You are on page 1of 15

J Physiol Biochem (2013) 69:559–573

DOI 10.1007/s13105-012-0227-2

MINI REVIEW

The physiological roles of placental corticotropin releasing


hormone in pregnancy and childbirth
Murray Thomson

Received: 11 September 2012 / Accepted: 10 December 2012 / Published online: 29 December 2012
# University of Navarra 2012

Abstract In response to stress, the hypothalamus health issues into their adult life as a result of foetal
releases cortiticotropin releasing hormone (CRH) that programming. Future treatment of these problems with
travels to the anterior pituitary, where it stimulates the CRH antagonists is an exciting possibility.
release of adrenocorticotropic hormone (ACTH).
ACTH travels to the adrenal cortex, where it stimulates Keywords Corticotropin releasing hormone (CRH) .
the release of cortisol and other steroids that liberate Adrenocorticotropic hormone (ACTH) . Cortisol .
energy stores to cope with the stress. During pregnancy, Placenta . Pituitary . Growth restriction . Foetus .
the placenta synthesises CRH and releases it into the Foetal programming . Diabetes . Obesity .
bloodstream at increasing levels to reach concentrations Hypertension . Atherosclerosis
1,000 to 10, 000 times of that found in the non-pregnant
individual. Urocortins, which are CRH analogues are
also secreted by the placenta. Desensitisation of the Introduction
maternal pituitary to CRH and resetting after birth may
be a factor in post-partum depression. Recently, CRH When physiological and psychological stress is pro-
has been found to modulate glucose transporter (GLUT) cessed in the cerebral hemispheres, the hypothalamus
proteins in placental tissue, and therefore there may be a is activated and as a consequence it releases corticotro-
link between CRH levels and foetal growth. Evidence pin releasing hormone (CRH) into the hypophyseal
suggests CRH is involved in the timing of birth by portal system that transports blood to the anterior pitu-
modulating signalling systems that control the contrac- itary. CRH activates the corticotropes of the anterior
tile properties of the myometrium. In the placenta, cor- pituitary to synthesize and release adrenocorticotropic
tisol stimulates CRH synthesis via activation of nuclear hormone (ACTH) and β-endorphin into the blood
factor kappa B (NF-κB), a component in a cellular stream [62]. Both ACTH and β-endorphin are derived
messenger system that may also be triggered by stres- from the protein precursor proopiomelanocortin [22]. β-
sors such as hypoxia and infection, indicating that intra- Endorphin interacts with opioid receptors to allow cop-
uterine stress could bring forward childbirth and cause ing with pain and psychological stress. ACTH travels to
low birth weight infants. Such infants could suffer the cortex of the adrenals where it stimulates the syn-
thesis and release of steroid hormones including cortisol
[42, 94]. Cortisol exerts a range of physiological func-
M. Thomson (*)
tions including the stimulation of gluconeogenesis and
School of Biological Sciences, University of Sydney,
Camperdown, NSW 2006, Australia glycogen catabolism, release of fatty acids from adipose
e-mail: murray.thomson@sydney.edu.au tissue and immune response modulation [8, 20, 72]. The
560 M. Thomson

hypothalamic–pituitary–adrenal system is regulated by contains a CRH-R1β variant which, like CRH-R1d,


negative feedback, ACTH and cortisol inhibit the re- lacks a 14 residue sequence in the seventh membrane
lease of CRH from the hypothalamus [112]. spanning region and this receptor has been designated
Research on the physiological roles of placental CRH CRH-R1β/d. CRH-R1β/d transcription is stimulated by
and its analogues conducted over the last 30 years has progesterone and estradiol 17β, two steroid hormones
increased our understanding of the molecular and cellu- that increase in the blood during pregnancy suggesting
lar events modulated by these molecules that are rele- that CRH-R1β/d may have special roles during preg-
vant to the biology and psychology of the pregnant nancy [45]. In humans, there are three known splice
woman and her offspring. During pregnancy the placen- variants of CRH-2; these are differentiated with the
ta also synthesises CRH and this placental CRH may be suffixes α, β and γ. CRH-R2α and CRH-R2β are
modulating important aspects of physiology including synthesised in the placenta, whereas CRH-R2γ is only
the induction of labour, glucose transport to the placen- expressed in the limbic system of the brain [38]. Differ-
tal cells and the foetus and the psychological mood of ent isoform combinations of CRH receptors found in
the mother. Recent evidence suggests that appropriate different tissues suggest that the combination of CRH
CRH modulation of pregnancy and placental function is receptor isoforms is a way that CRH family peptides can
vital to prevent conditions such as premature birth and stimulate different intracellular messenger systems.
low birth weight infants, conditions that can have seri-
ous health consequences for the rest of the infant’s life.
CRH from the placenta
CRH and related peptides
In the 1980s, it was discovered that during pregnancy,
concentrations of CRH in the plasma increased to reach
CRH and its related peptides are thought to have evolved
levels that were 1,000–10,000 times that of non-pregnant
from diuresis modulating proteins, homologues of which
women [13, 89]. The placenta was identified as the
are present in insects today [15]. CRH is a 41-amino-acid
possible source of the extra hormone levels; CRH im-
peptide that was first purified from sheep hypothalamus
munoreactivity was discovered in fixed placental tissue
[5, 108]. It is now known that there are three other related
and placental CRH was found to be identical in structure
proteins that together with CRH form the CRH family of
to hypothalamic CRH and biologically active on anterior
proteins, these are, urocortin (also known as urotensin I)
pituitary cells [74, 82, 84, 89, 97]. Fragments of living
which has 45 % homology to CRH, urocortin II (also
placental tissue continuously superfused with fresh me-
known as stresscopin-related peptide) with 55 % homol-
dium were shown to secrete CRH at levels that would
ogy and urocortin III (also known as stresscopin), which
account for the plasma levels of CRH at term [97]
has 32 % homology [6, 110].
providing strong evidence that the placenta is the major
source of CRH during pregnancy [88]. In contrast to the
massive rises in plasma CRH during pregnancy, ACTH
CRH receptors and cortisol levels in the plasma rise comparatively
modestly and initially this was puzzling as to why the
There are two separate genes that produce the two major much larger rise in CRH did not affect ACTH and
forms of CRH receptor. These are named CRH-R1 and cortisol levels more profoundly. CRH-binding protein
CRH-R2 and both proteins are seven transmembrane G- which is normally present in the plasma affords some
protein coupled receptors [16, 73, 76]. Differential splic- attenuation of CRH bioactivity but does not completely
ing of both the CRH-R1 and CRH-R2 genes produces shield the corticotrope from placental CRH, and desen-
variants that have different biological roles. CRH-R1β, sitization of the maternal anterior pituitary to stimulation
the largest of the R1 variants, has 444 amino acids and by placental CRH is another factor that likely keeps
forms the sequence from which other R1 variants are peripheral ACTH and cortisol from rising to pathological
derived [38]. CRH-R1α has 29 less amino acids in its levels in the pregnant woman [56, 58, 67, 94, 95, 98].
sequence, a difference that produces a higher affinity for The evidence that placental CRH reaches and stimulates
CRH [30]. In addition, the c, d, e, f and h forms of CRH- the anterior pituitary includes the following. Non-
R1 have been found. Furthermore, human myometrium pregnant humans increase release of ACTH after an
Roles of placental CRH in pregnancy and childbirth 561

