You are on page 1of 8

Placenta xxx (2017) 1e8

Contents lists available at ScienceDirect

Placenta
journal homepage: www.elsevier.com/locate/placenta

Notch signalling in placental development and gestational diseases


er*
S. Haider, J. Pollheimer, M. Kno
Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria

a r t i c l e i n f o a b s t r a c t

Article history: Activation of Notch signalling upon cell-cell contact of neighbouring cells controls a plethora of cellular
Received 13 December 2016 processes such as stem cell maintenance, cell lineage determination, cell proliferation, and survival.
Received in revised form Accumulating evidence suggests that the pathway also critically regulates these events during placental
9 January 2017
development and differentiation. Herein, we summarize our present knowledge about Notch signalling
Accepted 12 January 2017
in murine and human placentation and discuss its potential role in the pathophysiology of gestational
disorders. Studies in mice suggest that Notch controls trophectoderm formation, decidualization,
Keywords:
placental branching morphogenesis and endovascular trophoblast invasion. In humans, the particular
Placenta
Trophoblast
signalling cascade promotes formation of the extravillous trophoblast lineage and regulates trophoblast
Notch signalling proliferation, survival and differentiation. Expression patterns as well as functional analyses indicate
Gestational diseases distinct roles of Notch receptors in different trophoblast subtypes. Altered effects of Notch signalling
have been detected in choriocarcinoma cells, consistent with its role in cancer development and pro-
gression. Moreover, deregulation of Notch signalling components were observed in pregnancy disorders
such as preeclampsia and fetal growth restriction. In summary, Notch plays fundamental roles in
different developmental processes of the placenta. Abnormal signalling through this pathway could
contribute to the pathogenesis of gestational diseases with aberrant placentation and trophoblast
function.
2017 Elsevier Ltd. All rights reserved.

1. General role of Notch signalling intestinal stem cells and balances the cell fate of terminally
differentiating gut cells [3]. Interestingly, Notch was shown to have
Notch represents an evolutionarily conserved signalling an opposed role in intestinal stem cells of Drosophila, impairing
pathway critically involved in embryonic development and tissue their self-renewal. Hence, Notch signalling is highly complex and
homeostasis of adult organs. It controls a vast range of cellular depends on the cellular context as well as on its cross-talk with
processes including proliferation, survival, differentiation and other developmental signalling cascades mediated through mem-
motility. Signalling through Notch requires direct cell-cell contact bers of the Wingless (Wnt), epidermal growth factor (EGF) or
allowing for short-range communication between neighbouring transforming growth factor-b (TGF-b) family [4,5]. Conditions of the
cells [1]. Hence, the pathway is ideally suited to control self- particular microenvironment such as composition of the extracel-
renewal and differentiation in stem cell niches [2,3]. In the latter, lular matrix, shear stress or hypoxia also determine the various
Notch was shown to mediate binary cell fate decisions: different effects of Notch in cells and tissues [4]. Accordingly, Notch signal-
levels of Notch activation are created in two initially equipotent, ling components may act as tumour suppressors or oncogenes in
adjacent precursor cells thereby promoting adoption of distinct cell different organs and differentially inuence cancer development
fates. For example, in the developing mouse brain Notch inhibited and progression [6e8]. Considering its regulatory role in de novo
neuronal differentiation, but induced a glial cell fate and promoted vessel formation, angiogenesis and branching morphogenesis
astrocyte differentiation [2]. Likewise, Notch signalling is crucial for [9e11], it may not be surprising that Notch also controls these
epithelial stemness and cell fate specication. It maintains murine processes during placental development [12]. The present review
summarises our current knowledge about Notch signalling in mu-
rine and human placentation and discusses its role in the context of
* Corresponding author. Medical University of Vienna, Department of Obstetrics gestational diseases and choriocarcinoma cell function.
and Gynaecology, Reproductive Biology Unit, Waehringer Guertel 18-20, A-1090
Vienna, Austria.
E-mail address: martin.knoeer@meduniwien.ac.at (M. Kno er).

http://dx.doi.org/10.1016/j.placenta.2017.01.117
0143-4004/ 2017 Elsevier Ltd. All rights reserved.

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
2 S. Haider et al. / Placenta xxx (2017) 1e8

1.1. Canonical Notch signalling at two different cleavage sites in the TMD. These proteolytic steps
release NICD from the membrane allowing it to translocate into the
Canonical Notch signalling is activated upon interaction of a nucleus where it acts as a co-activator of transcription. Besides a
membrane-anchored ligand on the signal-sending cell with a Notch transactivation domain, NICD comprises a RAM (RBPJk-associated
receptor on the adjacent signal-receiving cell [1,13]. In contrast, module) domain for high afnity binding of the transcription factor
binding of the ligand to a receptor on the same cell was shown to recombination signal binding protein for immunoglobulin kappa J
inhibit the pathway [14]. In mice and humans four different Notch (RBPJk) as well as ankyrin repeats for RBPJk activation through
receptors (Notch1-4) and ve distinct Notch ligands, the Serrate- binding of co-activators of the mastermind-like (MAML) family. In
like ligands (Jagged1 and 2) and the Delta-like ligands (DLL1, 3 the absence of NICD RBPJk operates as a repressor recruiting his-
and 4) have been identied [1]. Notch receptors are single trans- tone deacetylases and other co-repressors such as CtBP (C-termi-
membrane proteins. After translation the Notch polypeptide chain nal-binding protein 1), CIR (CBF1 interacting co-repressor) or
is cleaved by furin-like convertases generating a heterodimer of the SHARP (SMRT and HDAC associated repressor protein) [5]. Upon
Notch extracellular domain (NECD) non-covalently linked to the Notch activation these repressors are displaced by NICD thereby
Notch transmembrane and intracellular domain (NTM-ICD) (Fig. 1). activating Notch target genes after binding of MAML proteins and
NECD harbours epidermal growth factor (EGF)-like repeats neces- further co-activators [17]. Activity of canonical Notch signalling is
sary for ligand interaction. These repeats can be modied by regulated by multiple mechanisms controlling post-translational
different types of glycosylation thereby governing differential Notch modication, receptor availability at the membrane, and
ligand binding and receptor activation [15]. expression of soluble Notch ligands, discussed elsewhere [1,13].
NTM-ICD contains a trans-membrane domain (TMD) and The expression of Notch targets strictly depends on the cell- and
different intracellular protein motifs required for the nuclear tissue-specic role of the pathway. Whereas NICD induces prolif-
function of the Notch intracellular domain (NICD). The latter arises eration by directly activating myc, cyclin D1 and CDK5, it promotes
after two sequential cleavages from the receptor [1]. Upon inter- growth arrest and differentiation by inducing kinase inhibitors
action of Notch with its ligand, members of the ADAM family, such such as p21 [18e20]. However, the prime targets of canonical Notch
as ADAM10 or ADAM17, cleave NTM-ICD 12 amino acids before the signalling are members of the Hairy/Enhancer of Split (HES) family,
trans-membrane domain thereby generating the intermediary HES1, HES5, HES7 and Hairy/Enhancer-of-split related with YRPW
membrane protein Notch extracellular truncation (NEXT). Subse- motif (HEY) proteins, HEY1, HEY2 and HEYL [21]. HES and HEY
quently, NEXT is recognized by g-secretase [16], cutting the protein proteins are basic helix loop helix proteins (bHLH) acting as

