Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Placenta 31 (2010) 951e957

Contents lists available at ScienceDirect

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

Current Opinion

Regulation of Hypoxia Inducible Factors (HIF) in Hypoxia and Normoxia During


Placental Development
J. Patel a, b, *, K. Landers b, R.H. Mortimer a, b, c, K. Richard a, b
a
School of Medicine, The University of Queensland, Royal Brisbane and Women’s Hospital, Herston, Queensland 4029, Australia
b
Conjoint Endocrine Laboratory, Pathology Queensland and Department of Endocrinology, Royal Brisbane and Women’s Hospital, Herston, Queensland 4029, Australia
c
Disciplines of Medicine, Obstetrics and Gynaecology, The University of Queensland, Herston, Queensland 4029, Australia

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

Article history: During the first trimester of pregnancy the human placenta develops in an hypoxic environment caused
Accepted 17 August 2010 by the occlusion of uterine spiral arterioles by extravillous trophoblasts (EVT). This period of low oxygen
tension is crucial for successful pregnancy. In low oxygen environments, Hypoxia Inducible Factors (HIF)
Keywords: are the main regulators in the transcription of a number of genes. Target genes can induce anaerobic
Hypoxia inducible factor processes, reducing oxygen consumption, or promote angiogenesis, which establishes and enhances the
Placenta
vascular environment. The HIFs can function throughout all stages of placental differentiation and
Oxygen
growth both in normal and pathological pregnancies (compromised by hypoxia/ischemia). Interestingly,
Trophoblast
HIFs respond to a multitude of changes during pregnancy, including 1) low oxygen, 2) renineangiotensin
system (RAS), 3) cytokines, and 4) growth factors, all of which regulate placental function. This review
explores oxygen-dependent and oxygen-independent regulation and the role of HIF in placental
development and differentiation.
Ó 2010 Elsevier Ltd. All rights reserved.

1. Introduction significant maternal blood flow and increasing oxygen levels (Fig. 1)
[8e10]. Recent in vitro studies have demonstrated the capacity of
The placenta is a remarkable organ derived from cells origi- EVTs to initiate apoptosis of vascular smooth muscle cells (VSMC)
nating in the extra-embryonic conceptus and maternal decidua and endothelial cells. This may represent the mechanism of the
that provides the conduit for materno-fetal transport/exchange of physiological modification of the uterine spiral arterioles that leads
nutrients and waste. Development and maturation of the placenta to the increased vascular compliance and circumference of early
is a complex process, requiring coordinated regulation of tropho- pregnancy (Fig. 1) [11e14]. The resulting increased blood flow and
blast invasion and proliferation and differentiation into the growth of the vascular and capillary network meets the demands of
maternal decidua [1e3]. This results in an adequate blood supply the growing fetus [15].
for continuing normal placental and fetal development. Impaired placental invasion and development during the first
In the first trimester, extravillous trophoblast (EVT) cells invade trimester can cause deleterious outcomes such as intrauterine
into the decidua, occluding uterine spiral arterioles (Fig. 1) [4]. This growth restriction (IUGR), preeclampsia, unexplained miscarriage,
restricts blood flow into the intervillous space (IVS) resulting in a low pre-term labour, placental abruption and stillbirth. Paradoxically,
oxygen environment that is essential for placental and embryonic excess blood flow early in pregnancy leads to reduced blood flow
development [3,5e7]. Measurements with oxygen sensitive probes later in gestation [16e18].
during ultrasonography at 8 weeks gestation have established that A low oxygen tension has been implicated as the key factor in the
the oxygen concentration within the IVS is <20 mmHg or 3e5% O2 process of placental invasion and development [3]. Although this
[8]. Oxygen concentrations within the underlying maternal decidua low oxygen tension is physiological at this stage of development it is
are approximately 60 mmHg or 8e10% O2. Between weeks 11e12 usually described as hypoxia and this review will use that term. All
of gestation, uterine spiral arterioles become patent, allowing cells respond to hypoxia with a series of adaptive modifications of
gene expression. This is particularly true in early pregnancy when
* Corresponding author. QIMR Bancroft Centre, 300 Herston Rd, Herston,
the feto-placental unit is developing in a low oxygen environment.
Queensland 4029, Australia. Tel.: þ617 3362 0496; fax: þ617 3636 8842. The Hypoxia Inducible Factors (HIFs) are the key mediators of this
E-mail address: jatin.patel@qimr.edu.au (J. Patel). process and facilitate placental vascularisation as well as signalling

0143-4004/$ e see front matter Ó 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.placenta.2010.08.008
952 J. Patel et al. / Placenta 31 (2010) 951e957

Fig. 1. (A) Demonstrates the narrow, small calibre nature of spiral arteries prior to pregnancy, which forms a capillary plexus below the uterine surface. (B) During the first trimester
of pregnancy invasion of endovascular EVT from the placental villi occurs. This leads to occlusion of the terminal portions of spiral arteries, creating a low oxygen environment
within the IVS (3e5% O2) that is essential for early placental and fetal development. (C) Late in the first trimester of pregnancy, the EVT invasion will cause changes in spiral artery
architecture, resulting in the formation of funnel-shaped flaccid arterioles with increased vascular compliance and circumference [8e13]. This results in increased oxygen levels
(8e10% O2) within the IVS for the rest of gestation. (Modified image by Moffet-King et al. [14]).

