Skip to main content

DNA methylation landscape in pregnancy-induced hypertension: progress and challenges

Abstract

Gestational hypertension (PIH), especially pre-eclampsia (PE), is a common complication of pregnancy. This condition poses significant risks to the health of both the mother and the fetus. Emerging evidence suggests that epigenetic modifications, particularly DNA methylation, may play a role in initiating the earliest pathophysiology of PIH. This article describes the relationship between DNA methylation and placental trophoblast function, genes associated with the placental microenvironment, the placental vascular system, and maternal blood and vascular function, abnormalities of umbilical cord blood and vascular function in the onset and progression of PIH, as well as changes in DNA methylation in the progeny of PIH, in terms of maternal, fetal, and offspring. We also explore the latest research on DNA methylation-based early detection, diagnosis and potential therapeutic strategies for PIH. This will enable the field of DNA methylation research to continue to enhance our understanding of the epigenetic regulation of PIH genes and identify potential therapeutic targets.

Introduction

Pregnancy-induced hypertension (PIH), the most common medical syndrome in pregnancy, is further categorized into four subtypes based on potential differences in pathogenesis and pregnancy outcomes: gestational hypertension, chronic hypertension, preeclampsia (PE), and PE with coexisting chronic hypertension [1,2,3]. PIH not only adversely affects pregnant women and fetuses/newborns, but also increases the risk of various chronic diseases, including cardiovascular disease and metabolic dysfunction, in both mother and offspring [4,5,6,7]. Therefore, elucidating the pathological mechanisms and developing effective therapeutics for PIH remains a priority. Accumulated findings have demonstrated that epigenetic modifications, particularly DNA methylation, relate to heritable changes in placental gene expression that contribute to the development of placental dysfunction and are a critical mediator in the onset of PIH [8, 9]. It is therefore believed that understanding the DNA methylation mechanisms underlying the initiation of PIH may contribute to the identification of novel therapeutic targets for PIH treatment, thereby improving fetal growth and reducing the risk of chronic disease in future generations.

DNA methylation, a stable epigenetic modification of DNA, involves the addition of a methyl group to cytosine in CpG-rich regions of the genome. This process is facilitated by DNA methyltransferase (DNMT), with S-adenosyl-methionine (SAM) acting as the methyl donor. DNMTs can be divided into two primary categories: de novo DNMTs (DNMT3a/3b/3L) and maintenance DNMTs (DNMT1/2) [10]. During active DNA demethylation, an intermediate modification called 5-hydroxymethylcytosine (5hmC) is formed, catalyzed by the ten-eleven translocation (TET1-3) proteins [11]. These enzymes play a pivotal role in embryonic development, establishing, maintaining, and dynamically regulating the somatic gene methylation pattern during embryogenesis. Abnormal DNA methylation has been observed in cells from various sources in individuals with PIH. Researchers have analyzed not only placental cells but also maternal blood cells, cell-free DNA, and umbilical cord blood leukocytes from fetuses. In this review, we present a succinct overview of the existing literature and perform a comprehensive assessment of the involvement of abnormal DNA methylation in the pathogenesis of PIH (Fig. 1). Additionally, we explore the potential utility of DNA methylation markers as predictive biomarkers for the onset and severity of PIH. Furthermore, we outline current investigations focusing on the application of DNA methylation-based approaches for the prevention, diagnosis, and potential therapeutic interventions for PIH.

Fig. 1
figure 1

The outline of this review

DNA methylation in PIH placenta

Although the exact pathogenesis of PIH remains unclear, the placenta is believed to play a key role, and DNA methylation may be associated with altered placental function in PIH progression. This section presents abnormal DNA methylation in PIH placenta, as well as a focus on describing the effects of DNA methylation on placental trophoblast and vascular function.

Global DNA methylation levels in PIH placenta

In recent years, researchers have investigated global DNA methylation in the PIH placentas. Jia et al. analyzed human PE placentas and found 296 genes with significantly aberrant DNA methylation [12]. Blair et al. found significant DNA methylation alterations at 38,840 CpG sites in whole chorionic villus samples from early onset pre-eclampsia (EOPE) placentas, most of which (74.5%) were hypomethylated in EOPE [13]. Xu et al. identified 536 upregulated genes with hypomethylation and 322 downregulated genes with hypermethylation in the PE placentas. Gene ontology analysis showed that these genes were primarily involved in processes such as mitotic cell cycle, calcium homeostasis, and cell proliferation [14]. Although the candidate genes in these studies are distinct, differences in their DNA methylation provide evidence that placental DNA methylation is altered during the pathophysiological course of PIH, and the identification of placental gene-specific methylation profiles may provide valuable insights into pathways that may be epigenetically regulated.

As the severity of PE symptoms can vary among pregnant women due to the disease's heterogeneity, Lim et al. investigated the genome-wide DNA methylation profile of PE placentas based on severity characteristics. They found that DNA hypomethylation was widespread in mild PE compared to PE with severe features. In PE with severe features, hypermethylated CpG sites showed physiopathological relevance [15]. Furthermore, the total amount of DNA methylation in the placenta has been observed to strongly correlate with the gestational age. Li et al. discovered that placental DNA methylation levels were higher in PE and premature birth compared to term birth. The effects of gestational age were ruled out, and a total of 808 differential methylation positions were discovered, the majority of which were hypomethylated in PE placentas [16]. Benjamin et al. reported that dynamic DNA methylation changes in the human placenta can be used to predict gestational age. They developed an accurate tool for predicting the gestational age of placentas and found that EOPE is associated with placental aging [17]. In addition, Chu et al. found evidence that fetal gender influences DNA methylation at autosomal loci in PE placentas [18]. Placenta development and invasion, both of which are influenced by the cytoskeleton and migration, occur more slowly in female fetuses compared to male fetuses [19, 20]. Male placentas have a greater capability for nutrient transfer than female placentas, and their functions throughout pregnancy are distinct [19].

Taken together, it is undeniable that patients with PIH experience dysregulation of placental DNA methylation. It is possible that maternal and fetal genes have independent or interactive impacts on the risk of PIH, and the disease exhibits heterogeneity with varying degrees of severity. Therefore, future studies analyzing placental global DNA methylation should carefully consider PIH's heterogeneity, gestational age, and fetal gender (Fig. 2).

Fig. 2
figure 2

Possible factors affecting the results of global DNA methylation levels in PIH placenta

DNA methylation and placental trophoblast functions

Different types of trophoblast cells have been identified in the placenta, including cytotrophoblast (CTB), syncytiotrophoblast (STB) and extravilloustrophoblast (EVT). These different cell types work together to perform various tasks, such as forming chemical and physical barriers, producing hormones, and maintaining immune tolerance. It is evident that trophoblast malfunctions are linked with various pregnancy disorders and pose a significant threat to the well-being of both the mother and the fetus. Numerous studies have examined placental trophoblast malfunction in PIH from a DNA methylation perspective, focusing on immune deficiency and hormone delivery.

DNA methylation and placental immunodysfunction

During the first trimester of pregnancy, there is a significant increase in innate immune activity, which facilitates the implantation and growth of the embryo and the placenta [21]. Immune dysregulation during implantation can lead to a lack of trophoblast invasion, resulting in a shallow arterial remodeling of the spiraled arteries in the myometrium. This, in turn, leads to inadequate placental perfusion and eventually placental ischemia [3]. Recent studies have reported that DNA methylation changes within many immune-related genes impair the placental trophectoderm, which impedes the development of maternal–fetal immune tolerance and contributes to PIH (Table 1).

Table 1 DNA methylation changes in PIH involved in the regulation of placental immunodeficiency and placental vascular system

Syncytins are a group of membrane glycoproteins. Syncytin-1 is mainly expressed by trophoblast cells, and mediates the fusion of trophoblasts to form STB. It has been reported to participate in regulating inflammation, anti-apoptosis, and immune suppression in PE placentas [22, 23]. Decreased expression of syncytin-1 has been observed in PE placentas, and this decrease has been linked to hypermethylation of its gene promoter [24,25,26]. Ruebner et al. found that overexpression of DNMT3a could be responsible for decreased syncytin-1 expression through promoter hypermethylation, which in turn inhibits trophoblast cell fusion [25]. Based on these findings, it is highly speculated that a broad aberrant DNA methylation pattern within the syncytin-1 gene downregulates its expression, which participates in the pathogenesis of PIH potentially by affecting trophoblast immune and endocrine functions.

Tissue factor (TF) is a membrane glycoprotein that plays a crucial role in fetal and placental damage in mouse models of PE and is involved in the etiology of PE [27, 28]. Liu et al. discovered that TF expression was increased in human PE placentas, and the TF gene promoter had significantly less DNA methylation [28]. The stable and high expression of the TF gene is essential for regulating the development and function of regulatory T (Treg) cells [29, 30], and its presence and relative stability are crucial for ensuring appropriate maternal–fetal tolerance. Consequently, aberrant TF expression in the placenta may lead to the loss of immunosuppressive function, hindering the development of placental immunological tolerance.

Galectins are highly expressed at the maternal–fetal interface and act as key regulator proteins of the immune response in maternal–fetal immune tolerance [31]. Three genes in a Chr19 cluster, encoding galectin 13/14/16, primarily confer immune tolerance in maternal–fetal interactions. Than et al. reported that the low DNA methylation of differentially methylated regions (DMRs) surrounding their transcription start sites may enable the activated transcription of these genes [32]. The dysregulation of these galectin genes in the PE placenta due to DNA methylation can affect immune tolerance at the maternal–fetal interface.

Human leukocyte antigens (HLA) molecules are highly correlated with mother's ability to carry and deliver a healthy allogeneic fetus. Immune tolerance between mother and fetus is partially mediated by HLA, with trophoblast cells expressing HLA-C, non-classical HLA-Ib molecules (HLA-E, -F, and -G), and HLA class II (HLA-DRB1, and -DPB1) antigens [33]. In physiological conditions, HLA-G, which is abundantly and specifically expressed on EVTs, promotes fetal-maternal immune tolerance by reducing NK cell toxicity and cytokine production [34, 35]. Soluble HLA-G also has a regional effect on spiral artery remodeling in the uterus [36]. The significance of HLA-G is further emphasized by the occurrence of PE when its expression at the maternal–fetal interface is abnormal [37, 38]. Several studies have reported that promoter DNA methylation influences HLA-G expression [39, 40]. Tang et al. observed extremely high levels of CpG methylation in the promoter region of dysregulated HLA-G in PE placentas, and demonstrated that DNMT1-mediated promoter hypermethylation of HLA-G is associated with PE [39]. In addition to HLA-G, HLA-DPB1 and HLA-DRB1 have been found to be involved in the interferon γ-mediated signaling pathway cluster specific to PE. Severe PE (SPE) cases show increased promoter DNA methylation of HLA-DPB1 and HLA-DRB1 compared to general PE cases. This difference in DNA methylation levels may indicate a relationship between vulnerability to illness and interferon γ-mediated signaling pathways in SPE [15].

In addition to the major molecules mentioned above, several genes are involved in immune tolerance at the maternal–fetal interface and exhibit altered expression in PE placentas due to DNA methylation modifications. For instance, CD39/ectonucleoside triphosphate diphosphohydrolase-1 (ENTPD1) and zinc finger DHHC-type containing 14 (ZDHHC14) have been found to have promoter hypomethylation in PE placentas, resulting in reduced expression of these genes [41]. ENTPD1, which is the primary ectonucleotidase in fetal trophoblast tissue, is also a surface marker of Treg cells. Reduced expression of ENTPD1 could contributes to the development of clinical characteristics associated with PE [42].

It is evident that abnormalities in trophoblast cell proliferation, migration, invasion, and adhesion during the first trimester of pregnancy result from placental immunodeficiency, which significantly raises the risk of PE. The DNA methylation perspective enhances our understanding of the pathogenesis; however, there is still a substantial knowledge gap, and further studies are necessary to establish the link between DNA methylation and the progression of PIH.

DNA methylation and placental hormone regulation

Glucocorticoids, which belong to the class of steroid hormones, participate in reproduction, including the initiation of pregnancy, successful fetal development, and delivery. Therefore, maintaining appropriate levels of circulating glucocorticoids in the fetus is vital for ensuring normal growth and development within the uterus. The primary bioavailable glucocorticoid is cortisol. It is believed that the enzyme 11β-hydroxysteroid dehydrogenase 2 (11βHSD2) is responsible for protecting the fetus from excessive exposure to maternal glucocorticoids by converting active cortisol into an inactive form [43]. This section aims to summarize whether abnormalities in the glucocorticoid receptor (nuclear receptor subfamily 3, group C, member 1, NR3C1) and 11βHSD2 are involved in the mechanism of PIH occurrence and whether this process is mediated by DNA methylation.

NR3C1 is responsible for the effects of cortisol, and the cortisol-NR3C1 complex can bind to glucocorticoid response elements in the promoters of target genes [44]. NR3C1 expression partially determines the impact of cortisol on blood pressure [45]. A lack of NR3C1 expression in the placenta has negative consequences for fetal health and growth [46]. When BeWo and JEG3 choriocarcinoma cells were exposed to the DNMT1 inhibitor 5-Aza-CdR, significant reductions in promoter DNA methylation of the NR3C1 gene were observed [47]. Evidence suggests that EOPE is associated with an increase in DNA methylation levels at CpG sites of the NR3C1 gene [48]. Notably, Putra et al. observed different patterns of NR3C1 promoter methylation in normotensive, hypertensive, and hypotensive pregnancies and found evidence of an independent association between placental NR3C1 proximal promoter methylation and maternal blood pressure [49]. This independent link between placental NR3C1 gene promoter methylation and maternal blood pressure was supported by data from other studies [48, 50]. It is without a doubt that differential methylation within the NR3C1 gene in placentas is a likely pathological mechanism in PIH.

Placenta contains a barrier against glucocorticoids, which is formed by 11βHSD2. Due to its high affinity for cortisol, 11βHSD2 acts as a placental glucocorticoid barrier, safeguarding the developing fetus from excess glucocorticoids. Disruption of the barrier has been linked to pregnancy complications and the future onset of chronic diseases [51,52,53]. Several studies have reported that the glucocorticoid barrier is damaged in PE placentas [54, 55], accompanied by the increased cortisol levels [56]. SCHOOF et al. found that in the European population (n = 18), 11βHSD2 is downregulated in PE placenta [57]. It is known that 11βHSD2 expression is tightly regulated by DNA methylation [58, 59]. However, Hu et al. found that in the Chinese population (n = 10), the DNA methylation level of the 11βHSD2 gene promoter region in the PE placenta did not change [60]. Current studies on epigenetic alterations of the 11βHSD2 gene in PE are limited and may yield unreliable results due to variations in sample size or population ethnicity.

The placenta plays a significant role in releasing hormones such as progesterone, and placental lactogen, which are crucial for normal pregnancy progression. However, there is currently a lack of studies comparing DNA methylation alterations in placental secretion function between PIH and normal pregnancies. This article aims to serve as a catalyst for further research into the role of DNA methylation in PIH-related placental insufficiency and hopes to inspire academics to investigate this topic.

DNA methylation and placental microenvironment

DNA methylation and imprinted genes

Epigenetic reprogramming, including genomic imprinting, occurs during germ cell development and continues into pre-implantation stages. The expression of imprinted genes is regulated by DNA methylation, which differs between parents. Imprinted genes, genes that are exclusively expressed from one allele-linked to the parental origin of these allele-, are expressed in the placenta. Some are expressed from the paternal allele, while other are expressed from the maternal allele, which serves as a connection between the mother and the developing baby. The human placenta contains a significant number of placenta-specific imprinted transcripts [61, 62]. Many genes in the placenta exhibit parent-of-origin specific imprinting, which may contribute to processes such as trophoblast proliferation, differentiation, angiogenesis, and nutrient transport [63]. Recent studies have reported associations between certain imprinted genes and PIH occurrence [64,65,66] (Fig. 3). These findings enhance our understanding of the pathogenesis of PIH.