Fig. 1. The canonical Notch pathway. Membrane-anchored Notch ligands contain a transmembrane domain (TDM), a cysteine (C)-rich domain and EGF-like repeats for binding to
Notch receptors. Moreover, the N-terminal DSL (Delta/Serrate/LAG-2 proteins) domain and the DOS (Delta and OSM-11-like proteins) motif contribute to receptor binding. In
contrast to Jagged 1 and Jagged 2, the C-rich region is absent from DLL1, whereas DLL3 and DLL4 additionally lack the DOS domain [1]. The co-ligands Delta-like homologue 1 and 2
(DLK1 and 2) harbour a DOS domain but do not contain a DSL motif. Depending on the receptor subtype, the Notch extracellular domain (NECD) contains differential numbers of
EGF-like repeats. In addition, NECD comprises cysteine-rich Lin12-Notch repeats (LNR) and a heterodimerization domain (HD) for binding to the Notch transmembrane and
intracellular domain (NTM-ICD). Upon Notch activation, cleavage by ADAM proteases generates the Notch extracellular truncation (NEXT) which is further processed by g-secretase
yielding Notch intracellular domain (NICD). The latter harbours a nuclear localisation sequence (NLS), the RBPJk-associated module (RAM) for binding to RBPJk and ankyrin (ANK)
repeats for interaction with mastermind-like (MAML) proteins. Nuclear translocation of NICD displaces co-repressors (Co-R) from RBPJk and recruits co-activators (Co-A) thereby
promoting transcription of canonical Notch target genes such as repressors of the HES and HEY family. Stability of NICD is regulated via ubiquitination of proline/glutamic acid/
serine/threonine-rich (PEST) motifs and proteosomal degradation.

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
S. Haider et al. / Placenta xxx (2017) 1e8 3

transcriptional repressors, particularly upon binding to co- oocyte survival and maturation [35e37]. Likewise, Notch is also
repressors of the Groucho/Transducin-like enhancer of split (TLE) involved in spermatogenesis [38]. Abnormal Notch activation has
family [22,23]. These protein complexes are critically involved in been noticed in ovarian and testicular cancer [39,40]. Functions of
the maintenance of stem and progenitor cells by repressing key individual Notch proteins and their cross-talk with other signalling
regulators of cell fate determination. pathways in germ cells are above the scope of this review and
Along those lines, the role of canonical Notch signalling in discussed elsewhere [41].
stemness and cell line lineage decisions is highly complex and
explained by different regulatory mechanisms [2,3,13]. In lateral 2.1. Notch signalling in blastocyst formation, implantation and
inhibition equivalent precursors undergo different cell fates since decidualization
high expression of a Notch ligand in a given cell activates the
pathway in a neighbouring cell. In the latter, NICD-RBPJk prevents Expression of Notch signalling components has been observed
Notch ligand expression through the induction of HES. As a in embryos commencing with the 4 cell-stage suggesting that the
consequence of subsequent feedback loops, the ligand-expressing pathway could be required for pre-implantation development [42].
cell is maintained in a Notch-inactive state whereas the activated Moreover, Notch expression in murine and human trophectoderm
cell adopts a different cell fate. Alternatively, Notch-dependent cell (TE) suggested that it could be involved in early lineage decisions
fate decisions are regulated by inhibitors of the NUMB family which [43,44]. However, homozygous mutation of RBPJk in mice did not
have been described as multifunctional adaptor proteins involved affect progression of zygotes to the blastocyst stage indicating that
in endocytosis and intracellular trafcking of membrane-bound Notch signalling might not be necessary for early embryonic
molecules [24]. NUMBs interact with proteins of the endocytic development [45]. On the other hand, RBPJk was shown to also
machinery, such as a-adaptin and Eps15, thereby promoting have Notch-independent roles, largely by acting as a transcriptional
internalisation and sequestration of Notch receptors. Moreover, repressor [46]. Indeed, a more recent study suggested that Notch1
NUMB was shown to antagonise canonical Notch signalling by ICD promotes expression of the key regulator of TE formation,
binding to NTM-ICD and membrane-tethered NICD, thus recruiting caudal-related homeobox transcription factor 2 (Cdx2), in
E3-ubiquitin ligases for proteasomal degradation. The adoption of conjunction with the Hippo signalling-dependent activator TEAD4
distinct cell fates is achieved by asymmetrical segregation of NUMB and converts outer blastomere cells to a TE cell fate [47].
during cell division of a progenitor, thereby creating two different Notch signalling also critically regulates implantation and
daughter cells with active and inactive Notch signalling, respec- decidualization [48]. Notch receptors and ligands are differentially
tively [13]. expressed during the menstrual cycle and have been detected in
the luminal and glandular epithelium of the non-pregnant uterus
1.2. Non-canonical Notch signalling as well as in stromal and endothelial cells [48]. Notch4 is markedly
expressed during the proliferative phase, whereas Jagged1, DLL4
Besides the canonical activation of RBPJk, full-length Notch re- and Notch1 are predominantly produced during the mid-secretory
ceptors as well as NICD exert non-canonical effects in cells and phase [49e52]. Changes in the expression of Notch1, DLL1 and
tissues in a ligand-dependent or -independent manner [25]. Notch Jagged1 were noticed in infertile women suggesting that levels of
has been initially identied as a cell adhesion molecule in some of the Notch receptors and ligands are critical for uterine
Drosophila and was also shown to promote cell-cell interactions in receptivity [53,54].
mammals independently of NICD generation [26]. Moreover, NTM- Indeed, precisely regulated levels of activated Notch1 are pivotal
ICD and/or membrane-tethered NICD bind signalling components, for successful implantation and decidualization [55]. Human
such as phosphatidylinositol-3-kinase (PI3K) and mammalian chorion gonadotrophin (hCG)-mediated induction of Notch1 was
target of rapamycin C2 (mTORC2) [27], promoting cell survival via shown to promote survival of uterine stromal cells and initiated
AKT (Fig. 2.) Notch-mediated activation of these kinases suppresses decidualization, whereas downregulation of Notch1 and up-
pro-apoptotic p53 or stabilizes X-linked inhibitor of apoptosis regulation of NUMB was required for completion of differentia-
(XIAP) [28,29]. Moreover, nuclear NICD interacts with a variety of tion [56,57]. Women suffering from endometriosis have decreased
non-canonical transcription factors, for example nuclear factor kB levels of Notch1, Notch4, Jagged2 and DLL4 [58]. Moreover, over-
(NFkB), hypoxia-inducible factor 1a (HIF1a) or Ying Yang1 (YY1) expression of Notch1 ICD in the murine uterus causes infertility due
[30e32], promoting their nuclear retention and/or activation of to the lack of uterine glands and DNA-methyltransferase 3b-
Notch targets upon binding to RBPJk (Fig. 2). Notch enhances NFkB mediated hypermethylation of the progesterone receptor gene [59].
signalling at various levels [33]. In addition to direct binding of the During the rst trimester of pregnancy Notch2 was the predomi-
NFkB p52 subunit, NICD interacts with IkB kinase a (IKKa) pro- nant receptor in decidual stromal cells which additionally express
moting phosphorylation of inhibitor of kB (IkB). Degradation of the Jagged1, DLL1 and DLL4, whereas decidual glands produce all Notch
latter allows for nuclear translocation of the active transcription receptors and ligands [60]. Disruption of Notch2 impaired expres-
factor and expression of its target genes. Notch's cross-talk to Wnt sion of the differentiation markers prolactin (PRL) and insulin-like
signalling is mediated through binding of its central player, active growth factor binding protein 1 (IGFBP1) suggesting that the re-
b-catenin, to the ankyrin repeats of the full-length receptor. Sub- ceptor is crucial for decidualization [60]. Moreover, different re-
sequently, Notch promotes lysosomal degradation of active b-cat- ceptors and ligands were detected on uterine NK (uNK) cells and
enin involving NUMB-mediated endocytosis thereby antagonizing DLL1 and DLL4 increased interferon-g secretion [60,61].
canonical Wnt signalling [34].
2.2. Notch signalling components control murine placental
2. Notch signalling in reproductive tissues development