for trophoblast differentiation [3]. Interestingly, recent evidence 2. Hypoxia inducible factors (HIFs)
suggests that HIFs are also activated by non-hypoxic stimuli such as
the renineangiotensin system (RAS), growth factors and immuno- To maintain cell survival, higher eukaryotes have cellular
genic cytokines, all of which play important roles in the regulation of mechanisms to adjust to low oxygen tensions. These oxygen-
placental development and maturation [19]. However, further dependent pathways are conserved and expressed in almost every
insight into the control of HIF-1a regulation under normoxic mammalian cell type [20]. The transcription factors activated
conditions by needs to be elucidated, to provide additional and during low oxygen conditions are called Hypoxia Inducible Factors
definitive evidence for its role in the complexities of placentology. (HIF). They are hetero-dimeric consisting of two subunits, HIF-
In this review we investigate the role of the well-characterised a and aryl hydrocarbon receptor nuclear translocator (Arnt; also
oxygen-dependent pathways of HIF modulation in placental called HIF-1b) [21]. They bring about their effects through inter-
development and differentiation but also consider the roles of the action through two Per-Arnt-Sim (PAS) domains, bind DNA via N-
less well-characterised oxygen-independent pathways in HIF terminal basic Helix-Loop-Helix (bHLH) domains, and activate
regulation. transcription with C-terminal transcriptional transactivation
J. Patel et al. / Placenta 31 (2010) 951e957 953

domains (TADs) [22]. HIF-1b is constitutively expressed whereas


the activity and expression of HIF-a depends on cellular oxygen
concentrations. In mammalian cells there are three HIF-a genes
(HIF-1a, HIF-2a and HIF-3a). HIF-1a is expressed ubiquitously,
while the expression of HIF-2a and HIF-3a are more restricted [21].
HIF-1a and HIF-2a dimerise with HIF-1b forming HIF-1 and HIF-2,
both leading to activation of key transcription factors. HIF-3a is
found in three isoforms (HIF-3a, neonatal and embryonic PAS
(NEPAS) and inhibitory PAS protein (IPAS)). HIF-3a isoforms
dimerise with HIF-1b forming HIF-3 and HIF-3NEPAS. In general, HIFs
mediate their effects by binding to Hypoxia Response Elements
(HREs) on DNA, which leads to the regulation of some 200 genes, of
which 70 have been studied in detail (Fig. 2). Of note, HIF-1 plays
a key role in maintaining homeostasis, inducing transcription of
key genes such as vascular endothelial growth factor (VEGF) and
erythropoietin (EPO), whereas HIF-2 and -3 have more specialised
and tissue specific regulatory roles (Fig. 2) [21e24].

3. Regulation of HIF through oxygen-dependent mechanisms

Under normoxic conditions (oxygen concentrations above 5%)


HIF-1a undergoes rapid degradation (<5 min) [25]. Hydroxylation
of conserved proline residues (HIF-1a Pro402 and Pro564) occurs via
prolyl hydroxylase domain proteins (PHD1-3), which are located
within a unique oxygen-dependent protein degradation domain.
This leads to proteosomal degradation by ubiquitinase, a prerequi-
site for von Hippel-Lindau (Pvhl) E3 ubiquitin ligase complex (also
containing elongin B, elongin C, Cul 2 and Rbx1), which interact
only with hydroxylated proteins [26e28].
In addition, there are two other oxygen-dependent inhibitors of
HIF-1a known as Factor inhibiting HIF-1 (FIH-1) and p300/CBP
interacting transactivator with ED-rich tail 2 (CITED2) [22]. FIH-1 is
an Fe(II)-dependent asparaginyl hydroxylase acting on the aspar-
agines residue in the TAD region, inhibiting the interaction of HIF- Fig. 3. During normoxic conditions three mechanisms can lead to the proteosomal
1a and HIF-2a with transcriptional coactivators p300/CBP [29,30]. degradation of HIF-1a. 1) Prolyl hydroxylase 1 (PHD1) prepares HIF-1a for degradation
via ubiquitinase by hydroxylating two conserved proline residues. 2) Factor inhibiting
Conversely, CITED2 is a negative regulator of HIF-1 that actively HIF-1 (FIH-1) and 3) p300/CBP interacting transactivator with ED-rich tail 2 (CITED2),
competes for p300/CBP binding (Fig. 3). In an hypoxic environment both prevent or compete for the binding of HIF-1a to p300/CBP, priming HIF-1a for
CITED2 is activated through hypoxia response elements (HRE) [31]. proteosomal degradation [25e27].
More importantly, CITED2 null mice have impaired trophoblast

invasion and inadequate placental blood supply suggesting that


CITED2 plays a vital role in placental development and differenti-
ation by controlling HIF-1[32].