Fig. 3
figure 3

The occurrence of PIH is associated with differential DNA methylation of placental genes. This figure illustrates the genes in the placenta that contribute to PIH due to variations in DNA methylation. These genes include imprinted genes, tumor suppressor genes, placental classical signaling pathways, placental microRNAs, and several other protein genes. Abbreviation: hyper, hypermethylation; hypo, hypomethylation; + , promote; -, inhibit; TFPI2, tissue factor pathway inhibitor 2; VHL, von Hippel Lindau tumor suppressor; RARRES1, retinoic acid receptor responder 1; TGF, transforming growth factor; DLL1, Delta-like 1; BCL-2, B-cell CLL/lymphoma 2; FABP4, fatty acid binding protein 4; ROS, reactive oxygen species; Bcl-xl, B-cell lymphoma-extra large; YAP1, Yes-associated protein 1; FOXO3, Forkhead-box O3; IGFBP5, insulin-like growth factor-binding protein 5; IGF, insulin-like growth factor; PAX3, Paired Box 3; YWHAQ, Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide; ERO1α, endoplasmic reticulum oxidase 1α; SERPINA3, serpin peptidase inhibitor clade A member 3; CMIP, C-Maf inducing protein; CTGF, connective tissue growth factor

Insulin-like growth factor 2 (IGF2), which is maternally imprinted and paternally expressed, plays a role in promoting placental growth and transport capacity by regulating the fetus's placental supply and the genetic demand for maternal nutrition [67, 68]. Berg et al. found that patients with EOPE exhibited altered DNA methylation of the IGF2 DMRs in their placental tissue, umbilical cord leukocytes, and vein endothelial cells, but not in those with late-onset preeclampsia (LOPE) [69]. He et al. discovered that the DNA methylation levels of IGF2 DMRs were significantly lower in the umbilical cord blood of infants born to PE. This suggests that reduced IGF2 DMRs methylation is associated with maternal PE and health problems in later life [70]. Furthermore, the average DNA methylation level at IGF2 DMRs was significantly correlated with PE, even after adjusting for birth weight, maternal age, gestational age at delivery, and fetal gender [70]. Insulin-like growth factor-binding protein 5 (IGFBP5) belongs to the IGFBP family of proteins, inhibits the migration of trophoblast cells induced by insulin-like growth factor-1 (IGF1) and IGF2 [71]. Jia et al. identified IGFBP5 as a critical downstream factor of DNMT3A. They discovered that downregulating DNMT3A in the several trophoblasts cell lines increases IGFBP5 expression by decreasing its promoter methylation, consequently suppressing trophoblast cell migration and invasion [72]. IGFBP5 is a secreted protein, and as such, it makes it a promising biomarker. Ma et al. reported that the IGF1 gene promoter is highly methylated in PE placentas, and these placentas have much lower levels of IGF1, moreover, PE is associated with hypermethylation of IGF1 promoter mediated by DNMT1[73].

The H19 exhibits high expression in invasive intermediate trophoblast cells and villous CTB, but its expression is barely detectable in STB [74, 75]. The variable expression pattern of H19 is consistent with its role in controlling trophoblast invasiveness, providing further evidence that the H19 gene is essential for early placental development. Gao et al. found that the overall DNA methylation levels within the H19 gene were considerably higher in EOPE placentas. Meanwhile, hypermethylation in the H19 gene promoter region was correlated with lower expression of H19 gene [65]. H19 exon 1 represents the longest functional nuclear portion of the H19 gene. Placental tissues from women with SPE exhibited lower DNA methylation levels in H19 exon 1[76]. After treatment with the methylation inhibitor 5-Aza-Dc, trophoblast methylation levels significantly decreased, H19 gene mRNA levels increased, and trophoblast cell proliferation, migration, and invasion were dramatically slowed down [76]. Additionally, the incidence of PE was lower in individuals with suppressed H19 mRNA expression due to promoter hypermethylation [77]. Recent clinical trials have examined the effectiveness of metformin in the treatment and prevention of PE [78]. These trials have shown that metformin can affect H19 gene DNA methylation, resulting in changes in H19 expression. The therapeutic benefit of metformin in PE is associated with a dose-dependent increase in H19 methylation and suppression of H19 expression. These studies collectively suggest that hypermethylation at specific CpG sites within the H19 gene may contribute to trophoblast dysfunction and the pathogenesis of PE. However, further extensive and carefully designed investigations in diverse racial and ethnic groups are necessary to validate these findings.

As a member of an imprinted gene cluster, tissue factor pathway inhibitor 2 (TFPI2) acts as an inhibitor of Kunitz-type serine proteinases. In PE placentas, TFPI2 expression is upregulated, which hinders proper villous interstitial remodeling, decidualization, and placental formation [79, 80]. Xiao et al. suggested that the epigenetic mechanism of TFPI2 may be involved in the pathogenesis of PE, and they found significantly lower levels of DNA methylation of the TFPI2 gene promoter in PE placentas compared to controls [80].

In summary, aberrant DNA methylation in imprinted genes such as H19, IGF2, and TFPI2 is associated with an increased risk of PIH. Disruptions in imprinted gene expression can result in phenotypic disorders in humans, as they play a crucial role in regulating growth and development during the embryonic and postnatal stages. Nonetheless, several unanswered questions remain, including the interaction between additional paternal or maternal imprinted genes and DNA methylation as well as their impact on trophoblast cells and placental function.

DNA methylation and tumor suppressor genes

The formation of the placenta, which is unique to mammals, can recapitulate many cancer features. The processes of placentation and tumorigenesis involve similar physiological events, such as rapid cell proliferation and invasion of surrounding tissues. The invasive behavior of cancer cells and placental trophoblast cells, as well as the role of aberrant DNA methylation, are well-known resemblances. The contribution of tumor suppressor genes in cancer is well-documented. Many tumor suppressor genes are expressed in the placenta, and also play a crucial role in placentation and the maintenance of trophoblast functions (Fig. 3).

During pregnancy, the tumor suppressor gene maspin plays a crucial role in controlling trophoblast invasion by being variably expressed in the human placenta. The presence of recombinant maspin greatly reduces the invasion of trophoblasts [81]. Increased maspin expression was observed in the PE placentas when DNA methylation levels in the maspin gene promoter region were low, suggesting that DNA methylation plays a role in the regulation of maspin expression and affects trophoblast invasion, contributing to the pathogenesis of PE [82]. Normal placental development depends on the von Hippel Lindau tumor suppressor (VHL) protein, which is under-expressed in EOPE placentas [83]. Alahari et al. shown that reduced methylation in the CpG-rich area of the VHL gene promoter facilitates VHL binding to E2F4, a transcriptional inhibitor, thereby inhibiting VHL transcription. Subsequently, the absence of VHL leads to an increase in hypoxia inducible factor (HIF) 1A, which disrupts the oxygen homeostasis characteristic of EOPE placentas [83]. This study provides the first evidence that promoter hypomethylation of the VHL gene at a crucial transcription factor binding site may be involved in the pathogenesis of EOPE. Previously, retinoic acid receptor responder 1 (RARRES1) was considered a tumor suppressor gene that is downregulated in cancer cells and promotes apoptosis [84, 85]. However, RARRES1 is under-expressed in most placental cells, while its expression is upregulated in CTB and STB cells due to promoter demethylation. The increase in RARRES1 in PE placentas may be attributed to disturbed trophoblast development, and RARRES1 may impede the invasion and migration of terminal placental EVTs [86, 87].

TP53 has been found to have implications for the life cycle of placental trophoblast cells. There is a 5.6-fold increase in the mRNA expression of the TP53 gene in PE placentas compared to control placentas. For the correlation, there is a strong trend toward a higher expression in methylated conditions (p = 0.06) [88]. Another tumor suppressor gene, bladder cancer-associated protein (BLCAP), has been found to be significantly expressed in the placenta of women with perinatal ectopic pregnancies. Interestingly, the promoter methylation level of BLCAP is significantly lowered in the PE placentas [16]. However, it is still unclear whether DNA methylation-mediated BLCAP expression has any effect on the progression of PE, and further investigation is needed in this area.

In the placenta, DNA methylation changes in tumor suppressor genes may impact trophoblast cell proliferation, apoptosis, migration, invasion, and adhesion, ultimately leading to a wide range of clinical symptoms associated with PIH. There are still many tumor suppressor genes that have not been thoroughly explored, especially in relation to DNA methylation in the context of PIH pregnancy.

DNA methylation and placental classical signaling pathways

The Wnt2 protein is a glycoprotein that interacts with the human brown fly receptor to activate the Wnt/β-catenin signaling pathway, resulting in the expression of target genes. In Wnt2 knockout mice, reduced placental vascularization and structural defects lead to increased fetal mortality due to trophoblastic invasion of the spiral arteries [89]. Methylation of the Wnt2 gene promoter has been observed in SPE placentas, which is associated with decreased Wnt2 gene expression [90, 91]. Liu et al. found a direct correlation between increased methylation of the Wnt2 gene promoter and decreased Wnt2 gene expression. They also demonstrated that DNA methylation plays a role in the regulation of Wnt2 gene expression, leading to trophoblast dysfunction, which in turn affects trophoblast invagination and helical artery remodeling, and thus is involved in the pathophysiology of PE [92]. Wnt3, which plays a crucial role in regulating decidualization, stromal cell proliferation, and trophoblast invasion, has been found to be hypomethylated in its gene promoter in PE placentas. This leads to increased expression that compensates for the low activation status of the Wnt/β-catenin signaling pathway [93].

Additionally, the transforming growth factor β (TGF-β) pathway plays a role in maintaining placental functions and establishing pregnancy. The TGF-β superfamily transmits signals by forming heteromeric complexes of type I and type II transmembrane serine/threonine kinase receptors [94]. Activation of TGF-β receptor 1 (TGFBR1) leads to the phosphorylation of receptor-specific Smad proteins, namely Smad2 and Smad3 [95]. TGF-β pathway exhibits extensive promoter hypomethylation in the PE placentas. This hypomethylation is accompanied by an enrichment of TGF-β pathway-related genes, including SMAD family member 3 (SMAD3), v-ski sarcoma viral oncogene homolog (SKI), runt-related transcription factor 3 (RUNX3), and TGFB-induced factor homeobox 1 (TGIF1) [96]. In the placenta of women with early-onset severe PE (EOSPE), trophoblast cells exhibit elevated levels of TGFBR1 due to downregulation of DNMT3A. Restoring normal trophoblast migration and invasion can be achieved by inhibiting TGFBR1 and TGF-β/Smad signaling. This indicates that DNA hypomethylation-mediated upregulation of the TGFBR1 gene is associated with the development of EOSPE by inhibiting trophoblast migration and invasion [72, 96].

The receptor-ligand Notch signaling family regulates various cellular processes, such as proliferation, apoptosis, invasion, and adhesion. Dysregulation of the Notch signaling pathway has been observed in PE placentas [97, 98]. Shimanuki et al. analyzed the Delta-like 1 (DLL1) gene, which is the Notch activating ligand, in PE placentas. They discovered a higher frequency of DLL1 gene DNA methylation and significantly lower DLL1 expressions in PE placentas[98]. Their analysis also revealed that EOSPE placentas with DLL1-DNA methylation exhibited an increase in syncytial knots and accelerated villous maturation [98]. These findings suggest that altered Notch signaling through DNA methylation may contribute to the pathogenesis of PE.

In conclusion, the Wnt/β-catenin signaling and TGF-β pathways are the most reported signaling pathways involved in the development of PIH. The Notch signaling pathway have also been reported to be associated with PIH (Fig. 3). These findings significantly enhance our understanding of the role of DNA methylation in the pathophysiology of PIH and suggest a potential novel therapeutic approach targeting DNA methylation for this disorder.

DNA methylation and placental MicroRNAs

Numerous studies have demonstrated the crucial role of miRNAs in the regulation of placental development and pathology [99, 100]. Previous research has indicated that PE placentas exhibit distinct miRNA expression profiles compared to normal placentas. Differentially expressed miRNAs are characteristic of mild and SPE. In many cases, these miRNAs often target key classical signaling pathways that control trophoblast proliferation and invasion, which are directly implicated in PIH [99, 100]. MiRNAs have also been recognized as potential therapeutic targets and diagnostic tools for PIH.

For instance, miR-34a has been found to regulate somatic cell reprogramming, cell cycle, apoptosis, and metastasis [101]. One study showed that PE placentas exhibits higher expression of pri-miR-34a due to promoter hypomethylation [102]. On the other hand, Guo et al. found significantly higher levels of miR-34a in the PE placentas, and demonstrated that upregulation of miR-34a induces trophoblast apoptosis in PE through inhibition of B-cell CLL/lymphoma 2 (BCL-2) protein expression [103]. Rezaei et al. further elucidated that hypomethylation of the miR-34a promoter increased susceptibility to PE, suggesting that miR-34a may be a target gene for PE development [104].

MiR-29b has been implicated in several studies on PE, where its presence has been found to decrease trophoblast cell proliferation and migration [105, 106]. It has been discovered that placental hypomethylation controls the induction of placental reactive oxygen species (ROS) production by upregulating the expression of miR-29b [107]. The Let-7 family of miRNAs exhibits strong sequence and functional conservation. Zha et al. indicates that let-7a is hypomethylated in EOSPE placentas and let-7a is highly expressed, leading to the downregulation of B-cell lymphoma-extra large (Bcl-xl) and Yes-associated protein 1 (YAP1), which decreases trophoblast invasion while promoting cell death [108]. Thus, elevated let-7a expression causes JEG-3 cells to become less viable, halts the cell cycle, and induces apoptosis [108, 109]. Luo et al. found that miR-155 showed significant up-regulation and an inverse correlation with promoter methylation levels in the PE placenta. MiR-155 was found to downregulate Forkhead-box O3 (FOXO3), which in turn increased apoptosis and decreased trophoblast cell viability, migration, and invasion in in vitro experiments [110].

Researchers compiled studies on placental miRNAs and their role in the pathogenesis of PE in a rodent PE animal model [111, 112]. They found that the expression of miR-148/152 family miRNAs was upregulated in the PE placenta. This finding was verified in pregnant rats in the PE model, and Yang et al. also discovered that the downregulation of DNMT1 was accompanied by an overall increase in miR-148a/152 expression, resulting in increased expression of fatty acid binding protein 4 (FABP4) [112]. This discovery was further confirmed in HTR8 cells, where overexpression of FABP4 led to upregulation of the pro-inflammatory cytokines IL-6 and TNF-a, as well as an increase in lipid accumulation. Both in vitro and in vivo models of PE have demonstrated the involvement of miR-148a/152-FABP4 in the development of PE [112, 113].

In summary, miRNAs such as miR-34a, miR-29b, let-7a, miR-155, and miR-148a/152 have an impact on the proliferation, migration, and invasion of trophoblast cells, ultimately leading to the initiation and development of PIH (Fig. 3). Considering PIH's heterogeneity, future research efforts should focus on identifying changes in placental miRNA profiles within patient subgroups or clarifying abnormal blood and placental miRNA expression profiles among different subsets of patients with placental equivalent diseases.

DNA methylation and other placental genes

In the above section, we systematically summarized the latest research progress on DNA methylation involved in regulating imprinted genes, tumor suppressor genes, classical pathway-related genes, and miRNAs expression in the placenta. In addition to these genes, current research has found that DNA methylation also participates in regulating the expression of other genes in the placenta and plays an important role in the occurrence of PIH.

Activated leukocyte cell adhesion molecule (ALCAM) is an immunoglobulin cell adhesion molecule that is overexpressed in human endometrial epithelial cells and blastocysts. The stage-specific expression during embryonic development suggests that ALCAM plays a role in the initial interaction between the embryo and the maternal endometrium [114]. A decrease in ALCAM expression and an increase in DNA methylation within the ALCAM gene promoter in PE placentas, meanwhile, there was a negative correlation between ALCAM promoter methylation and its expression [91, 115]. These findings suggest that alterations in DNA methylation-mediated ALCAM expression may contribute to the development of PE by reducing trophoblast vitality and invasiveness. This is because ALCAM expression is typically observed in proliferating cells and is crucial for blastocyst implantation, thereby enhancing maternal–fetal communication [116]. During embryonic development, PAX3 (Paired Box 3) acts as a transcription factor that influences the growth of placenta. Patients with PE have a higher methylation level of the PAX3 promoter and drastically reduced levels of PAX3 in their placentas. The decreased expression of PAX3 ultimately suppresses the proliferation and invasion abilities of trophoblast cells [117].

Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide (YWHAQ) is highly expressed in placental trophoblast cells and promotes trophoblast differentiation by controlling cell cycle progression [118]. Liu et al. reported that significant hypermethylation of the YWHAQ promoter in SPE placentas resulted in decreased YWHAQ and increased apoptosis of placental trophoblast cells, thereby resulting in increased levels of placental cell debris flowing into the maternal circulation [119]. This observation suggests that YWHAQ may be involved in the molecular mechanisms underlying the pathologic process of PE, but further evaluation is needed. Abnormal trophoblast cell apoptosis is implicated in the pathogenesis of PE, and endoplasmic reticulum (ER) stress is involved in the regulation of trophoblast cell apoptosis. ER oxidase 1α (ERO1α) is a glycosylated flavoenzyme that plays an important role in maintaining ER redox homeostasis. Xiong et al. reported that ERO1α expression is suppressed in trophoblast cells in the placenta of PE rats due to hypermethylation of the ERO1α promoter region [120]. These results demonstrate that promoter hypermethylation of ERO1α leads to trophoblast cell apoptosis through ER stress in the PE placentas, shedding light on the etiology of PE and potentially providing a therapeutic target for its treatment. One of the most critical steps in fertilization is the fusion of the egg plasma membrane, which is facilitated by sperm equatorial segment protein 1 (SPESP1). Yeung and Meng et al. found significant hypermethylation of the SPESP1 gene promoter as well as down-regulation of SPESP1 expression in PE placentas [91].