Besides other pathways Notch-mediated cell-cell communica- Regulation of angiogenesis represents one of the best charac-
tion plays a crucial role in the development of reproductive organs terised functions of Notch signalling. DLL4-Notch determines the
and their homeostasis in adults. In females, the particular signalling cell fate of endothelial precursors towards an arterial or venous
cascade controls formation of the reproductive tract, folliculo- phenotype and controls specication of endothelial tips and stalk
genesis, proliferation of granulosa cells during oogenesis, as well as cells as well as vessel stability and sprouting [11]. Likewise,

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
4 S. Haider et al. / Placenta xxx (2017) 1e8

Fig. 2. Non-canonical effects of Notch. Full-length Notch receptor, NTM-ICD and NICD provide a binding interface for multiple proteins exhibiting transcriptional or kinase activity.
Notch increases cell survival by activating PI3K/AKT/mTOR signalling and negatively affects Wnt-b-catenin signalling by triggering degradation of active b-catenin. Nuclear NICD
interacts with different transcription factors such as HIF1a or YY1 in a complex with RBPJk, bound to its cognate sequence, thereby integrating different environmental signals.
Furthermore, Notch promotes activation of NFkB through direct binding of its p52 subunit and indirectly by promoting phosphorylation and proteosomal degradation of IkB through
activation of IKKa.

homozygous mutations of Notch and its target genes mainly affect mediated vessel remodelling and differentiation of the above
the murine placental vascular system and/or branching morpho- mentioned TGC subtypes [72]. A similar phenotype was observed in
genesis of chorionic villi which goes in hand with vessel sprouting TLE3 mutant mice suggesting that Notch2 and TLE3 operate in the
[12]. Disruption of Notch1, Notch1/Notch4, Dll4, Hey1, Hey2 or same pathway [75].
RBPJk inhibited chorio-allantoic branching and/or formation of an
appropriate labyrinthine vascular network around midgestation 2.3. Notch signalling regulates human placental development
[62e67]. In addition, failures in chorion-allantoic fusion and
decreased numbers of proliferative trophoblasts have been re- Notch receptors and their ligands display trophoblast-subtype
ported in RBPJk mutant mice [62,68]. specic expression in the human placenta and also vary with
Like in invasive human EVTs [69], Notch2 seems to be the pre- gestational age and trophoblast differentiation [72,76,77]. Although
dominant receptor in diverse mouse trophoblast subtypes. Notch2 not functionally tested, expression of Notch1, DLL1, DLL4 and Jag-
has been detected in the chorion, the ectoplacental cone, in spon- ged1 in endothelial cells of villi suggests a role in placental vessel
giotrophoblasts as wells as in sinusoidal trophoblast giant cells formation and/or angiogenesis [77]. Akin to its expression in
(TGCs) and TGCs lining the maternal blood vessels [70e72]. Com- invasive mouse trophoblasts, Notch2 was predominantly detected
plementary expression of Notch2 and its putative ligands DLL1, in different extravillous trophoblast (EVT) subtypes of rst and
DLL4, Jagged1 or Jagged2 has been detected in adjacent cell layers second trimester tissues and increased during EVT formation
of the chorion, junctional zone and labyrinth, respectively, sug- in vivo and in vitro [69,72,76,77]. Interestingly, in distal HLA-G cell
gesting that the receptor could orchestrate different steps of column trophoblasts (CCTs) Notch2 was mainly detected at the cell
placental development [73]. Although the receptor may not play a membrane, whereas intramural EVTs of remodelling spiral arteries
role in trophoblast cell type specication, it is critically involved in additionally expressed nuclear Notch2 ICD [69]. Hence, it is antic-
establishing the placental-maternal circulation. Notch2 gene ipated that Notch2-dependent signalling is activated by adjacent
knock-out prevented formation of blood sinuses and caused maternal endothelial cells expressing DLL1, DLL4, Jagged1 and
delayed placental perfusion [70]. The role of Notch2 in murine Jagged2 in rst trimester decidual tissues [69]. Human Notch2 ICD
placentation was investigated in more detail using a homozygous might full a similar role as its murine counterpart being critical for
deletion of Notch2 in spongiotrophoblast precursors giving rise to endovascular trophoblast invasion and adaption of blood ow to
invasive trophoblast subtypes, spiral artery-associated TGCs and the placenta [72]. In contrast, disruption of Notch2 increased
canal TGCs [74]. Conditional deletion of the gene inhibited arterial in vitro motility in human primary trophoblast preparations con-
invasion and reduced the size of maternal blood canals as well as taining Notch2 distal cell column trophoblasts [69]. Therefore, we
perfusion indicating a crucial role for Notch2 in trophoblast- speculate that maintenance of column integrity by cell-cell