4. Regulation of HIF through oxygen-independent


mechanisms

There is a growing body of evidence suggesting that HIF-1 is


regulated through oxygen-independent factors that include
hormones, cytokines and growth factors. For the purpose of this
review we have focused on the hormone Angiotensin II (Ang II), the
cytokines interleukin-1b (IL-1b), tumour necrosis factor a (TNF-a)
and NF-kb and the growth factors, transforming growth factor-b1
(TGF-b1) and insulin-like growth factor (IGF) (Table 1). Interest-
ingly, many of these are up-regulated during pregnancy, resulting
in increased levels in the maternal circulation, which affect
placental development and maturation throughout gestation
[19,33e36].

4.1. Angiotensin II (Ang II)

Fig. 2. During hypoxia, HIF-1a translocates into the nucleus dimerising with HIF-1b
The role that Ang II plays in early trophoblast invasion and
(Arnt) to form a complex. This protein complex will bind to associated Hypoxia differentiation is well documented. There are increases in angio-
Response Elements (HREs), leading to the transcription of HIF-1 target genes [18e21]. tensin (AT1) receptor concentration and mRNA expression in the
954 J. Patel et al. / Placenta 31 (2010) 951e957

Table 1
Stimuli known to affect placental function: RenineAngiotensin System (RAS), immunogenic cytokines and growth factorsa.

Regulator of HIF-1a Effect on Placental Function References


Angiotensin (II) Increased EVT outgrowth, increased cellular proliferation and increased expression of sFlt-1. [40,42,43]
Interleukin-1b (IL-1b) Up-regulation of uPA, MMP-2 and MMP-9 [52e54]
Uterine specific natural killer cells (uNK) Up-regulation of MMP-9, decreased apoptosis [57]
Transforming Growth Factor-b1 (TGF-b1) Inhibition of trophoblast invasion, increased secretion of PAI-1 and PAI-2, reduction in uPA [65e67]
activity and MMP-2 and MMP-9 secretion
Insulin-Like Growth Factor-II (IGF-II) Increased trophoblast outgrowth [76,77]
a
HIF-1a, hypoxia inducible factor-1a; EVT, extravillous trophoblast cells; sFlt-1, soluble fms-like receptor-1; uPA, urokinase plasminogen activator; MMP, matrix metal-
loproteases; PAI, plasminogen activator inhibitor.

first trimester placenta [33]. The maternal renineangiotensin invasion can be dysregulated leading to deleterious outcomes such as
system (RAS) undergoes major changes following conception. A 1) failure to maintain implantation resulting in miscarriage and 2)
spike in plasma RAS concentrations early in pregnancy is due to inadequate spiral artery remodelling leading to preeclampsia later in
increased release of renin from the kidneys, ovaries and maternal pregnancy [47].
decidua [37]. mRNA expression of the precursor angiotensinogen Many of the uterine changes mediated through immunological
from the liver is increased in response to rising circulating estrogen processes described above occur in an hypoxic environment,
during pregnancy, providing further evidence of an intricate rela- leading to the accumulation and cellular downstream effects of
tionship shared by the early placenta and the RAS [38]. Craven et al. HIF-1a. The notion that cytokines may cause immunomodulatory
(1998) also suggested that RAS may play a role in assisting uteri- changes during normoxic conditions is intriguing. Matrix metal-
ne spiral artery dilatation and VSMC and endothelial cell dis- loproteases (MMP-2 and MMP-9) and urokinase plasminogen
organisation, prior to EVT invasion [39]. Current evidence suggests activator (uPA), have both been shown to be major targets of HIF
that AT1 receptor activation via Ang II leads to cascade signalling gene regulation in placental development [19]. Firstly, uPA plays
affecting a number of cellular systems associated with trophoblast a central role in placental migration and invasion into the maternal
invasion and angiogenesis, including plasminogen activator inhib- decidua [48]. It binds to its associated receptor (uPAR) on the
itor-1 (PAI-1) and soluble fms-like receptor-1 (sFlt-1) that are maternal decidual surface causing a downstream activation of
essential in the first trimester of pregnancy [33,39]. MMPs [49]. MMPs are essential in the degradation of extracellular
A recent study of first trimester placental explants by Araki- matrix (ECM), required for trophoblast invasion and remodelling of
Taguchi et al. (2008) revealed that Ang II mimicked the effects of the maternal decidua [50,51]. Studies in the human trophoblast cell