SERPINA3 (Serpin peptidase inhibitor clade A member 3), a serine protease inhibitor, is up-regulated in PIH placentas, which is associated with hypomethylation of the 5' region of the SERPINA3 gene [121, 122]. Overexpression of SERPINA3 in JEG-3 cells has been found to decrease cell adhesion to the extracellular matrix and neighboring cells [122]. C-Maf inducing protein (CMIP) has oncogenic properties and is expressed in a variety of malignant tumors. It was found that in PE placentas, the methylation level of the CMIP promoter was significantly reduced, and the expression level of CMIP was negatively correlated with the methylation level, which promotes trophoblast apoptosis and plays a role in the pathogenesis of PE. In addition, connective tissue growth factor (CTGF), a stroma-associated cellular protein, is a multifunctional signaling regulator involved in angiogenesis. In a study of a Chinese Han population, it was found that decreased promoter methylation resulted in increased expression of CTGF in SPE placentas, and the enhanced CTGF expression contributes to the regulation of trophoblast differentiation [123].

Aquaporin 1 (AQP1) is one of the most widely expressed water channels and plays an important role in regulating maternal–fetal fluid exchange and amniotic fluid volume [124]. It was found that AQP1 levels were elevated in PE placentas, which was associated with reduced DNA methylation in its promoter and significantly negatively correlated with the amniotic fluid index[125]. Elevated AQP1 may disrupt maternal–fetal body fluid homeostasis in PE pregnancies. In addition, guanine nucleotide binding protein (G protein) alpha 12 (GNA12) is an important regulatory molecule for signaling through transmembrane receptors. Study has shown that GNA12 expression is increased in PE placentas and hypomethylation is observed at three CpG sites of the GNA12 gene promoter [126, 127]. Death-domain associated protein (DAXX) is a histone chaperone protein that undergoes methylation changes in the DMR within the DAXX gene during CTB syncytization to STB and differentiation of in vitro cellular trophoblasts to invasive trophoblast cells. Novakovic et al. observed that loss of methylation in this DMR is associated with decreased DAXX expression [128]. However, this finding contradicts the results from the published EOPE comprehensive library of gene expression of placental villus methylation (GSE44712) [13], which states that DAXX hypomethylation leads to higher DAXX expression. Further research is needed to determine the specific role of differential DAXX expression in PE.

It is evident that trophoblast cells express a variety of proteins, and the abnormal expression of these proteins, regulated by DNA methylation, can result in abnormalities in trophoblast cell proliferation, differentiation, apoptosis, and invasion. These abnormalities may contribute to impaired placental function and the progression of PIH (Fig. 3).

DNA methylation and placental vascular system

In the previous section, we provided a summary of the research progress on DNA methylation and its regulation of gene expression in trophoblast cells in the context of the PIH placenta. As a vascular organ, the placenta is responsible for supplying blood to both the mother and the developing baby. Therefore, it is essential for the placenta to receive an adequate blood supply. Currently, it is widely accepted that defects in trophoblast cell invasion and spiral artery remodeling during the early stages of pregnancy are the main pathological causes of placental ischemia and the initiation of PIH. However, abnormalities in the development and vascular tone of placental vessels also contribute to insufficient blood supply to the placenta, ultimately leading to placental ischemia and dysfunction. In this section, we aim to summarize the current knowledge and provide a comprehensive evaluation of aberrant DNA methylation of genes associated with vascular development and function in PIH placentas (Table 1).

DNA methylation and placental angiogenesis, vascular remodeling

PIH is a placental vascular pathology, and hypoxia is known to influence placental angiogenesis. The oxygen-sensitive transcriptional activator HIF1 is a key mediator of the transcriptional response to hypoxic conditions. The HIF1 pathway has been identified as a master regulator of angiogenesis and is involved in vasculature formation through synergistic interactions with other proangiogenic factors such as vascular endothelial growth factor (VEGF), placental growth factor (PlGF), and angiopoietins [129]. Recently, Kaur et al. found that placental DNA methylation and expression of HIF1α and 3α genes are altered and associated with PIH, and birth outcomes [130]. Furthermore, DNA methylation in the promoter region of HIF1α was found to be negatively correlated with its expression levels in PIH placentas. Considering the significant role of HIF1 in angiogenesis and vasculogenesis, it should be considered a promising target for the treatment of PIH. VEGF is a crucial mediator of angiogenesis in both physiological and pathological processes. Sundrani et al. discovered that the methylation of the VEGF gene promoter was significantly reduced in early preterm PE placentas, while its expression was significantly increased. Disruptions in the levels of VEGF and its receptor can impair angiogenesis, resulting in placental insufficiency and endothelial dysfunction [131]. This finding highlights the significance of altered DNA methylation in placental angiogenesis and adverse pregnancy outcomes. Factors related to angiogenesis, such as soluble FMS-like tyrosine kinase-1 (sFlt-1) and PlGF, play significant roles in placental angiogenesis and vasculogenesis. Dave et al. observed hypomethylation at several CpG sites in the promoters of PlGF and Flt-1 genes in PE placentas. DNA methylation at different CpG sites was correlated with the two genes expression and was associated with maternal blood pressure [132].

Extracellular matrix (ECM) plays a crucial role in vascular remodeling during pregnancy. This process of trophoblasts invading the endometrial stroma, forming the myometrium and the deep layer of the placenta, is essential for fetal development [133]. Matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTs) are all examples of MMPs that are secreted from placental cells during this process [134, 135]. MMPs and their tissue inhibitors (TIMPs) play crucial roles in the trophoblast's proper invasion into maternal tissue. An imbalanced expression of MMPs and TIMPs impairs proper placentation, and DNA methylation may affect their expressions [136, 137]. The aberrant expressions of these proteins are tightly regulated by DNA methylation, which can impair the ECM remodeling process during pregnancy. In PE placentas, several genes, including MMP9, TIMP3, and ADAMTS7, were significantly less methylated within the gene promoter. Abnormal expression of MMP9 can increase the risk of developing PE by preventing adhesion molecule-mediated invasion of the EVTs [138]. Wang et al. found significantly higher expression levels of MMP9 in PE placentas, and this increased expression was well correlated with promoter demethylation [136].

TIMPs are present in a 1:1 stoichiometric ratio with MMPs in tissues, where they locally and temporarily suppress MMP activity [139]. TIMP3 is abundantly expressed in the placenta and plays a critical role in controlling placental vascular remodeling during placentation [139]. Yuen et al. confirmed significant hypomethylation of the TIMP3 promoter in EOPE placentas [140]. Xiang et al. discovered an inverse correlation between hypomethylation of the TIMP3 promoter and TIMP3 expression in PE placentas [141]. Similarly, Cruz et al. found hypomethylation of the TIMP3 gene promoter, as well as increased TIMP3 expression in corresponding placental samples from PE, EOPE, and LOPE compared to controls [142]. These findings demonstrate the significant role of DNA methylation in controlling TIMP3 expression. The hypomethylation of the TIMP3 promoter increase TIMP3 expression, leading to decreased trophoblast invasiveness during placental development and contributing to low placental perfusion in PE. Therefore, TIMP3 may serve as a prenatal marker for the development of PE. Additionally, abnormal expressions of some members of the ADAMTS family have been associated with PE progression [143]. Zhang et al. discovered that hypomethylation of the ADAMTS7 gene promoter resulted in higher expression of ADAMTS7 in PE placentas [144]. These findings suggest a role for ADAMTS7 in the pathophysiology of PE, as it can inhibit the migration, invasion, and survival of trophoblast cells in vitro, leading to inadequate repair and blood flow in the placental vascular spiral artery.

DNA methylation and placental vascular function

After placentation in early pregnancy, adequate blood flow in the placenta is vital for maintaining placental function. The blood arteries in the placenta lack their own neural innervation. Therefore, local synthesis of vasoactive chemicals and vascular regulators are essential for regulating vascular activity, function, and blood flow within the placenta [145, 146].

Arginine vasopressin (AVP), a traditional vasoconstrictor, controls vascular tone by binding to its receptors (primarily V1a (AVPR1A), V1b (AVPR1B), and V2 (AVPR2) subtypes) in vascular smooth muscle cells (SMCs). Gao et al. were the first to study the role of AVP in the placental vascular system and discovered that placental vessels had a diminished sensitivity to AVP-induced constriction in PE placentas. They also observed downregulation of AVPR1A and protein kinase C isoform β (PKCβ), as well as a dramatic increase in promoter DNA methylation within the two genes in the PE placental vessels [147]. This study revealed that aberrant DNA methylation-mediated gene expressions are correlated with vascular dysfunction in the PE placental circulation. Oxytocin (OXT), another traditional vasoconstrictor, is predominantly known as a potent stimulator of uterine smooth muscle and plays an important role in maintaining placental circulation [148]. The actions of OXT are mediated through its receptor (OXTR). In the context of pregnancy, OXT and OXTR are abundant in the human placenta. Oxytocin is increased in the placental system during late pregnancy and the onset of labor, and it exhibits concentration-dependent contractions in the placental vascular system [149]. DNA hypermethylation of CpG islands in the OXTR gene promoter leads to the transcriptional inactivation of the OXTR gene. This is why Fan et al. found that OXT-induced vasoconstriction was significantly reduced in the PE placental vessels [150].

Additionally, ROS can regulate vascular constriction and relaxation. NADPH oxidases (NOX) catalyze the production of ROS and regulate redox-dependent processes in various biological contexts. Recently, Yeung et al. reported that DNA hypermethylation within the promoter of the NOX5 gene were observed in the PE placentas [91]. Given the importance of the NOX/ROS pathway in regulating vascular tone, this finding suggests that DNA methylation of the NOX5 gene promoter may play a role in placental vascular dysfunction in PE placentas. As an important vascular mediator, the large-conductance Ca2+-activated K+ channel (BK) is a tetramer composed of a pore-forming alpha subunit (BKα) and up to four accessory beta subunits (BKβ). BK plays a crucial role in determining vascular tone. Chen et al. reported that placental hypoxia/ischemia during pregnancy inhibits pregnancy-induced demethylation of the BKβ1 gene and upregulation of BKβ1 expression in uterine arteries. They presented new insights into the role of DNA methylation in the association of gestational hypoxia/ischemia with aberrant uteroplacental circulation and an increased risk of PE [151].

In summary, whether it is angiogenesis and vascular remodeling during placentation or abnormal regulation of placental vascular function during mid-to-late pregnancy, it will affect placental function and lead to placental-related diseases such as PIH. Despite the aforementioned research (Table 1), there is currently a lack of data on DNA methylation of genes related to the placental vascular system in PIH placentas. Further in-depth research is urgently needed on the roles of DNA methylation in placental vascular angiogenesis, remodeling, and function regulation, as well as its role in the occurrence of PIH.

DNA methylation changes in PIH maternal blood

Researchers were able to identify a high number of DMRs that could be used as early diagnostic markers for PIH using maternal peripheral blood DNA [152, 153]. For example, three neuronal genes—GABRA1, BEX1, and GRIN2B—were significantly differentially methylated and expressed at low levels in the maternal blood, suggesting a correlation with seizure episodes of PIH patients, as determined by whole-genome methylation analysis [153]. Currently, whole-genome bisulfite sequencing was used on maternal blood to elucidate the methylation pattern associated with PIH, leading to the identification of new candidate risk genes. Additionally, these potential markers may have application in the early diagnosis of PE (Table 2).

Table 2 DNA Methylation Changes in PIH Maternal Blood

DNA methylation and maternal plasma

Pregnancy-related problems are associated with cell-free DNA (cfDNA) and cell-free fetal DNA (cffDNA) in maternal plasma [154, 155]. Total cfDNA levels are significantly higher in late pregnancy for pregnant women with PE [156]. This discovery opens the door for the analysis of cfDNA in maternal plasma to be used in clinical settings for the detection and monitoring of PE.

Recent research has proposed the use of cfDNA methylation and transcriptome indicators in early pregnancy to predict the onset of PE before the manifestation of clinical symptoms [157]. For example, RAS association domain family 1A (RASSF1A) DNA sequences were detected in the mother's blood; the gene was silenced and frequently inactivated by hypermethylation of the promoter region; and reduced RASSF1A expression was positively correlated with the severity of PE [158,159,160]. Rahat et al. concluded that there is an association of DNA methylation of the RASSF1A promoter and RASSF1A expression in PE maternal plasma, and the increased RASSF1A expression contributes to the development of PE [161]. This finding suggests that RASSF1A could be utilized as a non-invasive diagnostic tool for PE. Building upon these findings, Salvianti et al. prospectively assessed RASSF1A cfDNA in maternal plasma during pregnancy, specifically examining the methylation status of the promoter gene sequence. They found that total cfDNA and cffDNA were sufficiently predictive of PE in high-risk women compared with non-PE high-risk women between 17 and 30 weeks of gestation, and that cfDNA and cffDNA could be quantified with hypermethylated RASSF1A, suggesting that hypermethylated RASSF1A could provide evidence as a reliable marker for PE [162]. Apart from RASSF1A, DSCR3 (Down syndrome critical region gene 3) may also serve as an epigenetic marker for hypermethylated cffDNA. After 24–32 weeks, the concentration of cffDNA (DSCR3 and RASSF1A) significantly increased in patients with EOPE [163]. However, there are few current studies on DSCR3, and it is hoped that more subsequent studies will delve deeper into DSCR3 to determine whether it can be a biomarker.

Maspin is a well-known tumor suppressor gene. In PE plasma samples, the level of its promoter methylation exhibits further downregulation [164]. Unmethylated maspin DNA levels in the blood are 5.5 times higher in pregnant women with SPE compared to normal pregnant women in the third trimester. However, there is no statistically significant difference between moderate PE patients and normal pregnant women [165]. The methylation pattern of the TIMP3 promoter is similar to that of maspin. It is moderately methylated in normal placental tissue and almost completely methylated in maternal blood DNA. The level of unmethylated TIMP3 sequence is significantly higher in PE maternal plasma. This is consistent with the lower methylation level of the TIMP3 promoter in PE placentas [141]. Yuen et al. reported that gene-specific hypomethylation may be a common phenomenon in EOPE plasma [140]. Therefore, the alteration of unmethylated TIMP3 sequences in maternal plasma can be utilized to predict PE. An increase in the amount of unmethylated TIMP3 cfDNA was taken into account, which could improve the sensitivity of non-invasive screening for PE pregnancies.

STAT5A (signal transducer and activator of transcription 5A) is a transcription factor with well-known anti-apoptotic role. Its expression level in placental villi is highest in the first trimester and decreases over the remainder of pregnancy [166]. The pattern of promoter methylation of STAT5A in maternal plasma cfDNA in pregnant women with PE is consistent with that in placental villi, which are elevated. This suggests that it is a useful epigenetic marker in women with PE [166].Ye et al. investigated the potential association between the methylation of the GNA12 promoter and PE [127]. The methylation level at three CpG sites of the GNA12 promoter was significantly lower in both the PE placental and peripheral blood DNA samples. Consistent with the decreased methylation level, the expression level of GNA12 was higher in PE patients. These results showed that PE is associated with decreased methylation of the GNA12 promoter, which can be used as an early diagnostic biomarker of PE [127]. In addition, higher CTGF expression in trophoblast cells is associated with a reduced frequency of CTGF promoter methylation, which is observed in the peripheral blood of PE pregnant women compared to controls [123]. Evidence from the Han Chinese population indicates a link between PE and poor methylation of the CTGF promoter in the peripheral blood [123]. Nonetheless, more evidence is needed to establish CTGF as a gene factor appropriate for wide-scale application in early PE detection, as the sample size was very small.

Although it has been found that abnormal DNA methylation of genes in cfDNA and cffDNA can be used as markers for the prenatal diagnosis of PIH, more rigorous research is required to confirm the presence of cfDNA and cffDNA changes in the plasma of PE patients and establish a correlation between these changes and the placenta and maternal circulation. Furthermore, further investigation into DNA methylation is necessary to identify molecules that are abnormally produced in maternal plasma and determine their role in the etiology of PE.