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
S. Haider et al. / Placenta xxx (2017) 1e8 5

adhesion could be the main function of Notch2 in the distal portion mainly expressed by CCT progenitors. Notch1 exerts its effects by
of the cell column [69]. direct binding to the myc gene as well as by up-regulating IRF6, a
Similar to other developing systems, the role of Notch signalling negative regulator of p63 stability [24,80,81]. Rapid down-
in human placentation is complex and likely depends on the spe- regulation of Notch1 during differentiation might involve NUMB
cic cellular context. Notch1, Notch3 and Notch4 have been proteins, of which different isoforms have been detected in human
detected in cytotrophoblast (CTB) progenitors whereas Notch2 and placenta [82].
Notch3 are expressed by invasive trophoblasts [72,76]. The overall
Notch activity decreased during in vitro EVT formation suggesting
that the pathway could be mainly associated with maintenance and 2.4. Notch signalling in gestational diseases and choriocarcinoma
stemness of trophoblast precursors [78]. Indeed, Notch1 was cells
recently shown to be critical for trophoblast survival and prolifer-
ation of villous CTBs and CCT progenitors [24]. Conversely, inhibi- Gestational diseases such as early-onset preeclampsia and se-
tion of g-secretase or silencing of RBPJk did not provoke apoptosis vere intrauterine growth restriction (IUGR) are associated with
and promoted outgrowth in explant cultures and, as a consequence, defects in trophoblast invasion, spiral artery remodelling and
EVT differentiation [24,78]. These contrasting results might be placental vascularisation [83,84]. Therefore, critical pathways of
explained by the Notch-independent roles of g-secretase and RBPJk placental development such as Notch signalling could be altered in
as well as the non-canonical functions of Notch1 ICD [79], again these pregnancy disorders. Indeed, several studies demonstrated
suggesting high complexity of the pathway and individual roles of changes of Notch receptor and ligand expression in preeclamptic
its components. RBPJk and total Notch activity could be necessary placentae using qPCR, western blotting or immunohistochemistry
to balance rates of cell column growth and EVT differentiation. Of [85e89]. However, the majority of these expression data are con-
note, Notch1, regulated by hypoxia, seems to be critical for initia- tradictory which could be explained by differences in placental
tion and maintenance of the EVT lineage [24]. Notch1 appears in sampling or ethnicity, the utilisation of inadequate antibodies,
clusters of vCTBs at the basal side of the placenta around the 6th technical issues in the experimental procedure or differences in
week of gestation and is specically expressed in a subset of pro- gestational age of placentae. Notch1-4, DLL1, DLL3, DLL4, Jagged1
liferative, proximal CCTs at 12th week. Besides inhibiting cell fusion and Jagged2 were shown to be decreased in preeclamptic tissues
and EVT formation, overexpression of Notch1 ICD in CTBs sup- [85,86,88,89], whereas others reported no changes of most Notch
pressed p63 and TEAD4, two markers of villous CTB self-renewal, receptors and ligands [72,89], or even an increase of Notch3 [89].
and induced the stemness genes, myc and VE-cadherin, which are Along those lines, immunohistochemical analyses suggested
Notch1 expression in CTBs and the syncytium of normal,

Fig. 3. The role of Notch in placental anchoring villi. Proximal cell column trophoblasts (CCTs) express Notch1 ICD promoting proliferation and cell survival. Active Notch1 also
induces the extravillous trophoblast (EVT) progenitor-specic stemness markers myc and VE-cadherin and suppresses villous cytotrophoblast (vCTB)-specic TEAD4 and p63
expression, the latter by up-regulating IRF6. Notch1 ICD also suppresses formation of the syncytium (S) and EVT differentiation. One could speculate that Notch1 might also have a
non-canonical role in proximal CCT by binding b-catenin to the membrane. Downregulation of Notch1 in EVTs might then liberate b-catenin from the receptor, thereby provoking its
nuclear recruitment and Wnt-dependent EVT differentiation and/or migration, as recently shown [95]. In contrast to Notch1, Notch2 is present in the distal cell column and in
intramural EVTs. In the latter Notch2-dependent signalling could be required for trophoblast-endothelial interactions promoting endovascular invasion, remodelling and/or short
range communication with immune cells. VS, villous stroma.

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
6 S. Haider et al. / Placenta xxx (2017) 1e8