hypoxia by up-regulating the expression of HIF-1a mRNA expression line JEG-3 and in primary cultures of trophoblast cells from term
and protein [40]. This study also demonstrated EVT outgrowth and placentas have demonstrated that addition of IL-1b under normal
an increase in the expression of nuclear protein Ki67, which is cell culture conditions (21% O2) leads to up-regulation of uPA and
associated with cellular proliferation, providing further evidence for MMP-2 and MMP-9 [52e54]. Qian et al. (2004) have demonstrated
the roles of Ang II and HIF-1a in placental maturation during nor- HIF-1a activation by IL-1b and TNF-a results in increased secretion
moxic (>5% O2) conditions [40]. Conversely, Ang II has also been of VEGF, a potent activator of angiogenesis during times of hypoxia
implicated as a leading cause of IUGR and preeclampsia [33]. In 1999, [55]. The process of HIF-1a activation was driven by both IL-1b and
Wallukat et al. discovered that preeclamptic women produced auto- TNF-a, up-regulating expression of the extracellular signal-regu-
antibodies known as AT1 receptor agonistic autoantibody (AT1-AA), lated kinase (ERK) phosphorylation pathway [56].
which binds to a seven amino acid sequence on the second extra- Lash et al. (2010) demonstrated that decidual uNK cell superna-
cellular loop of the AT1 receptor [33,41]. Overstimulation of the AT1 tants from 12 to 14 weeks gestation stimulated EVT invasion from
receptor dramatically increases the expression and production of placental villous explants [57]. This was associated with increased
sFlt-1, which is a potent antagonist of angiogenesis by binding and MMP-9 levels and decreased apoptosis [57]. This provides further
inhibiting VEGF and placental growth factor (PIGF), reportedly support to the notion that cytokines released by uNK cells may act
seen only very early in normotensive pregnancies [42,43]. There via HIF-1a to activate HIF-1a targets such as MMP-9. This hypothesis
is approximately a two to five times greater expression of does, however, require further research, as suggested by Lash et al.
sFlt-1 in preeclamptic placentas than in placentas of normal preg- Other cell lines such as human hepatoma cells (HepG2) up-regulate
nancies [44,45]. HIF-1a protein after IL-1b and HIF-1 binding to nuclear target sites
following the addition of TNF-a [56]. Further studies have high-
4.2. Cytokines (IL-1b, TNF-a) lighted effects of TNF-a in stabilising HIF-1a in tubular LLC-PK(1)
cells [58]. This provides clear evidence of the key role cytokines can
Evidence is emerging of the interaction between cytokines (inter- play in mediating the effects of HIF-1 at normoxia in a number of
leukins (IL), tumour necrosis factors (TNF), natural killer cells (NK)) and tissue types, including placenta.
the maternal decidua, particularly just after conception and in early NF-kB is the key link that drives cytokine cellular signalling.
placental development [19,34]. This suggests that the role of cytokines There is a close relationship between HIF-1a and NF-kb during
is to ensure that the endometrium is receptive to the implanting hypoxia, which leads to the up-regulation of inflammatory modu-
blastocyst [46]. Endometrial receptivity is characterised by differenti- lators such as cycloxygenase-2 (COX-2), TNF-a and IL-6 [59].
ation and maturation of the endometrial glands into a secretory phase, However, studies have now demonstrated the ability of NF-kb to
as well as increasing the level of production of luminal epithelium cell up-regulate expression of HIF-1a under normoxic conditions. Jung
surface adhesion and extracellular matrix molecules [35]. Furthermore, et al. (2003) observed IL-1b activation of NF-kb via the PI3K/AKT/
there is differentiation of stromal fibroblasts into decidual cells and mTOR cellular signalling pathway resulted in the increase of COX-2,
influx and proliferation of uterine specific natural killer cells (uNK). which drove the increase of HIF-1a protein expression within
Successful implantation requires adequate decidualisation to assist in adenocarcinomic human alveolar (A549), Jurkat, MCF-7 and NIH-
the regulation of early trophoblast invasion and development [34]. 3T3 fibroblast cell lines [60,61]. Furthermore, using placental
However, in times of exaggerated immunological response, trophoblast explants for hypoxia/reperfusion experiments, showed increases in
J. Patel et al. / Placenta 31 (2010) 951e957 955

