmental origins of chronic adult disease. Acta Paediatr. Suppl. 2004, 93, 26–33. [Google Scholar] [CrossRef] [PubMed]
  145. Männistö, T.; Mendola, P.; Vääräsmäki, M.; Järvelin, M.R.; Hartikainen, A.L.; Pouta, A.; Suvanto, E. Elevated blood pressure in pregnancy and subsequent chronic disease risk. Circulation 2013, 127, 681–690. [Google Scholar] [CrossRef] [Green Version]
  146. Brown, M.C.; Best, K.E.; Pearce, M.S.; Waugh, J.; Robson, S.C.; Bell, R. Cardiovascular disease risk in women with pre-eclampsia: Systematic review and meta-analysis. Eur. J. Epidemiol. 2013, 28, 1–19. [Google Scholar] [CrossRef] [PubMed]
  147. Kvehaugen, A.S.; Dechend, R.; Ramstad, H.B.; Troisi, R.; Fugelseth, D.; Staff, A.C. Endothelial function and circulating biomarkers are disturbed in women and children after PE. Hypertension 2011, 58, 63–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Watkins, A.J.; Platt, D.; Papenbrock, T.; Wilkins, A.; Eckert, J.J.; Kwong, W.Y.; Osmond, C.; Hanson, M.; Fleming, T.P. Mouse embryo culture induces changes in postnatal phenotype including raised systolic blood pressure. Proc. Natl. Acad. Sci. USA 2007, 104, 5449–5454. [Google Scholar] [CrossRef] [Green Version]
  149. Rimoldi, S.F.; Sartori, C.; Rexhaj, E.; Bailey, D.M.; de Marchi, S.F.; McEneny, J.; von Arx, R.; Cerny, D.; Duplain, H.; Germond, M.; et al. Antioxidants improve vascular function in children conceived by assisted reproductive technologies: A randomized double-blind placebo-controlled trial. Eur. J. Prev. Cardiol. 2015, 22, 1399–1407. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, M.; Wu, L.; Zhao, J.; Wu, F.; Davies, M.J.; Wittert, G.A. Altered Glucose Metabolism in Mouse and Humans Conceived by IVF. Diabetes 2014, 63, 3189–3198. [Google Scholar] [CrossRef] [Green Version]
  151. Ceelen, M.; Van Weissenbruch, M.M.; Prein, J.; Smit, J.J.; Vermeiden, J.P.W.; Spreeuwenberg, M.; Van Leeuwen, F.E.; Delemarre-Van De Waal, H.A. Growth during Infancy and Early Childhood in Relation to Blood Pressure and Body Fat Measures at Age 8–18 Years of IVF Children and Spontaneously Conceived Controls Born to Subfertile Parents. Hum. Reprod. 2009, 24, 2788–2795. [Google Scholar] [CrossRef] [Green Version]
  152. Guo, X.Y.; Liu, X.M.; Jin, L.; Wang, T.T.; Ullah, K.; Sheng, J.Z.; Huang, H.F. Cardiovascular and Metabolic Profiles of Offspring Conceived by Assisted Reproductive Technologies: A Systematic Review and Meta-Analysis. Fertil. Steril. 2017, 107, 622–631. [Google Scholar] [CrossRef] [Green Version]
  153. Ceelen, M.; Van Weissenbruch, M.M.; Roos, J.C.; Vermeiden, J.P.W.; Van Leeuwen, F.E.; Delemarre-van De Waal, H.A. Body Composition in Children and Adolescents Born after in Vitro Fertilization or Spontaneous Conception. J. Clin. Endocrinol. Metab. 2007, 92, 3417–3423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Ceelen, M.; Van Weissenbruch, M.M.; Vermeiden, J.P.W.; Van Leeuwen, F.E.; Delemarre-Van De Waal, H.A. Cardiometabolic Differences in Children Born after In Vitro Fertilization: Follow-up Study. J. Clin. Endocrinol. Metab. 2008, 93, 1682–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Forse, T.; Eriksson, J.; Tuomilehto, J.; Reunanen, A.; Osmond, C.; Barker, D. The Fetal and Childhood Growth of Persons Who Develop Type 2 Diabetes. Ann. Intern. Med. 2000, 133, 176–182. [Google Scholar] [CrossRef] [PubMed]
  156. Waterland, R.A.; Garza, C. Potential Mechanisms of Metabolic Imprinting That Lead to Chronic Disease. Am. J. Clin. Nutr. 1999, 69, 179–197. [Google Scholar] [CrossRef]
  157. Millership, S.J.; Van de Pette, M.; Withers, D.J. Genomic Imprinting and Its Effects on Postnatal Growth and Adult Metabolism. Cell Mol. Life Sci. 2019, 76, 4009–4021. [Google Scholar] [CrossRef] [Green Version]
  158. Wale, P.L.; Gardner, D.K. The effects of chemical and physical factors on mammalian embryo culture and their importance for the practice of assisted human reproduction. Hum. Reprod. Update 2016, 22, 2–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Chen, J.Z.; Sheehan, P.M.; Brennecke, S.P.; Keogh, R.J. Vessel remodelling, pregnancy hormones and extravillous trophoblast function. Mol. Cell Endocrinol. 2012, 349, 138–144. [Google Scholar] [CrossRef]