preeclamptic or IUGR placentae obtained between the 24th week in cancer: it's NOTCH what you think, J. Exp. Med. 208 (10) (2011)
1931e1935.
and term [85,87e90]. However, at this time of pregnancy mRNA
[8] K. Forbes, B. Souquet, R. Garside, J.D. Aplin, M. Westwood, Transforming
and protein of the receptor are completely absent from all growth factor-{beta} (TGF{beta}) receptors I/II differentially regulate TGF
trophoblast subtypes [24,72]. Nevertheless, changes in methylation {beta}1 and IGF-binding protein-3 mitogenic effects in the human placenta,
of Notch4 and DLL1 were noticed in preeclamptic placentae sug- Endocrinology 151 (4) (2010) 1723e1731.
[9] K. Matsuzaki, Smad phospho-isoforms direct context-dependent TGF-beta
gesting epigenetic alterations of Notch components in this disease signaling, Cytokine Growth Factor Rev. 24 (4) (2013) 385e399.
[88]. Finally, analyses of Notch signalling in cultivated choriocar- [10] D.L. Bruce, G.P. Sapkota, Phosphatases in SMAD regulation, FEBS Lett. 586 (14)
cinoma cell lines suggested profound changes of its role in tumour (2012) 1897e1905.
[11] K. Matsuzaki, Smad phosphoisoform signaling specicity: the right place at
cells. In these studies Notch1 promoted cell migration but inhibited the right time, Carcinogenesis 32 (11) (2011) 1578e1588.
proliferation [91e93], whereas downregulation of Notch2 [12] M. Gasperowicz, F. Otto, The notch signalling pathway in the development of
increased cell growth [94]. These effects have not been observed the mouse placenta, Placenta 29 (8) (2008) 651e659.
[13] M.E. Fortini, Notch signaling: the core pathway and its posttranslational
upon manipulation of total Notch activity, Notch1 or Notch2 in regulation, Dev. Cell. 16 (5) (2009) 633e647.
primary CTBs [24,76]. [14] J. Prossler, Q. Chen, L. Chamley, J.L. James, The relationship between TGFbeta,
low oxygen and the outgrowth of extravillous trophoblasts from anchoring
villi during the rst trimester of pregnancy, Cytokine 68 (1) (2014) 9e15.
3. Summary [15] I. Caniggia, H. Mostach, J. Winter, M. Gassmann, S.J. Lye, M. Kuliszewski,
M. Post, Hypoxia-inducible factor-1 mediates the biological effects of oxygen
In conclusion, Notch signalling is a critical regulator of murine on human trophoblast differentiation through TGFbeta(3), J. Clin. Invest 105
(5) (2000) 577e587.
and human placental development. In mice, different Notch
[16] R. Kopan, M.X. Ilagan, Gamma-secretase: proteasome of the membrane? Nat.
pathway members control trophectoderm formation, angiogenesis, Rev. Mol. Cell Biol. 5 (6) (2004) 499e504.
vessel formation, giant cell differentiation and endovascular [17] A.S. McElhinny, J.L. Li, L. Wu, Mastermind-like transcriptional co-activators:
emerging roles in regulating cross talk among multiple signaling pathways,
trophoblast invasion. In the human placenta, Notch plays a crucial
Oncogene 27 (38) (2008) 5138e5147.
role in cell column formation, CCT survival and EVT differentiation [18] A. Rangarajan, C. Talora, R. Okuyama, M. Nicolas, C. Mammucari, H. Oh,
(Fig. 3). Despite recent progress much remains to be learned about J.C. Aster, S. Krishna, D. Metzger, P. Chambon, L. Miele, M. Aguet, F. Radtke,
Notch in human placentation and trophoblast development. Future G.P. Dotto, Notch signaling is a direct determinant of keratinocyte growth
arrest and entry into differentiation, EMBO J. 20 (13) (2001) 3427e3436.
studies should address epigenetic regulation of pathway members [19] C.H. Graham, J.J. Lysiak, K.R. McCrae, P.K. Lala, Localization of transforming
as well as the role of individual Notch ligands and receptors which growth factor-beta at the human fetal-maternal interface: role in trophoblast
have not yet been functionally tested in the human placenta. growth and differentiation, Biol. Reprod. 46 (4) (1992) 561e572.
[20] A.P. Weng, J.M. Millholland, Y. Yashiro-Ohtani, M.L. Arcangeli, A. Lau, C. Wai,
Furthermore, the cross-talk of Notch to other developmental C. Del Bianco, C.G. Rodriguez, H. Sai, J. Tobias, Y. Li, M.S. Wolfe, C. Shachaf,
pathways such as Hippo, EGF, TGF-b and Wnt signalling in CTBs D. Felsher, S.C. Blacklow, W.S. Pear, J.C. Aster, c-Myc is an important direct
should be elucidated. target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma, Genes
Dev. 20 (15) (2006) 2096e2109.
[21] I. Caniggia, S.J. Lye, J.C. Cross, Activin is a local regulator of human cyto-
Author's contributions trophoblast cell differentiation, Endocrinology 138 (9) (1997) 3976e3986.
[22] S. Chauvin, Y. Yinon, J. Xu, L. Ermini, J. Sallais, A. Tagliaferro, T. Todros, M. Post,
I. Caniggia, Aberrant TGFbeta signalling contributes to dysregulation of
S.H. drew the illustrations and provided support with respect to sphingolipid metabolism in intrauterine growth restriction, J. Clin. Endocrinol.
literature search and structure of the review. J.P. contributed to Metab. 100 (7) (2015) E986eE996.
writing and improvement of graphical design. M.K. wrote the [23] J. Xu, T. Sivasubramaniyam, Y. Yinon, A. Tagliaferro, J. Ray, O. Nevo, M. Post,
I. Caniggia, Aberrant TGFbeta signaling contributes to altered trophoblast
manuscript.
differentiation in preeclampsia, Endocrinology 157 (2) (2016) 883e899.
[24] S. Haider, G. Meinhardt, L. Saleh, C. Fiala, J. Pollheimer, M. Kno er, Notch1
Disclosure statement controls development of the extravillous trophoblast lineage in the human
placenta, Proc. Natl. Acad. Sci. U. S. A. 113 (48) (2016). E7710eE9.
[25] J.C. Cheng, H.M. Chang, P.C. Leung, Transforming growth factor-beta1 inhibits
The authors have nothing to declare. trophoblast cell invasion by inducing Snail-mediated down-regulation of
vascular endothelial-cadherin protein, J. Biol. Chem. 288 (46) (2013)
Conict of interest 33181e33192.
[26] A. Murata, M. Yoshino, M. Hikosaka, K. Okuyama, L. Zhou, S. Sakano, H. Yagita,
S. Hayashi, An evolutionary-conserved function of Mammalian notch family
The authors do not report any conict of interest. members as cell adhesion molecules, PLoS One 9 (9) (2014) e108535.
[27] C. Zeng, R. Xing, J. Liu, F. Xing, Role of CSL-dependent and independent Notch
signaling pathways in cell apoptosis, Apoptosis 21 (1) (2016) 1e12.
Acknowledgements [28] S.K. Mungamuri, X. Yang, A.D. Thor, K. Somasundaram, Survival signaling by
Notch1: mammalian target of rapamycin (mTOR)-dependent inhibition of
Research delineating signalling pathways in human trophoblast p53, Cancer Res. 66 (9) (2006) 4715e4724.
[29] E. Rosati, R. Sabatini, G. Rampino, A. Tabilio, M. Di Ianni, K. Fettucciari,
development and trophoblast differentiation were supported by A. Bartoli, S. Coaccioli, I. Screpanti, P. Marconi, Constitutively activated Notch
grant P-28417-B30 and P-25187-B13 of the Austrian Science Funds, signaling is involved in survival and apoptosis resistance of B-CLL cells, Blood
Vienna, Austria and by grant 16517 of the Austrian National Bank. 113 (4) (2009) 856e865.
[30] Y. Shi, J. Massague, Mechanisms of TGF-beta signaling from cell membrane to
the nucleus, Cell 113 (6) (2003) 685e700.
References [31] H.M. Shin, L.M. Minter, O.H. Cho, S. Gottipati, A.H. Fauq, T.E. Golde,
G.E. Sonenshein, B.A. Osborne, Notch1 augments NF-kappaB activity by
[1] R. Kopan, M.X. Ilagan, The canonical Notch signaling pathway: unfolding the facilitating its nuclear retention, EMBO J. 25 (1) (2006) 129e138.
activation mechanism, Cell 137 (2) (2009) 216e233. [32] H. Wang, J. Zou, B. Zhao, E. Johannsen, T. Ashworth, H. Wong, W.S. Pear,
[2] U. Koch, R. Lehal, F. Radtke, Stem cells living with a Notch, Development 140 J. Schug, S.C. Blacklow, K.L. Arnett, B.E. Bernstein, E. Kieff, J.C. Aster, Genome-
(4) (2013) 689e704. wide analysis reveals conserved and divergent features of Notch1/RBPJ
[3] C.N. Perdigoto, A.J. Bardin, Sending the right signal: Notch and stem cells, binding in human and murine T-lymphoblastic leukemia cells, Proc. Natl.
Biochim. Biophys. Acta 1830 (2) (2013) 2307e2322. Acad. Sci. U. S. A. 108 (36) (2011) 14908e14913.
[4] J. Massague, J. Seoane, D. Wotton, Smad transcription factors, Genes Dev. 19 [33] C. Osipo, T.E. Golde, B.A. Osborne, L.A. Miele, Off the beaten pathway: the
(23) (2005) 2783e2810. complex cross talk between Notch and NF-kappaB, Lab. Invest 88 (1) (2008)
[5] J. Massague, How cells read TGF-beta signals, Nat. Rev. Mol. Cell Biol. 1 (3) 11e17.
(2000) 169e178. [34] C. Kwon, P. Cheng, I.N. King, P. Andersen, L. Shenje, V. Nigam, D. Srivastava,
[6] U. Koch, F. Radtke, Notch and cancer: a double-edged sword, Cell Mol. Life Sci. Notch post-translationally regulates beta-catenin protein in stem and pro-
64 (21) (2007) 2746e2762. genitor cells, Nat. Cell Biol. 13 (10) (2011) 1244e1251.
[7] C. Lobry, P. Oh, I. Aifantis, Oncogenic and tumor suppressor functions of Notch [35] L. Ferguson, E.M. Kaftanovskaya, C. Manresa, A.M. Barbara, R.J. Poppiti, Y. Tan,