protein expression of HIF-1a, sFlt-1 and VEGF, which were reduced pregnancy leading to increased bioavailability, with both IGF-I and II
using sulfasalazine, an NF-kb antagonist [62]. Similar findings were having roles in trophoblast invasion, proliferation and survival (by
also obtained using labored placental samples. Increased level of reducing trophoblast apoptosis) [73]. IGF-II protein expression is
NF-kb due to the inflammatory response of labor lead to the also elevated in preeclamptic placentas, although its direct role in
production of increased levels of HIF-1a and sFlt-1. preeclamptic pathogenesis is unknown [36]. It is critical to note
however the ability of IGF-I and II to activate HIF-1a protein
4.3. Growth factors production under normoxic conditions. Although it has been argued
that the level of protein activation is very low, in some studies this
The process of implantation and early placental development has resulted in altered cellular function [74e77]. IGF-I has been
occurs via the controlled regulation of growth factors such as shown to up-regulate HIF-1a protein expression in a number of cell
transforming growth factors (TGF-b), insulin-like growth factors lines including HepG2 and L8 rat skeletal muscle myoblasts [75].
(IGF-I and IGF-II), and epidermal growth factors (EGF). IGF-I has also been shown to lead to the up-regulation and secretion
of VEGF from human osteoblast-like cells through activation of HIF-
4.3.1. Transforming growth factor-1 (TGF-b1) 2a [74,75]. Furthermore, a study by Pringle et al. demonstrated an
The effects of TGF-b on placental development have been increase of both HIF-1a and HIF-2a expression and trophoblast
studied extensively [3,63,64]. TGF-b has three isoforms, b1, b2 and outgrowth, following the addition of IGF-II to early murine tropho-
b3, all of which are produced and secreted by the maternal decidua blasts under normoxic conditions [76,77]. This supports the view
[50,64]. TGF-b1 and b2 have been found in the placenta throughout that additional research in this area is required to elucidate the role
gestation, whereas TGF-b3 exhibits time specific regulation, with of IGFs in placental maturation, especially through their capacity to
mRNA levels highest until the 10th week of gestation, with activate HIFs during normoxic conditions.
a marked drop in mRNA levels thereafter [63]. TGF-b3 has been
implicated as an inhibitor of trophoblast differentiation, thus 5. Conclusion
playing a key role in the regulation of the level of early placental
invasion [50]. It is important to note that TGF-b3 is over expressed The role of hypoxia in regulating trophoblast invasion and
in preeclamptic tissue, providing further evidence of its role in maturation throughout pregnancy is well recognised. Our under-
inhibiting placental development [63]. TGF-b1 has been studied to standing of the molecular mechanisms, including HIF-1a, HIF-1b,
a lesser extent, with conflicting data from numerous studies. Some HIF-2a, that regulate these effects is rapidly expanding. Further
studies have observed the inhibitory effects by TGF-b1 on tropho- development in this area requires an understanding of how these
blast invasion via the RhoA/Rho-associated kinase (ROCK) pathway molecular regulators affect the growth and maturation of the
that causes increases in the secretion of PAI-1 and PAI-2, whereas placenta under normoxic conditions. In this review we have dis-
others have observed a reduction in uPA activity and MMP-2 and cussed three key components (RAS, immunogenic cytokines and
MMP-9 secretion [65e67]. Conversely, TGF-b1 treatment of growth factors) that assist in placental development and which
primary isolated first trimester trophoblasts, did not alter the have also been demonstrated to be key modulators of HIF-1. There
extent of trophoblast invasion [66]. It is also important to note the is now growing evidence that the RAS, cytokines and growth factors
capacity of TGF-b1 to stabilise HIF-1a protein under normoxic can change cellular functions by up-regulating the HIF pathway
conditions. A study by Görlach et al. (2001) demonstrated TGF-b1 under normoxic conditions. Relatively few studies have been per-
up-regulation of HIF-1a protein within human vascular smooth formed on isolated placental cells or tissue, which warrants further
muscle cells (VSMC) as well as the increased translocation of HIF-1 investigation. Activation of HIFs during late pregnancy may play
into the nucleus [68]. A further study by McMahon et al. (2006) a role in the pathogenesis of IUGR and preeclampsia and conse-
using the hepatoma cell line HepG2 and the fibrosarcoma cell line quent adverse pregnancy outcomes.
HT1080, demonstrated that TGF-b1 inhibited prolyl hydroxylase 2
(PHD2), a potent oxygen sensor, leading to degradation of HIF-1a
protein during normoxia [26,69]. The inhibition of PHD2 results in References
preventing Vhl from binding to the HIF-1a protein, resulting in
[1] Huppertz B. The anatomy of the normal placenta. J Clin Pathol 2008;
protein stability [69]. This increases tumour growth by activation of 61(12):1296e302.
VEGF and other similar growth factors. The role of TGF-b1 in the [2] Jackson MR, Mayhew TM, Boyd PA. Quantitative description of the elaboration
complex processes involved in placental invasion and maturation and maturation of villi from 10 weeks of gestation to term. Placenta 1992;
13(4):357e70.
deserves further investigation, particularly in the second and third [3] Genbacev O, Zhou Y, Ludlow JW, Fisher SJ. Regulation of human placental
trimesters of pregnancy. TGF-b1 may well activate genes associated development by oxygen tension. Science 1997;277(5332):1669e72.
with the inhibition of trophoblast invasion and maturation leading [4] Jauniaux E, Gulbis B, Burton GJ. The human first trimester gestational sac
limits rather than facilitates oxygen transfer to the fetusea review. Placenta
to pathologies such as IUGR and preeclampsia. 2003;24(Suppl. A):S86e93.
[5] Huppertz B, Peeters LL. Vascular biology in implantation and placentation.
4.3.2. Insulin-like growth factor (IGF) Angiogenesis 2005;8(2):157e67.
[6] Osol G, Mandala M. Maternal uterine vascular remodeling during pregnancy.
IGF is found in two isoforms in the placenta (IGF-I and IGF-II) and Physiology (Bethesda) 2009;24:58e71.
has been observed in almost all cell types of the placenta including [7] Burton GJ, Jauniaux E, Watson AL. Maternal arterial connections to the
the maternal interface, as early as six weeks of pregnancy [70]. IGFs placental intervillous space during the first trimester of human pregnancy:
the Boyd collection revisited. Am J Obstet Gynecol 1999;181(3):718e24.
are also known to be key HIF-1 target genes. The placenta produces
[8] Rodesch F, Simon P, Donner C, Jauniaux E. Oxygen measurements in endo-
a range of IGF binding proteins (IGFBP 1e6), which bind the IGFs metrial and trophoblastic tissues during early pregnancy. Obstet Gynecol
more strongly than their associated cell membrane receptors [71]. 1992;80(2):283e5.
[9] Jauniaux E, Watson AL, Hempstock J, Bao YP, Skepper JN, Burton GJ. Onset of
During pregnancy, post-translational modifications (phosphoryla-
maternal arterial blood flow and placental oxidative stress. A possible factor in
tion and proteolysis) of the binding proteins are altered, reducing human early pregnancy failure. Am J Pathol 2000;157(6):2111e22.
binding affinity and hence increased bioavailability of IGF [36,71]. [10] Carter AM. Evolution of factors affecting placental oxygen transfer. Placenta
Interestingly, IGFBP-1, 2 and 3 are also HIF-1 regulated proteins, 2009;30(Suppl. A):S19e25.
[11] Harris LK, Keogh RJ, Wareing M, Baker PN, Cartwright JE, Aplin JD, et al.
emphasising their importance in early placental development Invasive trophoblasts stimulate vascular smooth muscle cell apoptosis by a fas
[19,72]. Maternal circulation of IGF-II also increases during early ligand-dependent mechanism. Am J Pathol 2006;169(5):1863e74.
956 J. Patel et al. / Placenta 31 (2010) 951e957