DNA methylation and maternal vascular function

Among the hallmarks of PE is vascular dysfunction in the mother. In PE maternal vessels, 65 genes are found to be hypomethylated, and there is evidence of overexpression of 75 molecular functions and 149 biological processes. These processes and functions include those involved in smooth muscle contraction, thrombosis, inflammation, redox homeostasis, and amino acid metabolism [167]. The most recently documented maternal vascular phenotypes in PE are the overexpression of MMPs in endothelial cells, vascular SMCs, and invading neutrophils [168, 169]. In PE patients, the great omental artery has been found to have significantly lower levels of methylation in the promoter regions of the MMP1 and MMP8 genes in neutrophils, which is strongly correlated with elevated expression [170]. As mentioned earlier, overexpressed MMP1 exerts its many biological functions through protease-activated receptor-1 (PAR1). Since PAR1 is only expressed during pregnancy, its activation leads to the translocation of TET2 and nuclear factor-kappa B (NF-κB) from the cytoplasm to the nucleus in neutrophils, resulting in increased expression of inflammation-related genes and a more rapid progression of PE [171].

A decrease in prostacyclin, a vasodilator and platelet inhibitor, and an increase in thromboxane, an effective vasoconstrictor and platelet activator, have been reported in the PE placentas. These changes have subsequently been confirmed in maternal blood and urine [172, 173]. Thromboxane A2 is produced by thromboxane A synthase 1 (TBXAS1), which is responsible for the isomerization of prostaglandin H2 (a precursor of both thromboxane and prostacyclin). Mousa et al. confirmed a strong correlation between decreased DNA methylation in the TBXAS1 gene promoter region and increased TBXAS1 expression in the great omental arteries of women with PE [174]. Increased TBXAS1 expression was also observed in maternal endothelial cells, vascular SMCs, invading neutrophils, and white blood cells of women with PE [174]. Furthermore, hypertension and chronic inflammatory illnesses are also linked to the cytokine family, which is a potent inflammatory agent [175]. Walsh et al. found significantly reduced DNA methylation of the interleukin-17 (IL17) gene promoter in omental arteries of PE women and demonstrated that IL17 levels are regulated by DNA methylation, and increases in the PE circulation [176].

In summary, these investigations demonstrate that DNA methylation is important in regulating maternal vascular function and contributes to the development of PIH. Clinical diagnosis and therapy of PIH are aided by the information provided by these studies, and the pathophysiology of this condition is better understood.

DNA methylation changes in PIH umbilical cord

DNA methylation in umbilical cord blood

Currently, only a small number of studies have investigated the variations in local or global DNA methylation in umbilical cord blood cells of newborns with PIH [177, 178]. Kazmi et al. detected the levels of global DNA methylation in cord blood among neonates with PIH and PE and found that the epigenome-wide correlation of PIH with offspring DNA methylation was moderately consistent with the equivalent epigenome-wide correlation of PE with offspring DNA methylation [178]. This demonstrates the presence of numerous DNA methylation markers in cord blood in cases of PIH and PE. Ching et al. initially examined DNA methylation throughout the entire genome in cord blood from neonates with EOPE and indicated that the DNA methylation pattern of a considerable number of genes changed in EOPE cord blood [179]. They found that promoters of 643 and 389 genes were hypomethylated and hypermethylated, respectively. Genes in the farnesoid X receptor and liver X receptor (FXR/LXR) pathways were enriched, indicating dysfunction of lipid metabolism in EOPE cord blood cells. Additionally, a group of genes involved in inflammation, lipid metabolism, and proliferation were persistently differentially methylated in EOPE cord blood cells [179]. These findings provide evidence of prominent DNA methylation modifications in cord blood DNAs associated with EOPE and suggest a connection between inflammation and lipid dysregulation in EOPE-associated newborns and a higher risk of cardiovascular diseases later in adulthood.

Leptin is a hormone that plays an important role in maintaining healthy cardiac metabolism and stable energy levels. Study has shown that in the cord blood of pregnant women with PE, there is reduced methylation of the leptin gene at the CpG14 locus and increased leptin level, which negatively affected maternal–fetal nutrient exchange and placental function [180]. HIF3A is a member of the HIF gene family and is thought to play an important role in vascular development, and metabolism. Mansell et al. first found that PE was associated with increased HIF3A expression due to hypomethylation of the HIF3A gene promoter in umbilical cord blood, which was consistent with hypoxia-induced up-regulation of HIF3A transcription [181]. However, further studies are needed to determine the underlying mechanisms.

Mitochondria are the energy factories of the cell; therefore, it is necessary to study the expression and activity of mitochondrial genes in cord blood cells. Novielli et al. first characterized mitochondrial DNA in cord blood from PE and found that DNA methylation levels of the D-loop (mitochondrial loci relevant to replication) gene were decreased and its expression increased in PE cases [182]. Low D-loop methylation levels are associated with poorer fetal prognosis, and this association is particularly evident in PE cases, where D-loop methylation levels are further reduced and strongly positively correlated with fetal weight and umbilical vein pO2 [182]. In the aforementioned placenta chapter, it is mentioned that the expression and activity of 11βHSD2 are reduced in the PE placenta, leading to increased exposure of the fetus to glucocorticoids. Infants born to mothers with PE have been found to have decreased levels of 11βHSD2 promoter methylation in cord blood, indicating a favorable association between 11βHSD2 promoter methylation and PE, as well as a possible increase in 11βHSD2 expression in PE offspring [183]. More research is required to understand the role that hypomethylation of the 11βHSD2 promoter plays in the progression of diseases in the PE offspring.

In conclusion, these investigations on cord blood have demonstrated the importance of DNA methylation in cord blood and its contribution to the development of PIH (Table 3), which has negative consequences for both the mother and the fetus. The information provided by these studies aids in the clinical diagnosis and therapy of PIH, and enhances our understanding of the pathophysiology of this condition.

Table 3 Changes in DNA methylation in umbilical cord and in offspring of PIH cases

DNA methylation in umbilical cord vessels

Umbilical vein plays a crucial role in the circulation between the placenta and the fetus, as it carries oxygenated and nutrient-rich blood from the placenta to the fetus. 5hmC is produced from 5mC and promotes DNA methylation plasticity through oxidation by the TET enzyme. 5hmC is not only an intermediate modification in the active demethylation process of DNA, but also an important epigenetic marker for neurodevelopment, and embryonic stem cell differentiation [184]. Sun et al. discovered that in patients with PE, both TET2 expression and 5hmC levels were significantly reduced in the umbilical veins [11]. Additionally, a decreased sensitivity to vasoconstriction of AVP and OXT was observed in the umbilical vein of PE patients, which has been linked to hypermethylation of AVPR1a, OXTR, and PKCβ. This is because hypermethylation of these genes leads to a relatively low gene expression pattern in the umbilical vein of PE patients [185].

Endothelial colony-forming cells (ECFCs) have been found to have a decreased number and compromised activities in PE umbilical cord [186]. ECFCs are a relatively homogeneous subset of late-growth endothelial progenitor cells that possess the ability to multiply and migrate to the site of vascular formation, directly contributing to endothelial development [187]. Brodowski et al. conducted a study on the methylation pattern of ECFCs in the umbilical cord of PE cases. They observed a differential methylation pattern of umbilical cord ECFCs in PE compared to normal pregnancies, with 1266 CpG sites showing differences in passage 3 and 2362 sites in passage 5. Moreover, 1260 of the PE-related methylation changes detected in passage 3 were confirmed in passage 5, suggesting that the methylation modifications induced by PE are stable [188]. Further research is necessary to comprehend the causes of DNA methylation alterations in PE umbilical cord blood and vessels, as well as the potential effects of these changes on the health of future generations.

The placenta plays a significant role in the deterioration of the mother's cardiovascular system during pregnancy. Throughout pregnancy, DNA methylation patterns can shift unexpectedly, occurring in the placental, maternal, and fetal sides. Therefore, it is crucial to detect maternal and fetal blood to investigate placental diseases. The availability and utilization of cfDNA and cffDNA in obstetrics have provided opportunities for non-invasive prenatal diagnosis of PIH.

DNA methylation changes in PIH neonates and children

Strong evidence suggests that the timing of development from conception to infancy is influenced by interactions between the genome and the environment, which can have long-lasting effects on offspring [189,190,191]. Conditions such as obesity, hypertension, and poor cardiometabolic markers in children have been associated with these conditions in the mother [191, 192]. Epigenetic imprinting, particularly DNA methylation, occurs during development, allowing the environment to not only influence gene expression but also impact future generations [191, 193]. This implies that DNA methylation play a crucial role in regulating the physiopathology of PIH in both the fetus and postnatal stages (Table 3).

The variations in DNA methylation in the blood, placenta, and umbilical cord of PIH mothers, as discussed earlier, may have effects on the developing fetus. Recent clinical studies in neonates and children have confirmed this. A few years ago, Suzuki et al. conducted DNA methylation analysis of fetal exposure to PE using amniotic membrane samples from delivered placentas [194]. They identified different DMRs between amniotic epithelial cells and mesenchymal stromal cells, confirming that exposure to PE alters fetal DNA methylation. This study also supported the use of amniotic membranes as a substitute for the fetus to address genome-wide DNA methylation. Several cohort studies have reported an increased risk of cardiovascular disease and pulmonary vascular malfunction in adults who were born to mothers with PIH [195, 196]. For example, Julian et al. found that offspring of women with PIH had higher pulmonary artery systolic pressure compared to controls [196]. To determine whether DNA methylation of genes involved in the regulation of vascular function is present in offspring of women with PIH, they compared DNA methylation and gene expression patterns in peripheral blood mononuclear cells from young male offspring of PIH women with normotensive controls [196]. They found that three genes, namely collagen triple helix repeat containing 1 (CTHRC1), AT rich interactive domain 1B (ARID1B), and SPARC related modular calcium binding 2 (SMOC2), had six different DMRs in the offspring of patients with PIH. Among these genes, ARID1B had impaired gene expression, and CTHRC1 tended to be higher. The transcriptional activity of the two genes was inversely proportional to their DNA methylation status [196].

Based on these findings, it is speculated that DNA methylation may play a role in mediating the increased risk of vascular disease later in life associated with PIH. Due to ethical constraints on human research, experimental animals were used to investigate the effects of maternal PIH on gene DNA methylation in multiple generations of offspring and its role in the occurrence of offspring diseases [197, 198]. For instance, Guan et al. [197] found that DNA hypomethylation of the G signaling gene in F1 sperm and F2 thoracic aorta, along with hyperactivation of downstream PI3K-Akt signaling, suggests that DNA methylation plays a crucial role in the development of hypertension in PIH offspring. Since PIH is a "major obstetric syndrome" involving multiple pathological processes that can overlap and ultimately stimulate the same pathway, such as endothelial activation, intravascular inflammation, and syncytial trophoblastic stress, it is important to investigate the link between intergenerational transmission and DNA methylation in a precise PIH animal model.

In conclusion, existing clinical and animal experiments have shown that alterations in DNA methylation, induced by PIH, are linked to unfavorable outcomes in children and offspring following birth. These outcomes include an elevated susceptibility to cardiovascular and metabolic diseases in PIH offspring. However, due to the complex nature of PIH pathogenesis, there is still a significant knowledge gap in this field. Therefore, it is imperative and essential to conduct further research in this area.

Conclusion and outlook

Pregnancy offers a unique opportunity to investigate stress and stress responses. In the case of pathological pregnancies, understanding epigenetic mechanisms, particularly DNA methylation, may assist in identifying new biomarkers associated with disease risk related to pregnancy. These biomarkers could be crucial for developing tools to detect risk factors and exposure levels at an early stage. In this review, we summarize the latest research progress on the role of DNA methylation in the pathophysiology of PIH. This review supports the notion that many genes involved in the initiation and development of PIH, mediated by DNA methylation, could serve as diagnostic markers for the disease. Moreover, addressing DNA methylation may prove to be a valuable therapeutic strategy for PIH.

This review focuses on three major aspects: maternal, fetal, and offspring, and provides a comprehensive account of the relationship between DNA methylation and the trophoblastic function of the placenta, genes related to the placental microenvironment, the placental vascular system, maternal blood and vascular function, abnormalities of umbilical cord blood and vascular function in the genesis and development of PIH, as well as the alteration of DNA methylation in the offspring of PIH. By doing so, our aim is to enhance understanding of the dynamic biological processes underlying the development of PIH. These insights will assist clinicians in recognizing adverse pregnancy outcomes caused by PIH and tailoring interventions to reduce risks, thereby promoting healthy pregnancies for all mothers. However, for most of the current studies, DNA methylation is usually identified in whole tissue/blood and there is some heterogeneity in these samples in terms of cell type, which can be a strong limitation.

The present study provides important information in understanding and exploring the epigenetic mechanisms of PIH pathogenesis. It is clear that changes in DNA methylation may play a crucial role in the development of PIH and that changes in DNA methylation play a key role in the screening, diagnosis and treatment of PIH. However, larger and more robust studies are needed to strengthen the existing understanding, and further research on DNA methylation must continue to provide a comprehensive picture of the epigenetic landscape. This review aims to serve as a catalyst for future investigations into the role of DNA methylation in the development of PIH, and hopes to inspire academics to delve deeper into this topic.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

PIH:

Pregnancy-induced hypertension

PE:

Preeclampsia

SPE:

Severe preeclampsia

EOPE:

Early-onset preeclampsia

LOPE:

Late-onset preeclampsia

EOSPE:

Early-onset severe preeclampsia

DM:

Differential methylation

DMRs:

Differential methylation regions

DNMT:

DNA methyltransferase

CTB:

Cytotrophoblast

STB:

Syncytiotrophoblast

EVT:

Extravilloustrophoblast

TET:

Ten–eleven translocation

MMP:

Matrix metalloproteinase

TIMP:

Tissue inhibitors of metalloproteinase

IGF:

Insulin-like growth factor

HIF:

Hypoxia inducible factor

References

  1. Wu P, Green M, Myers JE. Hypertensive disorders of pregnancy. BMJ. 2023;381:e071653. https://doi.org/10.1136/bmj-2022-071653. Epub 2023/07/01.

    Article  PubMed  Google Scholar 

  2. Gestational Hypertension and Preeclampsia: Acog Practice Bulletin, Number 222. Obstet Gynecol. 2020;135(6):e237–e60. https://doi.org/10.1097/AOG.0000000000003891. Epub 2020/05/23.

  3. Steegers EA, von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-Eclampsia. Lancet. 2010;376(9741):631–44. https://doi.org/10.1016/S0140-6736(10)60279-6. Epub 2010/07/06.

    Article  PubMed  Google Scholar 

  4. Geelhoed JJ, Fraser A, Tilling K, Benfield L, Davey Smith G, Sattar N, et al. Preeclampsia and gestational hypertension are associated with childhood blood pressure independently of family adiposity measures: the avon longitudinal study of parents and children. Circulation. 2010;122(12):1192–9. https://doi.org/10.1161/CIRCULATIONAHA.110.936674. Epub 2010/09/09.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Jayet PY, Rimoldi SF, Stuber T, Salmon CS, Hutter D, Rexhaj E, et al. Pulmonary and systemic vascular dysfunction in young offspring of mothers with preeclampsia. Circulation. 2010;122(5):488–94. https://doi.org/10.1161/CIRCULATIONAHA.110.941203. Epub 2010/07/21.

    Article  PubMed  Google Scholar 

  6. Garovic VD, White WM, Vaughan L, Saiki M, Parashuram S, Garcia-Valencia O, et al. Incidence and long-term outcomes of hypertensive disorders of pregnancy. J Am Coll Cardiol. 2020;75(18):2323–34. https://doi.org/10.1016/j.jacc.2020.03.028. Epub 2020/05/10.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Hauspurg A, Countouris ME, Catov JM. Hypertensive disorders of pregnancy and future maternal health: how can the evidence guide postpartum management? Curr Hypertens Rep. 2019;21(12):96. https://doi.org/10.1007/s11906-019-0999-7. Epub 2019/11/30.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Andrews S, Krueger C, Mellado-Lopez M, Hemberger M, Dean W, Perez-Garcia V, et al. Mechanisms and function of de novo DNA methylation in placental development reveals an essential role for Dnmt3b. Nat Commun. 2023;14(1):371. https://doi.org/10.1038/s41467-023-36019-9. Epub 2023/01/24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Deng J, Zhao HJ, Zhong Y, Hu C, Meng J, Wang C, et al. H3k27me3-modulated hofbauer cell bmp2 signalling enhancement compensates for shallow trophoblast invasion in preeclampsia. EBioMedicine. 2023;93:104664. https://doi.org/10.1016/j.ebiom.2023.104664. Epub 2023/06/19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lyko F, The DNA. Methyltransferase family: a versatile toolkit for epigenetic regulation. Nat Rev Genet. 2018;19(2):81–92. https://doi.org/10.1038/nrg.2017.80. Epub 2017/10/17.