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
S. Haider et al. / Placenta xxx (2017) 1e8 7

A.I. Agoulnik, Constitutive Notch signaling causes abnormal development of O. Mandelboim, Notch activation enhances IFNgamma secretion by human
the oviducts, abnormal angiogenesis, and cyst formation in mouse female peripheral blood and decidual NK cells, J. Reprod. Immunol. 84 (1) (2010) 1e7.
reproductive tract, Biol. Reprod. 94 (3) (2016) 67. [62] L.T. Krebs, J.R. Shutter, K. Tanigaki, T. Honjo, K.L. Stark, T. Gridley, Hap-
[36] D.A. Vanorny, R.D. Prasasya, A.J. Chalpe, S.M. Kilen, K.E. Mayo, Notch signaling loinsufcient lethality and formation of arteriovenous malformations in Notch
regulates ovarian follicle formation and coordinates follicular growth, Mol. pathway mutants, Genes Dev. 18 (20) (2004) 2469e2473.
Endocrinol. 28 (4) (2014) 499e511. [63] A. Fischer, N. Schumacher, M. Maier, M. Sendtner, M. Gessler, The Notch target
[37] C.P. Zhang, J.L. Yang, J. Zhang, L. Li, L. Huang, S.Y. Ji, Z.Y. Hu, F. Gao, Y.X. Liu, genes Hey1 and Hey2 are required for embryonic vascular development,
Notch signaling is involved in ovarian follicle development by regulating Genes Dev. 18 (8) (2004) 901e911.
granulosa cell proliferation, Endocrinology 152 (6) (2011) 2437e2447. [64] Y. Xue, X. Gao, C.E. Lindsell, C.R. Norton, B. Chang, C. Hicks, M. Gendron-
[38] T.X. Garcia, M.C. Hofmann, NOTCH signaling in Sertoli cells regulates gonocyte Maguire, E.B. Rand, G. Weinmaster, T. Gridley, Embryonic lethality and
fate, Cell Cycle 12 (16) (2013) 2538e2545. vascular defects in mice lacking the Notch ligand Jagged1, Hum. Mol. Genet. 8
[39] T. Hayashi, T. Yamada, Y. Kageyama, K. Kihara, Expression failure of the notch (5) (1999) 723e730.
signaling system is associated with the pathogenesis of testicular germ cell [65] A. Duarte, M. Hirashima, R. Benedito, A. Trindade, P. Diniz, E. Bekman, L. Costa,
tumor, Tumour Biol. 25 (3) (2004) 99e105. D. Henrique, J. Rossant, Dosage-sensitive requirement for mouse Dll4 in artery
[40] S.L. Rose, Notch signaling pathway in ovarian cancer, Int. J. Gynecol. Cancer 19 development, Genes Dev. 18 (20) (2004) 2474e2478.
(4) (2009) 564e566. [66] A. Limbourg, M. Ploom, D. Elligsen, I. Sorensen, T. Ziegelhoeffer, A. Gossler,
[41] A.V. Sirotkin, Growth factors controlling ovarian functions, J. Cell Physiol. 226 H. Drexler, F.P. Limbourg, Notch ligand Delta-like 1 is essential for postnatal
(9) (2011) 2222e2225. arteriogenesis, Circ. Res. 100 (3) (2007) 363e371.
[42] Q.T. Wang, K. Piotrowska, M.A. Ciemerych, L. Milenkovic, M.P. Scott, [67] F.P. Limbourg, K. Takeshita, F. Radtke, R.T. Bronson, M.T. Chin, J.K. Liao,
R.W. Davis, M. Zernicka-Goetz, A genome-wide study of gene activity reveals Essential role of endothelial Notch1 in angiogenesis, Circulation 111 (14)
developmental signaling pathways in the preimplantation mouse embryo, (2005) 1826e1832.
Dev. Cell 6 (1) (2004) 133e144. [68] C. Oka, T. Nakano, A. Wakeham, J.L. de la Pompa, C. Mori, T. Sakai, S. Okazaki,
[43] J. Adjaye, J. Huntriss, R. Herwig, A. BenKahla, T.C. Brink, C. Wierling, M. Kawaichi, K. Shiota, T.W. Mak, T. Honjo, Disruption of the mouse RBP-J
C. Hultschig, D. Groth, M.L. Yaspo, H.M. Picton, R.G. Gosden, H. Lehrach, Pri- kappa gene results in early embryonic death, Development 121 (10) (1995)
mary differentiation in the human blastocyst: comparative molecular por- 3291e3301.
traits of inner cell mass and trophectoderm cells, Stem Cells 23 (10) (2005) [69] K. Plessl, S. Haider, C. Fiala, J. Pollheimer, M. Kno er, Expression pattern and
1514e1525. function of Notch2 in different subtypes of rst trimester cytotrophoblast,
[44] S. Cormier, S. Vandormael-Pournin, C. Babinet, M. Cohen-Tannoudji, Devel- Placenta 36 (4) (2015) 365e371.
opmental expression of the Notch signaling pathway genes during mouse [70] Y. Hamada, T. Hiroe, Y. Suzuki, M. Oda, Y. Tsujimoto, J.R. Coleman, S. Tanaka,
preimplantation development, Gene Expr. Patterns 4 (6) (2004) 713e717. Notch2 is required for formation of the placental circulatory system, but not
[45] C. Souilhol, S. Cormier, K. Tanigaki, C. Babinet, M. Cohen-Tannoudji, RBP- for cell-type specication in the developing mouse placenta, Differentiation
Jkappa-dependent notch signaling is dispensable for mouse early embryonic 75 (3) (2007) 268e278.
development, Mol. Cell Biol. 26 (13) (2006) 4769e4774. [71] H. Nakayama, Y. Liu, S. Stifani, J.C. Cross, Developmental restriction of Mash-2
[46] R. Sanalkumar, S.B. Dhanesh, J. James, Non-canonical activation of Notch expression in trophoblast correlates with potential activation of the notch-2
signaling/target genes in vertebrates, Cell Mol. Life Sci. 67 (17) (2010) pathway, Dev. Genet. 21 (1) (1997) 21e30.
2957e2968. [72] N.M. Hunkapiller, M. Gasperowicz, M. Kapidzic, V. Plaks, E. Maltepe,
[47] T. Rayon, S. Menchero, A. Nieto, P. Xenopoulos, M. Crespo, K. Cockburn, J. Kitajewski, J.C. Cross, S.J. Fisher, A role for Notch signaling in trophoblast
S. Canon, H. Sasaki, A.K. Hadjantonakis, J.L. de la Pompa, J. Rossant, endovascular invasion and in the pathogenesis of pre-eclampsia, Develop-
M. Manzanares, Notch and hippo converge on Cdx2 to specify the tro- ment 138 (14) (2011) 2987e2998.
phectoderm lineage in the mouse blastocyst, Dev. Cell 30 (4) (2014) 410e422. [73] M. Gasperowicz, A. Rai, J.C. Cross, Spatiotemporal expression of Notch re-
[48] C. Cuman, E. Menkhorst, A. Winship, M. Van Sinderen, T. Osianlis, ceptors and ligands in developing mouse placenta, Gene Expr. Patterns 13 (7)
L.J. Rombauts, E. Dimitriadis, Fetal-maternal communication: the role of Notch (2013) 249e254.