  160. Imudia, A.N.; Awonuga, A.O.; Doyle, J.O.; Kaimal, A.J.; Wright, D.L.; Toth, T.L.; Styer, A.K. Peak serum estradiol level during controlled ovarian hyperstimulation is associated with increased risk of small for gestational age and PE in singleton pregnancies after in vitro fertilization. Fertil. Steril. 2012, 97, 1374–1379. [Google Scholar] [CrossRef]
  161. Jones, M.L.; Mark, P.J.; Mori, T.A.; Keelan, J.A.; Waddell, B.J. Maternal dietary omega-3 fatty acid supplementation reduces placental oxidative stress and increases fetal and placental growth in the rat. Biol. Reprod. 2013, 88, 37. [Google Scholar] [CrossRef]
  162. Myatt, L. Review: Reactive oxygen and nitrogen species and functional adaptation of the placenta. Placenta 2010, 31, S66–S69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. De Geyter, C.; Wyns, C.; Calhaz-Jorge, C.; de Mouzon, J.; Ferraretti, A.P.; Kupka, M.; Nyboe Andersen, A.; Nygren, K.G.; Goossens, V. 20 years of the European IVF-monitoring Consortium registry: What have we learned? A comparison with registries from two other regions. Hum. Reprod. 2020, 35, 2832–2849. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, J.X.; Norman, R.J.; Wilcox, A.J. Incidence of spontaneous abortion among pregnancies produced by assisted reproductive technology. Hum. Reprod. 2004, 19, 272–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Molecules regulating proper placental development, whose dysregulation is involved in an abnormal placental development. NO, nitric oxide; VEGF, vascular endothelial growth factor; GATA3, GATA binding protein 3; PlGF, placental growth factor; sFLT-1, soluble fms-like tyrosine kinase 1; sEndoglin, soluble endoglin; pEGFL7, placental epidermal growth factor-like domain 7; cEGFL7, circulating epidermal growth factor-like domain 7; ELA, elabela.
Figure 1. Molecules regulating proper placental development, whose dysregulation is involved in an abnormal placental development. NO, nitric oxide; VEGF, vascular endothelial growth factor; GATA3, GATA binding protein 3; PlGF, placental growth factor; sFLT-1, soluble fms-like tyrosine kinase 1; sEndoglin, soluble endoglin; pEGFL7, placental epidermal growth factor-like domain 7; cEGFL7, circulating epidermal growth factor-like domain 7; ELA, elabela.
Ijms 23 00659 g001
Figure 2. ART techniques affecting epigenetics in the pre-implantation embryo. Epigenetic dysregulation is involved in an abnormal placentation and genetic imprinting disorders. sFLT-1 denotes soluble fms-like tyrosine kinase 1, GATA3 GATA binding protein 3, BWS Beckwith–Wiedemann syndrome, AS Angelman syndrome, PWS Prader–Willi syndrome, SRS Silver–Russell syndrome.
Figure 2. ART techniques affecting epigenetics in the pre-implantation embryo. Epigenetic dysregulation is involved in an abnormal placentation and genetic imprinting disorders. sFLT-1 denotes soluble fms-like tyrosine kinase 1, GATA3 GATA binding protein 3, BWS Beckwith–Wiedemann syndrome, AS Angelman syndrome, PWS Prader–Willi syndrome, SRS Silver–Russell syndrome.
Ijms 23 00659 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Manna, C.; Lacconi, V.; Rizzo, G.; De Lorenzo, A.; Massimiani, M. Placental Dysfunction in Assisted Reproductive Pregnancies: Perinatal, Neonatal and Adult Life Outcomes. Int. J. Mol. Sci. 2022, 23, 659. https://doi.org/10.3390/ijms23020659

AMA Style

Manna C, Lacconi V, Rizzo G, De Lorenzo A, Massimiani M. Placental Dysfunction in Assisted Reproductive Pregnancies: Perinatal, Neonatal and Adult Life Outcomes. International Journal of Molecular Sciences. 2022; 23(2):659. https://doi.org/10.3390/ijms23020659

Chicago/Turabian Style

Manna, Claudio, Valentina Lacconi, Giuseppe Rizzo, Antonino De Lorenzo, and Micol Massimiani. 2022. "Placental Dysfunction in Assisted Reproductive Pregnancies: Perinatal, Neonatal and Adult Life Outcomes" International Journal of Molecular Sciences 23, no. 2: 659. https://doi.org/10.3390/ijms23020659

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