    Article  CAS  PubMed  Google Scholar 

  11. Sun M, Song MM, Wei B, Gao Q, Li L, Yao B, et al. 5-Hydroxymethylcytosine-mediated alteration of transposon activity associated with the exposure to adverse in utero environments in human. Hum Mol Genet. 2016;25(11):2208–19. https://doi.org/10.1093/hmg/ddw089. Epub 2016/10/30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Jia RZ, Zhang X, Hu P, Liu XM, Hua XD, Wang X, et al. Screening for differential methylation status in human placenta in preeclampsia using a Cpg Island plus promoter microarray. Int J Mol Med. 2012;30(1):133–41. https://doi.org/10.3892/ijmm.2012.983. Epub 2012/05/04.

    Article  CAS  PubMed  Google Scholar 

  13. Blair JD, Yuen RK, Lim BK, McFadden DE, von Dadelszen P, Robinson WP. Widespread DNA hypomethylation at gene enhancer regions in placentas associated with early-onset pre-eclampsia. Mol Hum Reprod. 2013;19(10):697–708. https://doi.org/10.1093/molehr/gat044. Epub 2013/06/19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Xu W, Ru P, Gu Z, Zhang R, Pang X, Huang Y, et al. Comprehensive analysis of differently expressed and methylated genes in preeclampsia. Comput Math Methods Med. 2020;2020:2139270. https://doi.org/10.1155/2020/2139270. Epub 2020/11/19.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Lim JH, Kang YJ, Bak HJ, Kim MS, Lee HJ, Kwak DW, et al. Epigenome-wide DNA methylation profiling of preeclamptic placenta according to severe features. Clin Epigenetics. 2020;12(1):128. https://doi.org/10.1186/s13148-020-00918-1. Epub 2020/08/25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li Y, Cui S, Shi W, Yang B, Yuan Y, Yan S, et al. Differential placental methylation in preeclampsia, preterm and term pregnancies. Placenta. 2020;93:56–63. https://doi.org/10.1016/j.placenta.2020.02.009. Epub 2020/04/07.

    Article  CAS  PubMed  Google Scholar 

  17. Mayne BT, Leemaqz SY, Smith AK, Breen J, Roberts CT, Bianco-Miotto T. Accelerated placental aging in early onset preeclampsia pregnancies identified by DNA methylation. Epigenomics. 2017;9(3):279–89. https://doi.org/10.2217/epi-2016-0103. Epub 2016/11/30.

    Article  CAS  PubMed  Google Scholar 

  18. Chu T, Bunce K, Shaw P, Shridhar V, Althouse A, Hubel C, et al. Comprehensive analysis of preeclampsia-associated DNA methylation in the placenta. PLoS One. 2014;9(9):e107318. https://doi.org/10.1371/journal.pone.0107318. Epub 2014/09/24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kalisch-Smith JI, Simmons DG, Dickinson H, Moritz KM. Review: sexual dimorphism in the formation, function and adaptation of the placenta. Placenta. 2017;54:10–6. https://doi.org/10.1016/j.placenta.2016.12.008. Epub 2016/12/17.

    Article  CAS  PubMed  Google Scholar 

  20. Kalisch-Smith JI, Simmons DG, Pantaleon M, Moritz KM. Sex differences in rat placental development: from pre-implantation to late gestation. Biol Sex Differ. 2017;8:17. https://doi.org/10.1186/s13293-017-0138-6. Epub 2017/05/20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. True H, Blanton M, Sureshchandra S, Messaoudi I. Monocytes and macrophages in pregnancy: the good, the bad, and the ugly. Immunol Rev. 2022;308(1):77–92. https://doi.org/10.1111/imr.13080. Epub 2022/04/23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Huang Q, Li J, Wang F, Oliver MT, Tipton T, Gao Y, et al. Syncytin-1 modulates placental trophoblast cell proliferation by promoting G1/S transition. Cell Signal. 2013;25(4):1027–35. https://doi.org/10.1016/j.cellsig.2013.01.008. Epub 2013/01/22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Langbein M, Strick R, Strissel PL, Vogt N, Parsch H, Beckmann MW, et al. Impaired cytotrophoblast cell-cell fusion is associated with reduced syncytin and increased apoptosis in patients with placental dysfunction. Mol Reprod Dev. 2008;75(1):175–83. https://doi.org/10.1002/mrd.20729. Epub 2007/06/05.

    Article  CAS  PubMed  Google Scholar 

  24. Zhuang XW, Li J, Brost BC, Xia XY, Chen HB, Wang CX, et al. Decreased expression and altered methylation of syncytin-1 gene in human placentas associated with preeclampsia. Curr Pharm Des. 2014;20(11):1796–802. https://doi.org/10.2174/13816128113199990541. Epub 2013/07/31.

    Article  CAS  PubMed  Google Scholar 

  25. Ruebner M, Strissel PL, Ekici AB, Stiegler E, Dammer U, Goecke TW, et al. Reduced syncytin-1 expression levels in placental syndromes correlates with epigenetic hypermethylation of the Ervw-1 promoter region. PLoS One. 2013;8(2):e56145 Epub 2013/03/05.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ma X, Li J, Brost B, Cheng W, Jiang SW. Decreased Expression and DNA Methylation Levels of Gatad1 in Preeclamptic Placentas. Cell Signal. 2014;26(5):959–67. https://doi.org/10.1016/j.cellsig.2014.01.013. Epub 2014/01/28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Girardi G. Role of tissue factor in pregnancy complications: crosstalk between coagulation and inflammation. Thromb Res. 2011;127(Suppl 3):S43–6. https://doi.org/10.1016/S0049-3848(11)70012-3. Epub 2011/01/26.

    Article  CAS  PubMed  Google Scholar 

  28. Liu R, Ma Q, Wen A, Tian G, Li M, Wang W, et al. Increased tissue factor expression and promoter hypomethylation in preeclampsia placentas in a Chinese population. Pregnancy Hypertens. 2017;10:90–5. https://doi.org/10.1016/j.preghy.2017.06.008. Epub 2017/11/21.

    Article  PubMed  Google Scholar 

  29. Wong AE, Kwaan HC, Grobman WA, Weiss I, Wong CA. Microparticle source and tissue factor expression in pregnancy. Ann Hematol. 2015;94(8):1285–90. https://doi.org/10.1007/s00277-015-2355-6. Epub 2015/04/01.

    Article  CAS  PubMed  Google Scholar 

  30. Godoi LC, Gomes KB, Alpoim PN, Carvalho M, Lwaleed BA, Sant’Ana Dusse LM. Preeclampsia: the role of tissue factor and tissue factor pathway inhibitor. J Thromb Thrombolysis. 2012;34(1):1–6. https://doi.org/10.1007/s11239-012-0708-8. Epub 2012/03/31.

    Article  CAS  PubMed  Google Scholar 

  31. Than NG, Romero R, Kim CJ, McGowen MR, Papp Z, Wildman DE. Galectins: guardians of eutherian pregnancy at the maternal-fetal interface. Trends Endocrinol Metab. 2012;23(1):23–31. https://doi.org/10.1016/j.tem.2011.09.003. Epub 2011/11/01.

    Article  CAS  PubMed  Google Scholar 

  32. Than NG, Romero R, Xu Y, Erez O, Xu Z, Bhatti G, et al. Evolutionary origins of the placental expression of chromosome 19 cluster galectins and their complex dysregulation in preeclampsia. Placenta. 2014;35(11):855–65. https://doi.org/10.1016/j.placenta.2014.07.015. Epub 2014/10/01.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Saftlas AF, Beydoun H, Triche E. Immunogenetic determinants of preeclampsia and related pregnancy disorders: a systematic review. Obstet Gynecol. 2005;106(1):162–72. https://doi.org/10.1097/01.AOG.0000167389.97019.37. Epub 2005/07/05.

    Article  PubMed  Google Scholar 

  34. Chen LJ, Han ZQ, Zhou H, Zou L, Zou P. Inhibition of Hla-G expression via rnai abolishes resistance of extravillous trophoblast cell line Tev-1 to Nk Lysis. Placenta. 2010;31(6):519–27. https://doi.org/10.1016/j.placenta.2010.03.008. Epub 2010/05/01.

    Article  CAS  PubMed  Google Scholar 

  35. Liu H, Liu X, Jin H, Yang F, Gu W, Li X. Proteomic analysis of knock-down Hla-G in invasion of human trophoblast cell line jeg-3. Int J Clin Exp Pathol. 2013;6(11):2451–9 Epub 2013/11/15.

    PubMed  PubMed Central  Google Scholar 

  36. Le Bouteiller P, Fons P, Herault JP, Bono F, Chabot S, Cartwright JE, et al. Soluble Hla-G and control of angiogenesis. J Reprod Immunol. 2007;76(1–2):17–22. https://doi.org/10.1016/j.jri.2007.03.007. Epub 2007/05/01.

    Article  CAS  PubMed  Google Scholar 

  37. Xu X, Zhou Y, Wei H. Roles of Hla-G in the maternal-fetal immune microenvironment. Front Immunol. 2020;11:592010. https://doi.org/10.3389/fimmu.2020.592010. Epub 2020/11/17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dunk CE, Bucher M, Zhang J, Hayder H, Geraghty DE, Lye SJ, et al. Human leukocyte antigen Hla-C, Hla-G, Hla-F, and Hla-E placental profiles are altered in early severe preeclampsia and preterm birth with chorioamnionitis. Am J Obstet Gynecol. 2022;227(4):641 e1–e13. https://doi.org/10.1016/j.ajog.2022.07.021. Epub 2022/07/22.

    Article  CAS  PubMed  Google Scholar 

  39. Tang Y, Liu H, Li H, Peng T, Gu W, Li X. Hypermethylation of the Hla-G promoter is associated with preeclampsia. Mol Hum Reprod. 2015;21(9):736–44. https://doi.org/10.1093/molehr/gav037. Epub 2015/06/28.

    Article  CAS  PubMed  Google Scholar 

  40. Verloes A, Spits C, Vercammen M, Geens M, LeMaoult J, Sermon K, et al. The role of methylation, DNA polymorphisms and micrornas on Hla-G expression in human embryonic stem cells. Stem Cell Res. 2017;19:118–27. https://doi.org/10.1016/j.scr.2017.01.005. Epub 2017/01/31.

    Article  CAS  PubMed  Google Scholar 

  41. Zhu L, Lv R, Kong L, Li X. Reduced methylation downregulates Cd39/Entpd1 and Zdhhc14 to suppress trophoblast cell proliferation and invasion: implications in preeclampsia. Pregnancy Hypertens. 2018;14:59–67. https://doi.org/10.1016/j.preghy.2018.03.012. Epub 2018/12/12.

    Article  PubMed  Google Scholar 

  42. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by Cd39 and Cd73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–65. https://doi.org/10.1084/jem.20062512. Epub 2007/05/16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chapman K, Holmes M, Seckl J. 11beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. Physiol Rev. 2013;93(3):1139–206. https://doi.org/10.1152/physrev.00020.2012. Epub 2013/08/01.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132(5):1033–44. https://doi.org/10.1016/j.jaci.2013.09.007. Epub 2013/10/03.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Baum M, Moe OW. Glucocorticoid-mediated hypertension: does the vascular smooth muscle hold all the answers? J Am Soc Nephrol. 2008;19(7):1251–3. https://doi.org/10.1681/ASN.2008040410. Epub 2008/05/30.

    Article  PubMed  Google Scholar 

  46. Sawady J, Mercer BM, Wapner RJ, Zhao Y, Sorokin Y, Johnson F, et al. The National Institute of Child Health and Human Development maternal-fetal medicine units network beneficial effects of antenatal repeated steroids study: impact of repeated doses of antenatal corticosteroids on placental growth and histologic findings. Am J Obstet Gynecol. 2007;197(3):281 e1–8. https://doi.org/10.1016/j.ajog.2007.06.041. Epub 2007/09/11.

    Article  CAS  PubMed  Google Scholar 

  47. Hogg K, Robinson WP, Beristain AG. Activation of endocrine-related gene expression in placental choriocarcinoma cell lines following DNA methylation knock-down. Mol Hum Reprod. 2014;20(7):677–89. https://doi.org/10.1093/molehr/gau020. Epub 2014/03/14.

    Article  CAS  PubMed  Google Scholar 

  48. Hogg K, Blair JD, McFadden DE, von Dadelszen P, Robinson WP. Early onset pre-eclampsia is associated with altered DNA methylation of cortisol-signalling and steroidogenic genes in the placenta. PLoS One. 2013;8(5):e62969. https://doi.org/10.1371/journal.pone.0062969. Epub 2013/05/15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Dwi Putra SE, Reichetzeder C, Meixner M, Liere K, Slowinski T, Hocher B. DNA methylation of the glucocorticoid receptor gene promoter in the placenta is associated with blood pressure regulation in human pregnancy. J Hypertens. 2017;35(11):2276–86. https://doi.org/10.1097/HJH.0000000000001450. Epub 2017/08/18.

    Article  CAS  PubMed  Google Scholar 

  50. Torres-Salazar Q, Martinez-Lopez Y, Reyes-Romero M, Perez-Morales R, Sifuentes-Alvarez A, Salvador-Moysen J. Differential methylation in promoter regions of the genes Nr3c1 and Hsp90aa1, involved in the regulation, and bioavailability of cortisol in leukocytes of women with preeclampsia. Front Med (Lausanne). 2020;7:206. https://doi.org/10.3389/fmed.2020.00206. Epub 2020/07/14.

    Article  PubMed  Google Scholar 

  51. Zhu P, Wang W, Zuo R, Sun K. Mechanisms for establishment of the placental glucocorticoid barrier, a guard for life. Cell Mol Life Sci. 2019;76(1):13–26. https://doi.org/10.1007/s00018-018-2918-5. Epub 2018/09/19.

    Article  CAS  PubMed  Google Scholar 

  52. Aufdenblatten M, Baumann M, Raio L, Dick B, Frey BM, Schneider H, et al. Prematurity is related to high placental cortisol in preeclampsia. Pediatr Res. 2009;65(2):198–202. https://doi.org/10.1203/PDR.0b013e31818d6c24. Epub 2008/12/03.

    Article  CAS  PubMed  Google Scholar 

  53. Dy J, Guan H, Sampath-Kumar R, Richardson BS, Yang K. Placental 11beta-hydroxysteroid dehydrogenase type 2 is reduced in pregnancies complicated with idiopathic intrauterine growth restriction: evidence that this is associated with an attenuated ratio of cortisone to cortisol in the umbilical artery. Placenta. 2008;29(2):193–200. https://doi.org/10.1016/j.placenta.2007.10.010. Epub 2007/12/07.

    Article  CAS  PubMed  Google Scholar 

  54. Causevic M, Mohaupt M. 11beta-hydroxysteroid dehydrogenase type 2 in pregnancy and preeclampsia. Mol Aspects Med. 2007;28(2):220–6. https://doi.org/10.1016/j.mam.2007.04.003. Epub 2007/05/29.

    Article  CAS  PubMed  Google Scholar 

  55. Kosicka K, Siemiatkowska A, Glowka FK. 11beta-hydroxysteroid dehydrogenase 2 in preeclampsia. Int J Endocrinol. 2016;2016:5279462. https://doi.org/10.1155/2016/5279462. Epub 2016/05/21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kosicka K, Siemiatkowska A, Krzyscin M, Breborowicz GH, Resztak M, Majchrzak-Celinska A, et al. Glucocorticoid metabolism in hypertensive disorders of pregnancy: analysis of plasma and urinary cortisol and cortisone. PLoS One. 2015;10(12):e0144343. https://doi.org/10.1371/journal.pone.0144343. Epub 2015/12/05.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Schoof E, Girstl M, Frobenius W, Kirschbaum M, Dorr HG, Rascher W, et al. Decreased gene expression of 11beta-hydroxysteroid dehydrogenase type 2 and 15-hydroxyprostaglandin dehydrogenase in human placenta of patients with preeclampsia. J Clin Endocrinol Metab. 2001;86(3):1313–7. https://doi.org/10.1210/jcem.86.3.7311. Epub 2001/03/10.