signalling in embryo implantation, Reproduction 147 (3) (2014) R75eR86. [74] D.G. Simmons, A.L. Fortier, J.C. Cross, Diverse subtypes and developmental
[49] L. Cobellis, F. Caprio, E. Trabucco, A. Mastrogiacomo, G. Coppola, L. Manente, origins of trophoblast giant cells in the mouse placenta, Dev. Biol. 304 (2)
N. Colacurci, M. De Falco, A. De Luca, The pattern of expression of Notch (2007) 567e578.
protein members in normal and pathological endometrium, J. Anat. 213 (4) [75] M. Gasperowicz, C. Surmann-Schmitt, Y. Hamada, F. Otto, J.C. Cross, The
(2008) 464e472. transcriptional co-repressor TLE3 regulates development of trophoblast giant
[50] J. Mazella, S. Liang, L. Tseng, Expression of Delta-like protein 4 in the human cells lining maternal blood spaces in the mouse placenta, Dev. Biol. 382 (1)
endometrium, Endocrinology 149 (1) (2008) 15e19. (2013) 1e14.
[51] A. Mikhailik, J. Mazella, S. Liang, L. Tseng, Notch ligand-dependent gene [76] S. Haider, G. Meinhardt, P. Velicky, G.R. Otti, G. Whitley, C. Fiala, J. Pollheimer,
expression in human endometrial stromal cells, Biochem. Biophys. Res. M. Kno er, Notch signaling plays a critical role in motility and differentiation
Commun. 388 (3) (2009) 479e482. of human rst-trimester cytotrophoblasts, Endocrinology 155 (1) (2014)
[52] Y. Mitsuhashi, A. Horiuchi, T. Miyamoto, H. Kashima, A. Suzuki, T. Shiozawa, 263e274.
Prognostic signicance of Notch signalling molecules and their involvement [77] F. Herr, I. Schreiner, N. Baal, C. Pfarrer, M. Zygmunt, Expression patterns of
in the invasiveness of endometrial carcinoma cells, Histopathology 60 (5) Notch receptors and their ligands Jagged and Delta in human placenta,
(2012) 826e837. Placenta 32 (8) (2011) 554e563.
[53] C. Cuman, E.M. Menkhorst, L.J. Rombauts, S. Holden, D. Webster, M. Bilandzic, [78] P. Velicky, S. Haider, G.R. Otti, C. Fiala, J. Pollheimer, M. Kno er, Notch-
T. Osianlis, E. Dimitriadis, Preimplantation human blastocysts release factors dependent RBPJkappa inhibits proliferation of human cytotrophoblasts and
that differentially alter human endometrial epithelial cell adhesion and gene their differentiation into extravillous trophoblasts, Mol. Hum. Reprod. 20 (8)
expression relative to IVF success, Hum. Reprod. 28 (5) (2013) 1161e1171. (2014) 756e766.
[54] M. Van Sinderen, C. Cuman, T. Gamage, K. Rainczuk, T. Osianlis, L. Rombauts, [79] L.R. Perumalsamy, M. Nagala, P. Banerjee, A. Sarin, A hierarchical cascade
E. Dimitriadis, Localisation of the Notch family in the human endometrium of activated by non-canonical Notch signaling and the mTOR-Rictor complex
fertile and infertile women, J. Mol. Histol. 45 (6) (2014) 697e706. regulates neglect-induced death in mammalian cells, Cell Death Differ. 16 (6)
[55] P. Banerjee, A.T. Fazleabas, Endometrial responses to embryonic signals in the (2009) 879e889.
primate, Int. J. Dev. Biol. 54 (2e3) (2010) 295e302. [80] F. Moretti, B. Marinari, N. Lo Iacono, E. Botti, A. Giunta, G. Spallone, G. Garaffo,
[56] Y. Afshar, J.W. Jeong, D. Roqueiro, F. DeMayo, J. Lydon, F. Radtke, R. Radnor, E. Vernersson-Lindahl, G. Merlo, A.A. Mills, C. Ballaro, S. Alema, S. Chimenti,
L. Miele, A. Fazleabas, Notch1 mediates uterine stromal differentiation and is L. Guerrini, A. Costanzo, A regulatory feedback loop involving p63 and IRF6
critical for complete decidualization in the mouse, FASEB J. 26 (1) (2012) links the pathogenesis of 2 genetically different human ectodermal dysplasias,
282e294. J. Clin. Invest 120 (5) (2010) 1570e1577.
[57] Y. Afshar, L. Miele, A.T. Fazleabas, Notch1 is regulated by chorionic gonado- [81] G. Restivo, B.C. Nguyen, P. Dziunycz, E. Ristorcelli, R.J. Ryan, O.Y. Ozuysal, M. Di
tropin and progesterone in endometrial stromal cells and modulates Piazza, F. Radtke, M.J. Dixon, G.F. Hofbauer, K. Lefort, G.P. Dotto, IRF6 is a
decidualization in primates, Endocrinology 153 (6) (2012) 2884e2896. mediator of Notch pro-differentiation and tumour suppressive function in
[58] R.W. Su, M.R. Strug, N.R. Joshi, J.W. Jeong, L. Miele, B.A. Lessey, S.L. Young, keratinocytes, EMBO J. 30 (22) (2011) 4571e4585.
A.T. Fazleabas, Decreased Notch pathway signaling in the endometrium of [82] M. Haider, Q. Qiu, M. Bani-Yaghoub, B.K. Tsang, A. Gruslin, Characterization
women with endometriosis impairs decidualization, J. Clin. Endocrinol. and role of NUMB in the human extravillous trophopblast, Placenta 32 (6)
Metab. 100 (3) (2015) E433eE442. (2011) 441e449.
[59] R.W. Su, M.R. Strug, J.W. Jeong, L. Miele, A.T. Fazleabas, Aberrant activation of [83] S.J. Fisher, Why is placentation abnormal in preeclampsia? Am. J. Obstet.
canonical Notch1 signaling in the mouse uterus decreases progesterone re- Gynecol. 213 (4 Suppl) (2015) S115eS122.
ceptor by hypermethylation and leads to infertility, Proc. Natl. Acad. Sci. U. S. [84] R. Pijnenborg, J. Anthony, D.A. Davey, A. Rees, A. Tiltman, L. Vercruysse, A. van
A. 113 (8) (2016) 2300e2305. Assche, Placental bed spiral arteries in the hypertensive disorders of preg-
[60] er, Notch2 controls
G.R. Otti, L. Saleh, P. Velicky, C. Fiala, J. Pollheimer, M. Kno nancy, Br. J. Obstet. Gynaecol. 98 (7) (1991) 648e655.
prolactin and insulin-like growth factor binding protein-1 expression in [85] L. Cobellis, A. Mastrogiacomo, E. Federico, M.T. Schettino, M. De Falco,
decidualizing human stromal cells of early pregnancy, PLoS One 9 (11) (2014) L. Manente, G. Coppola, M. Torella, N. Colacurci, A. De Luca, Distribution of
e112723. Notch protein members in normal and preeclampsia-complicated placentas,
[61] I. Manaster, R. Gazit, D. Goldman-Wohl, N. Stern-Ginossar, S. Mizrahi, S. Yagel, Cell Tissue Res. 330 (3) (2007) 527e534.