[12] Zygmunt M, Herr F, Munstedt K, Lang U, Liang OD. Angiogenesis and vascu- [42] Karumanchi SA, Epstein FH. Placental ischemia and soluble fms-like tyrosine
logenesis in pregnancy. Eur J Obstet Gynecol Reprod Biol 2003;110(Suppl. 1): kinase 1: cause or consequence of preeclampsia? Kidney Int 2007;
S10e8. 71(10):959e61.
[13] Ashton SV, Whitley GS, Dash PR, Wareing M, Crocker IP, Baker PN, et al. [43] Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth factor
Uterine spiral artery remodeling involves endothelial apoptosis induced by activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci U S
extravillous trophoblasts through Fas/FasL interactions. Arterioscler Thromb A 1993;90(22):10705e9.
Vasc Biol 2005;25(1):102e8. [44] Tsatsaris V, Goffin F, Munaut C, Brichant JF, Pignon MR, Noel A, et al. Over-
[14] Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol 2002; expression of the soluble vascular endothelial growth factor receptor in
2(9):656e63. preeclamptic patients: pathophysiological consequences. J Clin Endocrinol
[15] Burton GJ. Oxygen, the Janus gas; its effects on human placental development Metab 2003;88(11):5555e63.
and function. J Anat 2009;215(1):27e35. [45] Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, et al. Excess placental
[16] Pijnenborg R, Anthony J, Davey DA, Rees A, Tiltman A, Vercruysse L, et al. soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial
Placental bed spiral arteries in the hypertensive disorders of pregnancy. Br J dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest
Obstet Gynaecol 1991;98(7):648e55. 2003;111(5):649e58.
[17] Pijnenborg R, Luyten C, Vercruysse L, Van Assche FA. Attachment and differ- [46] Psychoyos A. Hormonal control of ovoimplantation. Vitam Horm 1973;31:201e56.
entiation in vitro of trophoblast from normal and preeclamptic human [47] Redman CW, Sargent IL. Placental stress and pre-eclampsia: a revised view.
placentas. Am J Obstet Gynecol 1996;175(1):30e6. Placenta 2009;30(Suppl. A):S38e42.
[18] Brosens IA, Robertson WB, Dixon HG. The role of the spiral arteries in the [48] Naruse K, Lash GE, Bulmer JN, Innes BA, Otun HA, Searle RF, et al. The
pathogenesis of pre-eclampsia. J Pathol 1970;101(4):Pvi. urokinase plasminogen activator (uPA) system in uterine natural killer cells in
[19] Pringle KG, Kind KL, Sferruzzi-Perri AN, Thompson JG, Roberts CT. Beyond the placental bed during early pregnancy. Placenta 2009;30(5):398e404.
oxygen: complex regulation and activity of hypoxia inducible factors in [49] Kjoller L, Kanse SM, Kirkegaard T, Rodenburg KW, Ronne E, Goodman SL, et al.
pregnancy. Hum Reprod Update; 2009. Plasminogen activator inhibitor-1 represses integrin- and vitronectin-medi-
[20] Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW. Hypoxia-inducible factor (HIF-1) ated cell migration independently of its function as an inhibitor of plasmin-
alpha: its protein stability and biological functions. Exp Mol Med 2004; ogen activation. Exp Cell Res 1997;232(2):420e9.
36(1):1e12. [50] Caniggia I, Winter J, Lye SJ, Post M. Oxygen and placental development during
[21] Chen L, Endler A, Shibasaki F. Hypoxia and angiogenesis: regulation of the first trimester: implications for the pathophysiology of pre-eclampsia.
hypoxia-inducible factors via novel binding factors. Exp Mol Med 2009; Placenta 2000;21(Suppl. A):S25e30.
41(12):849e57. [51] Caniggia I, Mostachfi H, Winter J, Gassmann M, Lye SJ, Kuliszewski M, et al.
[22] Dunwoodie SL. The role of hypoxia in development of the mammalian Hypoxia-inducible factor-1 mediates the biological effects of oxygen on
embryo. Dev Cell 2009;17(6):755e73. human trophoblast differentiation through TGFbeta(3). J Clin Invest 2000;105
[23] Ietta F, Wu Y, Winter J, Xu J, Wang J, Post M, et al. Dynamic HIF1A regulation (5):577e87.
during human placental development. Biol Reprod 2006;75(1):112e21. [52] Karmakar S, Das C. Regulation of trophoblast invasion by IL-1beta and TGF-
[24] Carroll VA, Ashcroft M. Targeting the molecular basis for tumour hypoxia. beta1. Am J Reprod Immunol 2002;48(4):210e9.
Expert Rev Mol Med 2005;7(6):1e16. [53] Bauer S, Pollheimer J, Hartmann J, Husslein P, Aplin JD, Knofler M. Tumor
[25] Jewell UR, Kvietikova I, Scheid A, Bauer C, Wenger RH, Gassmann M. Induction necrosis factor-alpha inhibits trophoblast migration through elevation of
of HIF-1alpha in response to hypoxia is instantaneous. FASEB J 2001; plasminogen activator inhibitor-1 in first-trimester villous explant cultures.
15(7):1312e4. J Clin Endocrinol Metab 2004;89(2):812e22.
[26] Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, et al. Targeting [54] Huber AV, Saleh L, Bauer S, Husslein P, Knofler M. TNFalpha-mediated
of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regu- induction of PAI-1 restricts invasion of HTR-8/SVneo trophoblast cells.
lated prolyl hydroxylation. Science 2001;292(5516):468e72. Placenta 2006;27(2e3):127e36.
[27] Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, et al. HIFalpha targeted [55] Qian D, Lin HY, Wang HM, Zhang X, Liu DL, Li QL, et al. Normoxic induction of
for VHL-mediated destruction by proline hydroxylation: implications for O2 the hypoxic-inducible factor-1 alpha by interleukin-1 beta involves the