    Article  CAS  PubMed  Google Scholar 

  58. Alikhani-Koopaei R, Fouladkou F, Frey FJ, Frey BM. Epigenetic regulation of 11 beta-hydroxysteroid dehydrogenase type 2 expression. J Clin Invest. 2004;114(8):1146–57. https://doi.org/10.1172/JCI21647. Epub 2004/10/19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Green BB, Armstrong DA, Lesseur C, Paquette AG, Guerin DJ, Kwan LE, et al. The role of placental 11-beta hydroxysteroid dehydrogenase type 1 and type 2 methylation on gene expression and infant birth weight. Biol Reprod. 2015;92(6):149. https://doi.org/10.1095/biolreprod.115.128066. Epub 2015/03/20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hu W, Wang H, Huang H. Analysis of gene expression and preliminary study of methylation about 11beta-Hsd2 gene in placentas of Chinese pre-eclampsia patients of han ethnicity. J Obstet Gynaecol Res. 2015;41(3):343–9. https://doi.org/10.1111/jog.12555. Epub 2014/10/22.

    Article  CAS  PubMed  Google Scholar 

  61. Pilvar D, Reiman M, Pilvar A, Laan M. Parent-of-origin-specific allelic expression in the human placenta is limited to established imprinted loci and it is stably maintained across pregnancy. Clin Epigenetics. 2019;11(1):94. https://doi.org/10.1186/s13148-019-0692-3. Epub 2019/06/28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Monk D. Genomic Imprinting in the Human Placenta. Am J Obstet Gynecol. 2015;213(4 Suppl):S152–62. https://doi.org/10.1016/j.ajog.2015.06.032. Epub 2015/10/03.

    Article  PubMed  Google Scholar 

  63. Coan PM, Burton GJ, Ferguson-Smith AC. Imprinted genes in the placenta- a review. Placenta. 2005;26(Suppl A):S10–20. https://doi.org/10.1016/j.placenta.2004.12.009. Epub 2005/04/20.

    Article  CAS  PubMed  Google Scholar 

  64. Yu L, Chen M, Zhao D, Yi P, Lu L, Han J, et al. The H19 gene imprinting in normal pregnancy and pre-eclampsia. Placenta. 2009;30(5):443–7. https://doi.org/10.1016/j.placenta.2009.02.011. Epub 2009/04/04.

    Article  CAS  PubMed  Google Scholar 

  65. Gao WL, Li D, Xiao ZX, Liao QP, Yang HX, Li YX, et al. Detection of global DNA methylation and paternally imprinted H19 gene methylation in preeclamptic placentas. Hypertens Res. 2011;34(5):655–61. https://doi.org/10.1038/hr.2011.9. Epub 2011/02/18.

    Article  CAS  PubMed  Google Scholar 

  66. LaRocca J, Binder AM, McElrath TF, Michels KB. The impact of first trimester phthalate and phenol exposure on igf2/h19 genomic imprinting and birth outcomes. Environ Res. 2014;133:396–406. https://doi.org/10.1016/j.envres.2014.04.032. Epub 2014/06/29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kuhnel E, Kleff V, Stojanovska V, Kaiser S, Waldschutz R, Herse F, et al. Placental-specific overexpression of Sflt-1 alters trophoblast differentiation and nutrient transporter expression in an iugr mouse model. J Cell Biochem. 2017;118(6):1316–29. https://doi.org/10.1002/jcb.25789. Epub 2016/11/20.

    Article  CAS  PubMed  Google Scholar 

  68. Constancia M, Hemberger M, Hughes J, Dean W, Ferguson-Smith A, Fundele R, et al. Placental-specific Igf-Ii Is a major modulator of placental and fetal growth. Nature. 2002;417(6892):945–8. https://doi.org/10.1038/nature00819. Epub 2002/06/28.

    Article  CAS  PubMed  Google Scholar 

  69. van den Berg CB, Herzog EM, Duvekot JJ, van der Spek PJ, Steegers EAP, Stoop MP, et al. Differences in DNA methylation of insulin-like growth factor 2 and cadherin 13 in patients with preeclampsia. Pregnancy Hypertens. 2020;19:150–8. https://doi.org/10.1016/j.preghy.2020.01.010. Epub 2020/02/03.

    Article  PubMed  Google Scholar 

  70. He J, Zhang A, Fang M, Fang R, Ge J, Jiang Y, et al. Methylation levels at Igf2 and gnas dmrs in infants born to preeclamptic pregnancies. BMC Genomics. 2013;14:472. https://doi.org/10.1186/1471-2164-14-472. Epub 2013/07/13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Crosley EJ, Dunk CE, Beristain AG, Christians JK. Igfbp-4 and -5 are expressed in first-trimester villi and differentially regulate the migration of Htr-8/Svneo cells. Reprod Biol Endocrinol. 2014;12:123. https://doi.org/10.1186/1477-7827-12-123. Epub 2014/12/06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jia Y, Li T, Huang X, Xu X, Zhou X, Jia L, et al. Dysregulated DNA methyltransferase 3a upregulates Igfbp5 to suppress trophoblast cell migration and invasion in preeclampsia. Hypertension. 2017;69(2):356–66. https://doi.org/10.1161/HYPERTENSIONAHA.116.08483. Epub 2017/01/05.

    Article  CAS  PubMed  Google Scholar 

  73. Ma M, Zhou QJ, Xiong Y, Li B, Li XT. Preeclampsia is associated with hypermethylation of Igf-1 promoter mediated by Dnmt1. Am J Transl Res. 2018;10(1):16–39 Epub 2018/02/10.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Rachmilewitz J, Gileadi O, Eldar-Geva T, Schneider T, de-Groot N, Hochberg A. Transcription of the H19 gene in differentiating cytotrophoblasts from human placenta. Mol Reprod Dev. 1992;32(3):196–202. https://doi.org/10.1002/mrd.1080320303. Epub 1992/07/01.

    Article  CAS  PubMed  Google Scholar 

  75. Adam GI, Cui H, Miller SJ, Flam F, Ohlsson R. Allele-specific in situ hybridization (Asish) analysis: a novel technique which resolves differential allelic usage of H19 within the same cell lineage during human placental development. Development. 1996;122(3):839–47. https://doi.org/10.1242/dev.122.3.839. Epub 1996/03/01.

    Article  CAS  PubMed  Google Scholar 

  76. Lu L, Hou Z, Li L, Yang Y, Wang X, Zhang B, et al. Methylation pattern of H19 exon 1 is closely related to preeclampsia and trophoblast abnormalities. Int J Mol Med. 2014;34(3):765–71. https://doi.org/10.3892/ijmm.2014.1816. Epub 2014/06/28.

    Article  CAS  PubMed  Google Scholar 

  77. Harati-Sadegh M, Kohan L, Teimoori B, Mehrabani M, Salimi S. The effects of placental long noncoding Rna H19 polymorphisms and promoter methylation on h19 expression in association with preeclampsia susceptibility. IUBMB Life. 2020;72(3):413–25. https://doi.org/10.1002/iub.2199. Epub 2019/11/27.

    Article  CAS  PubMed  Google Scholar 

  78. Romero R, Erez O, Huttemann M, Maymon E, Panaitescu B, Conde-Agudelo A, et al. Metformin, the aspirin of the 21st century: its role in gestational diabetes mellitus, prevention of preeclampsia and cancer, and the promotion of longevity. Am J Obstet Gynecol. 2017;217(3):282–302. https://doi.org/10.1016/j.ajog.2017.06.003. Epub 2017/06/18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhou Q, Xiong Y, Chen Y, Du Y, Zhang J, Mu J, et al. Effects of tissue factor pathway inhibitor-2 expression on biological behavior of bewo and jeg-3 cell lines. Clin Appl Thromb Hemost. 2012;18(5):526–33. https://doi.org/10.1177/1076029611429785. Epub 2011/12/29.

    Article  CAS  PubMed  Google Scholar 

  80. Xiao X, Tao X, Wang Y, Zhu L, Ye Y, Liu H, et al. Hypomethylation of tissue factor pathway inhibitor 2 in human placenta of preeclampsia. Thromb Res. 2017;152:7–13. https://doi.org/10.1016/j.thromres.2017.02.005. Epub 2017/02/17.

    Article  CAS  PubMed  Google Scholar 

  81. Dokras A, Gardner LM, Kirschmann DA, Seftor EA, Hendrix MJ. The tumour suppressor gene maspin is differentially regulated in cytotrophoblasts during human placental development. Placenta. 2002;23(4):274–80. https://doi.org/10.1053/plac.2001.0784. Epub 2002/04/24.

    Article  CAS  PubMed  Google Scholar 

  82. Liu Q, Qiao FY, Shi XW, Liu HY, Gong X, Wu YY. Promoter hypomethylation and increased maspin expression in preeclamptic placentas in a chinese population. Placenta. 2014;35(11):876–82. https://doi.org/10.1016/j.placenta.2014.08.088. Epub 2014/08/26.

    Article  CAS  PubMed  Google Scholar 

  83. Alahari S, Garcia J, Post M, Caniggia I. The von hippel lindau tumour suppressor gene is a novel target of E2f4-mediated transcriptional repression in preeclampsia. Biochim Biophys Acta Mol Basis Dis. 2018;1864(10):3298–308. https://doi.org/10.1016/j.bbadis.2018.07.016. Epub 2018/07/28.

    Article  CAS  PubMed  Google Scholar 

  84. Peng Z, Shen R, Li YW, Teng KY, Shapiro CL, Lin HJ. Epigenetic repression of rarres1 is mediated by methylation of a proximal promoter and a loss of Ctcf binding. PLoS One. 2012;7(5):e36891. https://doi.org/10.1371/journal.pone.0036891. Epub 2012/05/23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Oldridge EE, Walker HF, Stower MJ, Simms MS, Mann VM, Collins AT, et al. Retinoic acid represses invasion and stem cell phenotype by induction of the metastasis suppressors Rarres1 and Lxn. Oncogenesis. 2013;2(4):e45. https://doi.org/10.1038/oncsis.2013.6. Epub 2013/04/17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Das BC, Thapa P, Karki R, Das S, Mahapatra S, Liu TC, et al. Retinoic acid signaling pathways in development and diseases. Bioorg Med Chem. 2014;22(2):673–83. https://doi.org/10.1016/j.bmc.2013.11.025. Epub 2014/01/08.

    Article  CAS  PubMed  Google Scholar 

  87. Huebner H, Hartner A, Rascher W, Strick RR, Kehl S, Heindl F, et al. Expression and regulation of retinoic acid receptor responders in the human placenta. Reprod Sci. 2018;25(9):1357–70. https://doi.org/10.1177/1933719117746761. Epub 2017/12/17.

    Article  CAS  PubMed  Google Scholar 

  88. Harati-Sadegh M, Kohan L, Teimoori B, Mehrabani M, Salimi S. Analysis of polymorphisms, promoter methylation, and mrna expression profile of maternal and placental p53 and p21 genes in preeclamptic and normotensive pregnant women. J Biomed Sci. 2019;26(1):92. https://doi.org/10.1186/s12929-019-0586-x. Epub 2019/11/11.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Monkley SJ, Delaney SJ, Pennisi DJ, Christiansen JH, Wainwright BJ. Targeted disruption of the Wnt2 gene results in placentation defects. Development. 1996;122(11):3343–53. https://doi.org/10.1242/dev.122.11.3343. Epub 1996/11/01.

    Article  CAS  PubMed  Google Scholar 

  90. Zhang Z, Zhang L, Zhang L, Jia L, Wang P, Gao Y. Association of Wnt2 and Sfrp4 expression in the third trimester placenta in women with severe preeclampsia. Reprod Sci. 2013;20(8):981–9. https://doi.org/10.1177/1933719112472740. Epub 2013/01/17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Yeung KR, Chiu CL, Pidsley R, Makris A, Hennessy A, Lind JM. DNA methylation profiles in preeclampsia and healthy control placentas. Am J Physiol Heart Circ Physiol. 2016;310(10):H1295-303. https://doi.org/10.1152/ajpheart.00958.2015. Epub 2016/03/13.

    Article  PubMed  Google Scholar 

  92. Liu Y, Ma Y. Promoter methylation status of Wnt2 in placenta from patients with preeclampsia. Med Sci Monit. 2017;23:5294–301. https://doi.org/10.12659/msm.903602. Epub 2017/11/08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhang L, Sang M, Li Y, Li Y, Yuan E, Yang L, et al. Wnt3 hypomethylation counteracts low activity of the wnt signaling pathway in the placenta of preeclampsia. Cell Mol Life Sci. 2021;78(21–22):6995–7008. https://doi.org/10.1007/s00018-021-03941-4. Epub 2021/10/06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Akhurst RJ, Hata A. Targeting the Tgfbeta signalling pathway in disease. Nat Rev Drug Discov. 2012;11(10):790–811. https://doi.org/10.1038/nrd3810. Epub 2012/09/25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Shi Y, Massague J. Mechanisms of Tgf-beta signaling from cell membrane to the nucleus. Cell. 2003;113(6):685–700. https://doi.org/10.1016/s0092-8674(03)00432-x. Epub 2003/06/18.

    Article  CAS  PubMed  Google Scholar 

  96. Jia Y, Xie H, Zhang J, Ying H. Induction of Tgf-beta receptor I expression in a DNA methylation-independent manner mediated by Dnmt3a downregulation is involved in early-onset severe preeclampsia. FASEB J. 2020;34(10):13224–38. https://doi.org/10.1096/fj.202000253RR. Epub 2020/08/15.

    Article  CAS  PubMed  Google Scholar 

  97. Roca C, Adams RH. Regulation of vascular morphogenesis by notch signaling. Genes Dev. 2007;21(20):2511–24. https://doi.org/10.1101/gad.1589207. Epub 2007/10/17.

    Article  CAS  PubMed  Google Scholar 

  98. Shimanuki Y, Mitomi H, Fukumura Y, Makino S, Itakura A, Yao T, et al. Alteration of delta-like ligand 1 and notch 1 receptor in various placental disorders with special reference to early onset preeclampsia. Hum Pathol. 2015;46(8):1129–37. https://doi.org/10.1016/j.humpath.2015.03.013. Epub 2015/05/28.

    Article  CAS  PubMed  Google Scholar 

  99. Li H, Ouyang Y, Sadovsky E, Parks WT, Chu T, Sadovsky Y. Unique microrna signals in plasma exosomes from pregnancies complicated by preeclampsia. Hypertension. 2020;75(3):762–71. https://doi.org/10.1161/HYPERTENSIONAHA.119.14081. Epub 2020/01/28.

    Article  CAS  PubMed  Google Scholar 

  100. Skalis G, Katsi V, Miliou A, Georgiopoulos G, Papazachou O, Vamvakou G, et al. Micrornas in preeclampsia. Microrna. 2019;8(1):28–35. https://doi.org/10.2174/2211536607666180813123303. Epub 2018/08/14.

    Article  CAS  PubMed  Google Scholar 

  101. Agarwal S, Hanna J, Sherman ME, Figueroa J, Rimm DL. Quantitative assessment of mir34a as an independent prognostic marker in breast cancer. Br J Cancer. 2015;112(1):61–8. https://doi.org/10.1038/bjc.2014.573. Epub 2014/12/05.

    Article  CAS  PubMed  Google Scholar 

  102. Doridot L, Houry D, Gaillard H, Chelbi ST, Barbaux S, Vaiman D. Mir-34a expression, epigenetic regulation, and function in human placental diseases. Epigenetics. 2014;9(1):142–51. https://doi.org/10.4161/epi.26196. Epub 2013/10/02.

    Article  CAS  PubMed  Google Scholar 

  103. Guo M, Zhao X, Yuan X, Li P. Elevated microrna-34a contributes to trophoblast cell apoptosis in preeclampsia by targeting Bcl-2. J Hum Hypertens. 2017;31(12):815–20. https://doi.org/10.1038/jhh.2017.65. Epub 2017/10/13.

    Article  CAS  PubMed  Google Scholar 

  104. Rezaei M, Eskandari F, Mohammadpour-Gharehbagh A, Harati-Sadegh M, Teimoori B, Salimi S. Hypomethylation of the Mirna-34a Gene Promoter Is Associated with Severe Preeclampsia. Clin Exp Hypertens (2018):1–5. https://doi.org/10.1080/10641963.2018.1451534. Epub 2018/03/21

  105. Bounds KR, Chiasson VL, Pan LJ, Gupta S, Chatterjee P. Micrornas: new players in the pathobiology of preeclampsia. Front Cardiovasc Med. 2017;4:60. https://doi.org/10.3389/fcvm.2017.000600. Epub 2017/10/11.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Du J, Ji Q, Dong L, Meng Y, Xin G. Hdac4 knockdown induces preeclampsia cell autophagy and apoptosis by Mir-29b. Reprod Sci. 2021;28(2):334–42. https://doi.org/10.1007/s43032-020-00286-4. Epub 2020/08/12.

    Article  CAS  PubMed  Google Scholar 

  107. Sonkar R, Kay MK, Choudhury M. Pfos modulates interactive epigenetic regulation in first-trimester human trophoblast cell line Htr-8/Sv(Neo). Chem Res Toxicol. 2019;32(10):2016–27. https://doi.org/10.1021/acs.chemrestox.9b00198. Epub 2019/09/12.