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117
8 S. Haider et al. / Placenta xxx (2017) 1e8

[86] P. Fragkiadaki, N. Soulitzis, S. Sifakis, D. Koutroulakis, V. Gourvas, N. Vrachnis, [91] M. Massimiani, L. Vecchione, D. Piccirilli, P. Spitalieri, F. Amati, S. Salvi,
D.A. Spandidos, Downregulation of notch signaling pathway in late preterm S. Ferrazzani, H. Stuhlmann, L. Campagnolo, Epidermal growth factor-like
and term placentas from pregnancies complicated by preeclampsia, PLoS One domain 7 promotes migration and invasion of human trophoblast cells
10 (5) (2015) e0126163. through activation of MAPK, PI3K and NOTCH signaling pathways, Mol. Hum.
[87] Z. Sahin, N. Acar, O. Ozbey, I. Ustunel, R. Demir, Distribution of Notch family Reprod. 21 (5) (2015) 435e451.
proteins in intrauterine growth restriction and hypertension complicated [92] J. Wagener, W. Yang, K. Kazuschke, E. Winterhager, A. Gellhaus, CCN3 regu-
human term placentas, Acta Histochem. 113 (3) (2011 May) 270e276, http:// lates proliferation and migration properties in Jeg3 trophoblast cells via ERK1/
dx.doi.org/10.1016/j.acthis.2009.10.006. Epub 2009 Nov 12. 2, Akt and Notch signalling, Mol. Hum. Reprod. 19 (4) (2013) 237e249.
[88] Y. Shimanuki, H. Mitomi, Y. Fukumura, S. Makino, A. Itakura, T. Yao, S. Takeda, [93] Y. Yu, L. Wang, W. Tang, D. Zhang, T. Shang, RNA interference-mediated
Alteration of Delta-like ligand 1 and Notch 1 receptor in various placental knockdown of Notch-1 inhibits migration and invasion, down-regulates ma-
disorders with special reference to early onset preeclampsia, Hum. Pathol. 46 trix metalloproteinases and suppresses NF-kappaB signaling pathway in
(8) (2015) 1129e1137. trophoblast cells, Acta histochem. 116 (5) (2014) 911e919.
[89] W.X. Zhao, T.T. Huang, M. Jiang, R. Feng, J.H. Lin, Expression of notch family [94] W.X. Zhao, X. Zhuang, T.T. Huang, R. Feng, J.H. Lin, Effects of Notch2 and
proteins in placentas from patients with early-onset severe preeclampsia, Notch3 on cell proliferation and apoptosis of trophoblast cell lines, Int. J. Med.
Reprod. Sci. 21 (6) (2014) 716e723. Sci. 12 (11) (2015) 867e874.
[90] M. De Falco, L. Cobellis, D. Giraldi, A. Mastrogiacomo, A. Perna, N. Colacurci, [95] G. Meinhardt, S. Haider, P. Haslinger, K. Proestling, C. Fiala, J. Pollheimer,
L. Miele, A. De Luca, Expression and distribution of notch protein members in M. Kno er, Wnt-dependent T-cell factor-4 controls human etravillous
human placenta throughout pregnancy, Placenta 28 (2e3) (2007) 118e126. trophoblast motility, Endocrinology 155 (5) (2014) 1908e1920.

Please cite this article in press as: S. Haider, et al., Notch signalling in placental development and gestational diseases, Placenta (2017), http://
dx.doi.org/10.1016/j.placenta.2017.01.117

You might also like