sensing. Science 2001;292(5516):464e8. extracellular signal-regulated kinase 1/2 pathway in normal human cyto-
[28] Kaelin Jr WG. Molecular basis of the VHL hereditary cancer syndrome. Nat Rev trophoblast cells. Biol Reprod 2004;70(6):1822e7.
Cancer 2002;2(9):673e82. [56] Hellwig-Burgel T, Rutkowski K, Metzen E, Fandrey J, Jelkmann W. Interleukin-
[29] Koivunen P, Hirsila M, Gunzler V, Kivirikko KI, Myllyharju J. Catalytic prop- 1beta and tumor necrosis factor-alpha stimulate DNA binding of hypoxia-
erties of the asparaginyl hydroxylase (FIH) in the oxygen sensing pathway are inducible factor-1. Blood 1999;94(5):1561e7.
distinct from those of its prolyl 4-hydroxylases. J Biol Chem 2004; [57] Lash GE, Otun HA, Innes BA, Percival K, Searle RF, Robson SC, et al. Regulation
279(11):9899e904. of extravillous trophoblast invasion by uterine natural killer cells is dependent
[30] McNeill LA, Hewitson KS, Claridge TD, Seibel JF, Horsfall LE, Schofield CJ. on gestational age. Hum Reprod 2010;25(5):1137e45.
Hypoxia-inducible factor asparaginyl hydroxylase (FIH-1) catalyses hydrox- [58] Zhou J, Fandrey J, Schumann J, Tiegs G, Brune B. NO and TNF-alpha released
ylation at the beta-carbon of asparagine-803. Biochem J 2002;367 from activated macrophages stabilize HIF-1alpha in resting tubular LLC-PK1
(Pt 3):571e5. cells. Am J Physiol Cell Physiol 2003;284(2):C439e46.
[31] Freedman SJ, Sun ZY, Kung AL, France DS, Wagner G, Eck MJ. Structural basis [59] Taylor CT, Cummins EP. The role of NF-kappaB in hypoxia-induced gene
for negative regulation of hypoxia-inducible factor-1alpha by CITED2. Nat expression. Ann N Y Acad Sci 2009;1177:178e84.
Struct Biol 2003;10(7):504e12. [60] Jung YJ, Isaacs JS, Lee S, Trepel J, Neckers L. IL-1beta-mediated up-regulation of
[32] Withington SL, Scott AN, Saunders DN, Lopes Floro K, Preis JI, Michalicek J, HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link
et al. Loss of Cited2 affects trophoblast formation and vascularization of the between inflammation and oncogenesis. FASEB J 2003;17(14):2115e7.
mouse placenta. Dev Biol 2006;294(1):67e82. [61] Jung YJ, Isaacs JS, Lee S, Trepel J, Neckers L. Microtubule disruption utilizes an
[33] Irani RA, Xia Y. The functional role of the renineangiotensin system in NFkappa B-dependent pathway to stabilize HIF-1alpha protein. J Biol Chem
pregnancy and preeclampsia. Placenta 2008;29(9):763e71. 2003;278(9):7445e52.
[34] Dimitriadis E, Menkhorst E, Salamonsen LA and Paiva P. Review: LIF and IL11 [62] Cindrova-Davies T. Gabor Than Award Lecture 2008: pre-eclampsia e from
in trophoblasteendometrial interactions during the establishment of preg- placental oxidative stress to maternal endothelial dysfunction. Placenta
nancy. Placenta, 31:S99eS104. 2009;30(Supp. A):S55e65.
[35] Dimitriadis E, Nie G, Hannan NJ, Paiva P and Salamonsen LA. Local regulation [63] Caniggia I, Grisaru-Gravnosky S, Kuliszewsky M, Post M, Lye SJ. Inhibition of
of implantation at the human fetal-maternal interface. Int J Dev Biol 54 TGF-beta 3 restores the invasive capability of extravillous trophoblasts in
(2e3):313e322. preeclamptic pregnancies. J Clin Invest 1999;103(12):1641e50.
[36] Forbes K, Westwood M. The IGF axis and placental function. A mini review. [64] Lysiak JJ, Hunt J, Pringle GA, Lala PK. Localization of transforming growth
Horm Res 2008;69(3):129e37. factor beta and its natural inhibitor decorin in the human placenta and
[37] Hsueh WA, Luetscher JA, Carlson EJ, Grislis G, Fraze E, McHargue A. Changes in decidua throughout gestation. Placenta 1995;16(3):221e31.
active and inactive renin throughout pregnancy. J Clin Endocrinol Metab [65] Graham CH, Lala PK. Mechanism of control of trophoblast invasion in situ. J
1982;54(5):1010e6. Cell Physiol 1991;148(2):228e34.
[38] Brown MA, Gallery ED, Ross MR, Esber RP. Sodium excretion in normal and [66] Bass KE, Morrish D, Roth I, Bhardwaj D, Taylor R, Zhou Y, et al. Human
hypertensive pregnancy: a prospective study. Am J Obstet Gynecol 1988;159 cytotrophoblast invasion is up-regulated by epidermal growth factor:
(2):297e307. evidence that paracrine factors modify this process. Dev Biol 1994;
[39] Morgan T, Craven C, Ward K. Human spiral artery renineangiotensin system. 164(2):550e61.
Hypertension 1998;32(4):683e7. [67] Fafet P, Rebouissou C, Maudelonde T, Vignais ML. Opposite effects of trans-
[40] Araki-Taguchi M, Nomura S, Ino K, Sumigama S, Yamamoto E, Kotani-Ito T, forming growth factor-beta activation and rho-associated kinase inhibition on
et al. Angiotensin II mimics the hypoxic effect on regulating trophoblast human trophoblast migration in a reconstituted placental-endometrial
proliferation and differentiation in human placental explant cultures. Life Sci coculture system. Endocrinology 2008;149(9):4475e85.
2008;82(1e2):59e67. [68] Gorlach A, Diebold I, Schini-Kerth VB, Berchner-Pfannschmidt U, Roth U,
[41] Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jupner A, et al. Brandes RP, et al. Thrombin activates the hypoxia-inducible factor-1 signaling
Patients with preeclampsia develop agonistic autoantibodies against the pathway in vascular smooth muscle cells: role of the p22(phox)-containing
angiotensin AT1 receptor. J Clin Invest 1999;103(7):945e52. NADPH oxidase. Circ Res 2001;89(1):47e54.
J. Patel et al. / Placenta 31 (2010) 951e957 957