    Article  CAS  PubMed  Google Scholar 

  108. Zha W, Guan S, Liu N, Li Y, Tian Y, Chen Y, et al. Let-7a inhibits Bcl-Xl and Yap1 expression to induce apoptosis of trophoblast cells in early-onset severe preeclampsia. Sci Total Environ. 2020;745:139919. https://doi.org/10.1016/j.scitotenv.2020.139919. Epub 2020/07/30.

    Article  CAS  PubMed  Google Scholar 

  109. Chaulk SG, Lattanzi VJ, Hiemer SE, Fahlman RP, Varelas X. The hippo pathway effectors Taz/Yap regulate dicer expression and microrna biogenesis through Let-7. J Biol Chem. 2014;289(4):1886–91. https://doi.org/10.1074/jbc.C113.529362. Epub 2013/12/11.

    Article  CAS  PubMed  Google Scholar 

  110. Luo X, Pan C, Guo X, Gu C, Huang Y, Guo J, et al. Methylation mediated silencing of mir-155 suppresses the development of preeclampsia in vitro and in vivo by targeting foxo3. Mediators Inflamm. 2022;2022:4250621. https://doi.org/10.1155/2022/4250621. Epub 2022/06/07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Frazier S, McBride MW, Mulvana H, Graham D. From animal models to patients: the role of placental micrornas, Mir-210, Mir-126, and Mir-148a/152 in preeclampsia. Clin Sci (Lond). 2020;134(8):1001–25. https://doi.org/10.1042/CS20200023. Epub 2020/04/28.

    Article  CAS  PubMed  Google Scholar 

  112. Yang A, Zhang H, Sun Y, Wang Y, Yang X, Yang X, et al. Modulation of Fabp4 hypomethylation by dnmt1 and its inverse interaction with mir-148a/152 in the placenta of preeclamptic rats and Htr-8 Cells. Placenta. 2016;46:49–62. https://doi.org/10.1016/j.placenta.2016.08.086. Epub 2016/10/05.

    Article  CAS  PubMed  Google Scholar 

  113. Shangguan X, Liu F, Wang H, He J, Dong M. Alterations in serum adipocyte fatty acid binding protein and retinol binding protein-4 in normal pregnancy and preeclampsia. Clin Chim Acta. 2009;407(1–2):58–61. https://doi.org/10.1016/j.cca.2009.06.031. Epub 2009/07/04.

    Article  CAS  PubMed  Google Scholar 

  114. Fujiwara H, Tatsumi K, Kosaka K, Sato Y, Higuchi T, Yoshioka S, et al. Human blastocysts and endometrial epithelial cells express activated leukocyte cell adhesion molecule (Alcam/Cd166). J Clin Endocrinol Metab. 2003;88(7):3437–43. https://doi.org/10.1210/jc.2002-021888. Epub 2003/07/05.

    Article  CAS  PubMed  Google Scholar 

  115. Wei LL, Pan YS, Tang Q, Yang ZJ, Song WQ, Gao YF, et al. Decreased alcam expression and promoter hypermethylation is associated with preeclampsia. Hypertens Res. 2020;43(1):13–22. https://doi.org/10.1038/s41440-019-0337-0. Epub 2019/10/12.

    Article  PubMed  Google Scholar 

  116. Fujiwara K, Ohuchida K, Sada M, Horioka K, Ulrich CD 3rd, Shindo K, et al. Cd166/Alcam expression is characteristic of tumorigenicity and invasive and migratory activities of pancreatic cancer cells. PLoS One. 2014;9(9):e107247. https://doi.org/10.1371/journal.pone.0107247. Epub 2014/09/16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Feng Y, Wang J, He Y, Zhang H, Jiang M, Cao D, et al. Hoxd8/Diaph2-As1 Epigenetically Regulates Pax3 and Impairs Htr-8/Svneo Cell Function under Hypoxia. Biosci Rep (2019) 39(1). https://doi.org/10.1042/BSR20182022. Epub 2019/01/11.

  118. Cheng Y, Hu R, Jin H, Ma K, Zhou S, Cheng H, et al. Effect of 14-3-3 tau protein on differentiation in bewo choriocarcinoma cells. Placenta. 2010;31(1):60–6. https://doi.org/10.1016/j.placenta.2009.10.002. Epub 2009/11/13.

    Article  CAS  PubMed  Google Scholar 

  119. Liu H, Tang Y, Liu X, Zhou Q, Xiao X, Lan F, et al. 14-3-3 Tau (Ywhaq) gene promoter hypermethylation in human placenta of preeclampsia. Placenta. 2014;35(12):981–8. https://doi.org/10.1016/j.placenta.2014.09.016. Epub 2014/10/13.

    Article  CAS  PubMed  Google Scholar 

  120. Xiong J, Ding N, Gao T, Wang Y, Guo W, Zhang H, et al. Hypermethylation of endoplasmic reticulum disulfide oxidase 1alpha leads to trophoblast cell apoptosis through endoplasmic reticulum stress in preeclampsia. J Cell Biochem. 2018;119(10):8588–99. https://doi.org/10.1002/jcb.27101. Epub 2018/07/31.

    Article  CAS  PubMed  Google Scholar 

  121. Chelbi ST, Mondon F, Jammes H, Buffat C, Mignot TM, Tost J, et al. Expressional and epigenetic alterations of placental serine protease inhibitors: serpina3 is a potential marker of preeclampsia. Hypertension. 2007;49(1):76–83. https://doi.org/10.1161/01.HYP.0000250831.52876.cb. Epub 2006/11/08.

    Article  CAS  PubMed  Google Scholar 

  122. Chelbi ST, Wilson ML, Veillard AC, Ingles SA, Zhang J, Mondon F, et al. Genetic and epigenetic mechanisms collaborate to control serpina3 expression and its association with placental diseases. Hum Mol Genet. 2012;21(9):1968–78. https://doi.org/10.1093/hmg/dds006. Epub 2012/01/17.

    Article  CAS  PubMed  Google Scholar 

  123. Zhang L, Zhao F, Yang C, Tang Q, Zhang R, Li J, et al. Hypomethylation of Ctgf promoter in placenta and peripheral blood of pre-eclampsia women. Reprod Sci. 2020;27(2):468–76. https://doi.org/10.1007/s43032-019-00038-z. Epub 2020/02/06.

    Article  CAS  PubMed  Google Scholar 

  124. Prat C, Blanchon L, Borel V, Gallot D, Herbet A, Bouvier D, et al. Ontogeny of aquaporins in human fetal membranes. Biol Reprod. 2012;86(2):48. https://doi.org/10.1095/biolreprod.111.095448. Epub 2011/11/05.

    Article  CAS  PubMed  Google Scholar 

  125. Ding H, Ding Z, Zhao M, Ji B, Lei J, Chen J, et al. Correlation of amniotic fluid index and placental aquaporin 1 levels in terms of preeclampsia. Placenta. 2022;117:169–78. https://doi.org/10.1016/j.placenta.2021.12.010. Epub 2021/12/21.

    Article  CAS  PubMed  Google Scholar 

  126. Cox B, Sharma P, Evangelou AI, Whiteley K, Ignatchenko V, Ignatchenko A, et al. Translational analysis of mouse and human placental protein and mrna reveals distinct molecular pathologies in human preeclampsia. Mol Cell Proteomics. 2011;10(12):M111 012526. https://doi.org/10.1074/mcp.M111.012526. Epub 2011/10/12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ye W, Shen L, Xiong Y, Zhou Y, Gu H, Yang Z. Preeclampsia Is Associated with Decreased Methylation of the Gna12 Promoter. Ann Hum Genet. 2016;80(1):7–10. https://doi.org/10.1111/ahg.12136. Epub 2016/01/16.

    Article  CAS  PubMed  Google Scholar 

  128. Novakovic B, Evain-Brion D, Murthi P, Fournier T, Saffery R. Variable daxx gene methylation is a common feature of placental trophoblast differentiation, preeclampsia, and response to hypoxia. FASEB J. 2017;31(6):2380–92. https://doi.org/10.1096/fj.201601189RR. Epub 2017/02/23.

    Article  CAS  PubMed  Google Scholar 

  129. Zimna A, Kurpisz M. Hypoxia-inducible factor-1 in physiological and pathophysiological angiogenesis: applications and therapies. Biomed Res Int. 2015;2015:549412. https://doi.org/10.1155/2015/549412. Epub 2015/07/07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kaur L, Sundrani D, Dave K, Randhir K, Mehendale S, Bayyana S, et al. Hypoxia inducible factors (Hif1alpha and Hif3alpha) are differentially methylated in preeclampsia placentae and are associated with birth outcomes. Mol Cell Biochem. 2023;478(10):2309–18. https://doi.org/10.1007/s11010-023-04661-y. Epub 2023/01/29.

    Article  CAS  PubMed  Google Scholar 

  131. Sundrani DP, Reddy US, Joshi AA, Mehendale SS, Chavan-Gautam PM, Hardikar AA, et al. Differential placental methylation and expression of Vegf, Flt-1 and Kdr genes in human term and preterm preeclampsia. Clin Epigenetics. 2013;5(1):6. https://doi.org/10.1186/1868-7083-5-6. Epub 2013/04/30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. K MD, Kaur L, Randhir KN, Mehendale SS, Sundrani DP, Chandak GR, et al. Placental Growth Factor and Fms Related Tyrosine Kinase-1 Are Hypomethylated in Preeclampsia Placentae. Epigenomics 2021;13(4):257–69. https://doi.org/10.2217/epi-2020-0318. Epub 2021/01/21.

  133. Cai Z, Gong Z, Li Z, Li L, Kong W. Vascular extracellular matrix remodeling and hypertension. Antioxid Redox Signal. 2021;34(10):765–83. https://doi.org/10.1089/ars.2020.8110. Epub 2020/05/29.

    Article  CAS  PubMed  Google Scholar 

  134. Wells JM, Gaggar A, Blalock JE. Mmp generated matrikines. Matrix Biol. 2015;44–46:122–9. https://doi.org/10.1016/j.matbio.2015.01.016. Epub 2015/02/01.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Yamamoto K, Murphy G, Troeberg L. Extracellular regulation of metalloproteinases. Matrix Biol. 2015;44–46:255–63. https://doi.org/10.1016/j.matbio.2015.02.007. Epub 2015/02/24.

    Article  CAS  PubMed  Google Scholar 

  136. Wang Z, Lu S, Liu C, Zhao B, Pei K, Tian L, et al. Expressional and epigenetic alterations of placental matrix metalloproteinase 9 in preeclampsia. Gynecol Endocrinol. 2010;26(2):96–102. https://doi.org/10.3109/09513590903184100. Epub 2009/09/03.

    Article  CAS  PubMed  Google Scholar 

  137. van Dijk M, Visser A, Posthuma J, Poutsma A, Oudejans CB. Naturally occurring variation in trophoblast invasion as a source of novel (Epigenetic) biomarkers. Front Genet. 2012;3:22. https://doi.org/10.3389/fgene.2012.00022. Epub 2012/03/01.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Lockwood CJ, Oner C, Uz YH, Kayisli UA, Huang SJ, Buchwalder LF, et al. Matrix metalloproteinase 9 (Mmp9) expression in preeclamptic decidua and Mmp9 induction by tumor necrosis factor alpha and interleukin 1 beta in human first trimester decidual cells. Biol Reprod. 2008;78(6):1064–72. https://doi.org/10.1095/biolreprod.107.063743. Epub 2008/02/16.

    Article  CAS  PubMed  Google Scholar 

  139. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141(1):52–67. https://doi.org/10.1016/j.cell.2010.03.015. Epub 2010/04/08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Yuen RK, Penaherrera MS, von Dadelszen P, McFadden DE, Robinson WP. DNA methylation profiling of human placentas reveals promoter hypomethylation of multiple genes in early-onset preeclampsia. Eur J Hum Genet. 2010;18(9):1006–12. https://doi.org/10.1038/ejhg.2010.63. Epub 2010/05/06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Xiang Y, Zhang X, Li Q, Xu J, Zhou X, Wang T, et al. Promoter hypomethylation of timp3 is associated with pre-eclampsia in a Chinese Population. Mol Hum Reprod. 2013;19(3):153–9. https://doi.org/10.1093/molehr/gas054. Epub 2012/11/23.

    Article  CAS  PubMed  Google Scholar 

  142. Cruz JO, Conceicao I, Sandrim VC, Luizon MR. Comprehensive analyses of DNA methylation of the Timp3 promoter in placentas from early-onset and late-onset preeclampsia. Placenta. 2022;117:118–21. https://doi.org/10.1016/j.placenta.2021.12.003. Epub 2021/12/10.

    Article  CAS  PubMed  Google Scholar 

  143. Namli Kalem M, Kalem Z, Yuce T, Soylemez F. Adamts 1, 4, 12, and 13 levels in maternal blood, cord blood, and placenta in preeclampsia. Hypertens Preg. 2018;37(1):9–17. https://doi.org/10.1080/10641955.2017.1397690. Epub 2017/11/15.

    Article  CAS  Google Scholar 

  144. Zhang L, Zhao F, Li C, Li H, Tang Q, Chen Y, et al. Hypomethylation of DNA promoter upregulates adamts7 and contributes to Htr-8/Svneo and Jeg-3 cells abnormalities in pre-eclampsia. Placenta. 2020;93:26–33. https://doi.org/10.1016/j.placenta.2020.02.013. Epub 2020/04/07.

    Article  CAS  PubMed  Google Scholar 

  145. Walker DW, McLean JR. Absence of adrenergic nerves in the human placenta. Nature. 1971;229(5283):344–5. https://doi.org/10.1038/229344a0. Epub 1971/01/29.

    Article  CAS  PubMed  Google Scholar 

  146. Gao Q, Tang J, Li N, Liu B, Zhang M, Sun M, et al. What is precise pathophysiology in development of hypertension in pregnancy? precision medicine requires precise physiology and pathophysiology. Drug Discov Today. 2018;23(2):286–99. https://doi.org/10.1016/j.drudis.2017.10.021. Epub 2017/11/05.

    Article  CAS  PubMed  Google Scholar 

  147. Gao Q, Li H, Ding H, Fan X, Xu T, Tang J, et al. Hyper-methylation of avpr1a and pkcbeta gene associated with insensitivity to arginine vasopressin in human pre-eclamptic placental vasculature. EBioMedicine. 2019;44:574–81. https://doi.org/10.1016/j.ebiom.2019.05.056. Epub 2019/06/09.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Szukiewicz D, Bilska A, Mittal TK, Stangret A, Wejman J, Szewczyk G, et al. Myometrial contractility influences oxytocin receptor (Oxtr) expression in term trophoblast cells obtained from the maternal surface of the human placenta. BMC Pregnancy Childbirth. 2015;15:220. https://doi.org/10.1186/s12884-015-0656-3. Epub 2015/09/18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Russell JA, Leng G, Douglas AJ. The magnocellular oxytocin system, the fount of maternity: adaptations in pregnancy. Front Neuroendocrinol. 2003;24(1):27–61. https://doi.org/10.1016/s0091-3022(02)00104-8. Epub 2003/03/01.

    Article  CAS  PubMed  Google Scholar 

  150. Fan X, Xu T, Ding H, Li H, Yang Y, He Y, et al. DNA methylation-reprogrammed oxytocin receptor underlies insensitivity to oxytocin in pre-eclamptic placental vasculature. J Cell Mol Med. 2019;23(6):4118–26. https://doi.org/10.1111/jcmm.14299. Epub 2019/04/06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Chen M, Dasgupta C, Xiong F, Zhang L. Epigenetic upregulation of large-conductance Ca2+-activated K+ Channel expression in uterine vascular adaptation to pregnancy. Hypertension. 2014;64(3):610–8. https://doi.org/10.1161/HYPERTENSIONAHA.114.03407. Epub 2014/06/11.

    Article  CAS  PubMed  Google Scholar 

  152. Xiang Y, Zhang J, Li Q, Zhou X, Wang T, Xu M, et al. DNA methylome profiling of maternal peripheral blood and placentas reveal potential fetal DNA markers for non-invasive prenatal testing. Mol Hum Reprod. 2014;20(9):875–84. https://doi.org/10.1093/molehr/gau048. Epub 2014/07/06.

    Article  CAS  PubMed  Google Scholar 

  153. White WM, Brost B, Sun Z, Rose C, Craici I, Wagner SJ, et al. Genome-wide methylation profiling demonstrates hypermethylation in maternal leukocyte DNA in preeclamptic compared to normotensive pregnancies. Hypertens Pregnancy. 2013;32(3):257–69. https://doi.org/10.3109/10641955.2013.796970. Epub 2013/06/21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Lo YM, Corbetta N, Chamberlain PF, Rai V, Sargent IL, Redman CW, et al. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350(9076):485–7. https://doi.org/10.1016/S0140-6736(97)02174-0. Epub 1997/08/16.