[69] McMahon S, Charbonneau M, Grandmont S, Richard DE, Dubois CM. Trans- [73] Forbes K, Westwood M, Baker PN, Aplin JD. Insulin-like growth factor I and II
forming growth factor beta1 induces hypoxia-inducible factor-1 stabilization regulate the life cycle of trophoblast in the developing human placenta. Am J
through selective inhibition of PHD2 expression. J Biol Chem 2006;281 Physiol Cell Physiol 2008;294(6):C1313e22.
(34):24171e81. [74] Feldser D, Agani F, Iyer NV, Pak B, Ferreira G, Semenza GL. Reciprocal positive
[70] Han VK, Bassett N, Walton J, Challis JR. The expression of insulin-like growth regulation of hypoxia-inducible factor 1alpha and insulin-like growth factor 2.
factor (IGF) and IGF-binding protein (IGFBP) genes in the human placenta and Cancer Res 1999;59(16):3915e8.
membranes: evidence for IGFeIGFBP interactions at the feto-maternal inter- [75] Zelzer E, Levy Y, Kahana C, Shilo BZ, Rubinstein M, Cohen B. Insulin induces
face. J Clin Endocrinol Metab 1996;81(7):2680e93. transcription of target genes through the hypoxia-inducible factor HIF-
[71] Westwood M, Gibson JM, Davies AJ, Young RJ, White A. The phosphorylation 1alpha/ARNT. EMBO J 1998;17(17):5085e94.
pattern of insulin-like growth factor-binding protein-1 in normal plasma is [76] Pringle KG, Kind KL, Thompson JG, Roberts CT. Complex interactions between
different from that in amniotic fluid and changes during pregnancy. J Clin hypoxia inducible factors, insulin-like growth factor-II and oxygen in early
Endocrinol Metab 1994;79(6):1735e41. murine trophoblasts. Placenta 2007;28(11e12):1147e57.
[72] Forbes K, Souquet B, Garside R, Aplin JD and Westwood M. Transforming [77] Kwon YW, Kwon KS, Moon HE, Park JA, Choi KS, Kim YS, et al. Insulin-like
growth factor-{beta} (TGF{beta}) receptors I/II differentially regulate TGF growth factor-II regulates the expression of vascular endothelial growth
{beta}1 and IGF-binding protein-3 mitogenic effects in the human placenta. factor by the human keratinocyte cell line HaCaT. J Invest Dermatol 2004;
Endocrinology 151(4):1723e1731. 123(1):152e8.

You might also like