    Article  CAS  PubMed  Google Scholar 

  155. Yu SCY, Jiang P, Peng W, Cheng SH, Cheung YTT, Tse OYO, et al. Single-molecule sequencing reveals a large population of long cell-free dna molecules in maternal plasma. Proc Natl Acad Sci U S A. 2021;118(50). https://doi.org/10.1073/pnas.2114937118. Epub 2021/12/08.

  156. Kwak DW, Kim SY, Kim HJ, Lim JH, Kim YH, Ryu HM. Maternal total cell-free DNA in preeclampsia with and without intrauterine growth restriction. Sci Rep. 2020;10(1):11848. https://doi.org/10.1038/s41598-020-68842-1. Epub 2020/07/18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Del Vecchio G, Li Q, Li W, Thamotharan S, Tosevska A, Morselli M, et al. Cell-free DNA methylation and transcriptomic signature prediction of pregnancies with adverse outcomes. Epigenetics. 2021;16(6):642–61. https://doi.org/10.1080/15592294.2020.1816774. Epub 2020/10/14.

    Article  PubMed  Google Scholar 

  158. Tsui DW, Chan KC, Chim SS, Chan LW, Leung TY, Lau TK, et al. Quantitative aberrations of hypermethylated Rassf1a gene sequences in maternal plasma in pre-eclampsia. Prenat Diagn. 2007;27(13):1212–8. https://doi.org/10.1002/pd.1897. Epub 2007/11/13.

    Article  CAS  PubMed  Google Scholar 

  159. Zhao F, Wang J, Liu R, Yang J, Cui K, Wu Y, et al. Quantification and application of the placental epigenetic signature of the rassf1a gene in maternal plasma. Prenat Diagn. 2010;30(8):778–82. https://doi.org/10.1002/pd.2546. Epub 2010/07/28.

    Article  PubMed  Google Scholar 

  160. Kim MJ, Kim SY, Park SY, Ahn HK, Chung JH, Ryu HM. Association of fetal-derived hypermethylated Rassf1a concentration in placenta-mediated pregnancy complications. Placenta. 2013;34(1):57–61. https://doi.org/10.1016/j.placenta.2012.11.001. Epub 2012/11/29.

    Article  CAS  PubMed  Google Scholar 

  161. Rahat B, Thakur S, Hamid A, Bagga R, Kaur J. Association of aberrant methylation at promoter regions of tumor suppressor genes with placental pathologies. Epigenomics. 2016;8(6):767–87. https://doi.org/10.2217/epi.16.7. Epub 2016/06/24.

    Article  CAS  PubMed  Google Scholar 

  162. Salvianti F, Inversetti A, Smid M, Valsecchi L, Candiani M, Pazzagli M, et al. Prospective evaluation of rassf1a cell-free DNA as a biomarker of pre-eclampsia. Placenta. 2015;36(9):996–1001. https://doi.org/10.1016/j.placenta.2015.07.003. Epub 2015/07/18.

    Article  CAS  PubMed  Google Scholar 

  163. Kim HJ, Kim SY, Lim JH, Kwak DW, Park SY, Ryu HM. Quantification and application of potential epigenetic markers in maternal plasma of pregnancies with hypertensive disorders. Int J Mol Sci. 2015;16(12):29875–88. https://doi.org/10.3390/ijms161226201. Epub 2015/12/24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Chim SS, Tong YK, Chiu RW, Lau TK, Leung TN, Chan LY, et al. Detection of the placental epigenetic signature of the maspin gene in maternal plasma. Proc Natl Acad Sci U S A. 2005;102(41):14753–8. https://doi.org/10.1073/pnas.0503335102. Epub 2005/10/06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Qi YH, Teng F, Zhou Q, Liu YX, Wu JF, Yu SS, et al. Unmethylated-Maspin DNA in maternal plasma is associated with severe preeclampsia. Acta Obstet Gynecol Scand. 2015;94(9):983–8. https://doi.org/10.1111/aogs.12691. Epub 2015/06/23.

    Article  CAS  PubMed  Google Scholar 

  166. Rahat B, Thakur S, Bagga R, Kaur J. Epigenetic regulation of Stat5a and its role as fetal DNA epigenetic marker during placental development and dysfunction. Placenta. 2016;44:46–53. https://doi.org/10.1016/j.placenta.2016.06.003. Epub 2016/07/28.

    Article  CAS  PubMed  Google Scholar 

  167. Mousa AA, Archer KJ, Cappello R, Estrada-Gutierrez G, Isaacs CR, Strauss JF 3rd, et al. DNA methylation is altered in maternal blood vessels of women with preeclampsia. Reprod Sci. 2012;19(12):1332–42. https://doi.org/10.1177/1933719112450336. Epub 2012/08/21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Leik CE, Walsh SW. Neutrophils infiltrate resistance-sized vessels of subcutaneous fat in women with preeclampsia. Hypertension. 2004;44(1):72–7. https://doi.org/10.1161/01.HYP.0000130483.83154.37. Epub 2004/05/19.

    Article  CAS  PubMed  Google Scholar 

  169. Estrada-Gutierrez G, Cappello RE, Mishra N, Romero R, Strauss JF 3rd, Walsh SW. Increased expression of matrix metalloproteinase-1 in systemic vessels of preeclamptic women: a critical mediator of vascular dysfunction. Am J Pathol. 2011;178(1):451–60. https://doi.org/10.1016/j.ajpath.2010.11.003. Epub 2011/01/13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Mousa AA, Cappello RE, Estrada-Gutierrez G, Shukla J, Romero R, Strauss JF 3rd, et al. Preeclampsia is associated with alterations in DNA methylation of genes involved in collagen metabolism. Am J Pathol. 2012;181(4):1455–63. https://doi.org/10.1016/j.ajpath.2012.06.019. Epub 2012/08/07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Walsh SW, Nugent WH, Al Dulaimi M, Washington SL, Dacha P, Strauss JF 3rd. Proteases activate pregnancy neutrophils by a protease-activated receptor 1 pathway: epigenetic implications for preeclampsia. Reprod Sci. 2020;27(11):2115–27. https://doi.org/10.1007/s43032-020-00232-4. Epub 2020/06/17.

    Article  CAS  PubMed  Google Scholar 

  172. Chavarria ME, Lara-Gonzalez L, Gonzalez-Gleason A, Garcia-Paleta Y, Vital-Reyes VS, Reyes A. Prostacyclin/thromboxane early changes in pregnancies that are complicated by preeclampsia. Am J Obstet Gynecol. 2003;188(4):986–92. https://doi.org/10.1067/mob.2003.203. Epub 2003/04/25.

    Article  CAS  PubMed  Google Scholar 

  173. Mills JL, DerSimonian R, Raymond E, Morrow JD, Roberts LJ 2nd, Clemens JD, et al. Prostacyclin and thromboxane changes predating clinical onset of preeclampsia: a multicenter prospective study. JAMA. 1999;282(4):356–62. https://doi.org/10.1001/jama.282.4.356. Epub 1999/08/04.

    Article  CAS  PubMed  Google Scholar 

  174. Mousa AA, Strauss JF 3rd, Walsh SW. Reduced methylation of the thromboxane synthase gene is correlated with its increased vascular expression in preeclampsia. Hypertension. 2012;59(6):1249–55. https://doi.org/10.1161/HYPERTENSIONAHA.111.188730. Epub 2012/04/12.

    Article  CAS  PubMed  Google Scholar 

  175. Nguyen H, Chiasson VL, Chatterjee P, Kopriva SE, Young KJ, Mitchell BM. Interleukin-17 causes rho-kinase-mediated endothelial dysfunction and hypertension. Cardiovasc Res. 2013;97(4):696–704. https://doi.org/10.1093/cvr/cvs422. Epub 2012/12/25.

    Article  CAS  PubMed  Google Scholar 

  176. Walsh SW, Nugent WH, Archer KJ, Al Dulaimi M, Washington SL, Strauss JF 3rd. Epigenetic regulation of interleukin-17-related genes and their potential roles in neutrophil vascular infiltration in preeclampsia. Reprod Sci. 2022;29(1):154–62. https://doi.org/10.1007/s43032-021-00605-3. Epub 2021/05/08.

    Article  CAS  PubMed  Google Scholar 

  177. Nomura Y, Lambertini L, Rialdi A, Lee M, Mystal EY, Grabie M, et al. Global methylation in the placenta and umbilical cord blood from pregnancies with maternal gestational diabetes, preeclampsia, and obesity. Reprod Sci. 2014;21(1):131–7. https://doi.org/10.1177/1933719113492206. Epub 2013/06/15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Kazmi N, Sharp GC, Reese SE, Vehmeijer FO, Lahti J, Page CM, et al. Hypertensive disorders of pregnancy and DNA methylation in newborns. Hypertension. 2019;74(2):375–83. https://doi.org/10.1161/HYPERTENSIONAHA.119.12634. Epub 2019/06/25.

    Article  CAS  PubMed  Google Scholar 

  179. Ching T, Ha J, Song MA, Tiirikainen M, Molnar J, Berry MJ, et al. Genome-scale hypomethylation in the cord blood dnas associated with early onset preeclampsia. Clin Epigenetics. 2015;7(1):21. https://doi.org/10.1186/s13148-015-0052-x. Epub 2015/03/26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Mansell T, Ponsonby AL, Collier F, Burgner D, Vuillermin P, Lange K, et al. Genetic variation, intrauterine growth, and adverse pregnancy conditions predict leptin gene DNA methylation in blood at birth and 12 months of age. Int J Obes (Lond). 2020;44(1):45–56. https://doi.org/10.1038/s41366-019-0472-3. Epub 2019/10/23.

    Article  CAS  PubMed  Google Scholar 

  181. Mansell T, Ponsonby AL, Januar V, Novakovic B, Collier F, Burgner D, et al. Early-life determinants of hypoxia-inducible factor 3a gene (Hif3a) methylation: a birth cohort study. Clin Epigenetics. 2019;11(1):96. https://doi.org/10.1186/s13148-019-0687-0. Epub 2019/07/03.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Novielli C, Mando C, Tabano S, Anelli GM, Fontana L, Antonazzo P, et al. Mitochondrial DNA content and methylation in fetal cord blood of pregnancies with placental insufficiency. Placenta. 2017;55:63–70. https://doi.org/10.1016/j.placenta.2017.05.008. Epub 2017/06/19.

    Article  CAS  PubMed  Google Scholar 

  183. Hu W, Weng X, Dong M, Liu Y, Li W, Huang H. Alteration in methylation level at 11beta-hydroxysteroid dehydrogenase type 2 gene promoter in infants born to preeclamptic women. BMC Genet. 2014;15:96. https://doi.org/10.1186/s12863-014-0096-5. Epub 2014/09/10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Kraus TF, Guibourt V, Kretzschmar HA. 5-Hydroxymethylcytosine, the “Sixth Base”, during brain development and ageing. J Neural Transm (Vienna). 2015;122(7):1035–43. https://doi.org/10.1007/s00702-014-1346-4. Epub 2014/12/05.

    Article  CAS  PubMed  Google Scholar 

  185. Gao Q, Fan X, Xu T, Li H, He Y, Yang Y, et al. Promoter methylation changes and vascular dysfunction in pre-eclamptic umbilical vein. Clin Epigenetics. 2019;11(1):84. https://doi.org/10.1186/s13148-019-0685-2. Epub 2019/05/30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Ingram DA, Mead LE, Tanaka H, Meade V, Fenoglio A, Mortell K, et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood. 2004;104(9):2752–60. https://doi.org/10.1182/blood-2004-04-1396. Epub 2004/07/01.

    Article  CAS  PubMed  Google Scholar 

  187. Urbich C, Dimmeler S. Endothelial progenitor cells functional characterization. Trends Cardiovasc Med. 2004;14(8):318–22. https://doi.org/10.1016/j.tcm.2004.10.001. Epub 2004/12/15.

    Article  CAS  PubMed  Google Scholar 

  188. Brodowski L, Zindler T, von Hardenberg S, Schroder-Heurich B, von Kaisenberg CS, Frieling H, et al. Preeclampsia-associated alteration of DNA methylation in fetal endothelial progenitor cells. Front Cell Dev Biol. 2019;7:32. https://doi.org/10.3389/fcell.2019.00032. Epub 2019/04/06.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Faulk C, Dolinoy DC. Timing is everything: the when and how of environmentally induced changes in the epigenome of animals. Epigenetics. 2011;6(7):791–7. https://doi.org/10.4161/epi.6.7.16209. Epub 2011/06/04.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of in utero and early-life conditions on adult health and disease. N Engl J Med. 2008;359(1):61–73. https://doi.org/10.1056/NEJMra0708473. Epub 2008/07/04.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Gao Q, Tang J, Chen J, Jiang L, Zhu X, Xu Z. Epigenetic code and potential epigenetic-based therapies against chronic diseases in developmental origins. Drug Discov Today. 2014;19(11):1744–50. https://doi.org/10.1016/j.drudis.2014.05.004. Epub 2014/06/01.

    Article  CAS  PubMed  Google Scholar 

  192. West NA, Crume TL, Maligie MA, Dabelea D. Cardiovascular risk factors in children exposed to maternal diabetes in Utero. Diabetologia. 2011;54(3):504–7. https://doi.org/10.1007/s00125-010-2008-1. Epub 2010/12/15.

    Article  CAS  PubMed  Google Scholar 

  193. Cheong JN, Wlodek ME, Moritz KM, Cuffe JS. Programming of maternal and offspring disease: impact of growth restriction, fetal sex and transmission across generations. J Physiol. 2016;594(17):4727–40. https://doi.org/10.1113/JP271745. Epub 2016/03/13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Suzuki M, Maekawa R, Patterson NE, Reynolds DM, Calder BR, Reznik SE, et al. Amnion as a surrogate tissue reporter of the effects of maternal preeclampsia on the fetus. Clin Epigenetics. 2016;8:67. https://doi.org/10.1186/s13148-016-0234-1. Epub 2016/06/14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Perak AM, Lancki N, Kuang A, Labarthe DR, Allen NB, Shah SH, et al. Associations of maternal cardiovascular health in pregnancy with offspring cardiovascular health in early adolescence. JAMA. 2021;325(7):658–68. https://doi.org/10.1001/jama.2021.0247. Epub 2021/02/17.

    Article  PubMed  Google Scholar 

  196. Julian CG, Pedersen BS, Salmon CS, Yang IV, Gonzales M, Vargas E, et al. Unique DNA methylation patterns in offspring of hypertensive pregnancy. Clin Transl Sci. 2015;8(6):740–5. https://doi.org/10.1111/cts.12346. Epub 2015/11/08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Guan X, Dan GR, Yang Y, Ji Y, Lai WJ, Wang FJ, et al. Prenatal inflammation exposure-programmed hypertension exhibits multi-generational inheritance via disrupting DNA methylome. Acta Pharmacol Sin. 2022;43(6):1419–29. https://doi.org/10.1038/s41401-021-00772-8. Epub 2021/10/02.

    Article  CAS  PubMed  Google Scholar 

  198. Hofsink N, Dijkstra DJ, Stojanovska V, Scherjon SA, Plosch T. Preeclampsia-induced alterations in brain and liver gene expression and DNA methylation patterns in fetal mice. J Dev Orig Health Dis. 2023;14(1):146–51. https://doi.org/10.1017/S2040174422000344. Epub 2022/06/25.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Home for Researchers editorial team (www.home-for-researchers.com) for language editing service and Figure drawing (Figdraw. 2.0).

Funding

Supported partly by the Ministry of Science and Technology (2019YFA0802600), National Nature and Science Foundation of China (82271724, 82271712, and 81873841), Suzhou City “Wei Sheng Ren Cai (GSWS2019029)” program, General Programs of Jiangsu Commission of Health (M2021087), and Nature and Science Foundation of Jiangsu (BK20221243).

Author information

Authors and Affiliations

Authors

Contributions

QG, FD, MZ and JQ conceived, designed and wrote the manuscript. JL, MS, XW, CZ and ML help to revised manuscript. All authors reviewed and gave approval of the final draft.

Corresponding authors

Correspondence to Miao Sun, Meihua Zhang or Qinqin Gao.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deng, F., Lei, J., Qiu, J. et al. DNA methylation landscape in pregnancy-induced hypertension: progress and challenges. Reprod Biol Endocrinol 22, 77 (2024). https://doi.org/10.1186/s12958-024-01248-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-024-01248-0

Keywords