injection of CRH and because non-pregnant people have pregnancy and an increase in the amount of ion channels
the same levels of circulating CRH binding protein as and gap junctions in the tissue at this time allows the
women in early pregnancy, the CRH-BP clearly does not propagation of action potential driven contraction
stop all CRH in the circulation from reaching the pitui- throughout the tissue [47]. This process is facilitated
tary [21, 90, 114]. CRH-BP levels decrease in the later by a dramatic ‘rewiring’ of receptor and intracellular
stages of pregnancy thus further reducing the ability of messenger systems so that contractile processes are
CRH-BP to attenuate placental CRH activity at this time triggered more frequently [12]. It has been thought for
[55]. Furthermore, infusing rats for 50 h with CRH some time that CRH plays a role in the transition of the
desensitises the pituitary to subsequent CRH stimulation myometrium and the timing of birth. As supporting
[100], and pituitary cells continuously perfused with evidence, sharp increases in plasma CRH occurs sooner
medium desensitize within 60 min to prolonged expo- in women that give birth before 41 weeks and the
sure to CRH [96]. The evidence shows therefore that opposite is true for women who go into labour after this
placental CRH not only reaches the maternal pituitary, it time [88, 101]. In the first 27 weeks of pregnancy,
desensitizes it to subsequent CRH stimulation. women showing signs of going into preterm labour
Animal models are not readily available to study show normal levels of CRH in the circulation for that
rising levels of CRH during pregnancy as CRH appears stage of pregnancy but from weeks 28–36 in women
to only be expressed in the placenta of higher primates with impending premature delivery, higher CRH levels
and not in that of rodents and lemurs [77]. In monkeys for that time in pregnancy indicate that labour will occur
CRH reaches peak levels mid gestation then decline to a within a day [49, 85]. In post-term pregnancies, CRH
plateau level for the rest of the pregnancy until term [75], levels in the plasma are significantly lower than normal
unlike humans and great apes where levels rise exponen- while levels of CRH-BP are elevated [25]. CRH mRNA
tially throughout pregnancy to peak at labour. The mid- is elevated in the blood and placenta of women experi-
term peak of CRH may have been lost somewhere encing preterm preeclampsia as compared to women at
between ancestral anthropoids and the evolution of hom- normal term delivery [68]. There is substantial evidence,
inids [75]. Since the discovery of biologically active therefore, to show that placental CRH is involved in the
placental CRH many scientists have been determining timing of birth; the challenge now is to fully determine
the roles of placental CRH on the physiology of the the mechanisms through which CRH is helping to bring
pregnant woman and the implications for her offspring. on parturition and what roles the urocortins are playing.
The factors that could alter CRH expression, and there-
fore alter the timing of birth, are just beginning to be
The contractile transition in uterine myometrium understood, and these may include epigenetic chromatin
modification [1], stress and infection.
The upper section of the myometrium is in a state of Timed changes in the way the myometrium expresses
non-contraction or quiescence for most of pregnancy different CRH receptor types, and links these to intra-
whereas the lower section is contracted before child- cellular signalling systems, may play a major role in the
birth. At the time for labour, the lower section of myo- development of contractility in the myometrium. In this
metrium becomes relaxed whereas the upper region way, CRH could function to keep the upper myome-
goes through a transition to become highly contractile trium in a state of quiescence for most of the pregnancy
and thereby expel the foetus. The complex interplay and then at the last stage of pregnancy when CRH
between endocrine signals, intracellular messenger sys- receptors are switched to interact with contractile ele-
tems and cellular components that regulates this transi- ments of the cell, high levels of CRH may trigger
tion is poorly understood, but research performed over parturition [30]. When it is time for the myometrium
the last decade has established some key points and to be contractile, calcium stimulates the interaction of
implicated CRH in the process. At the time for labour mysosin and actin filaments in the smooth muscle. This
the upper myometrium becomes sensitive to contractile action is initiated by receptors such as the oxytocin
signals from hormones including oxytocin and prosta- receptor and the endothelin receptor that transmit their
glandins [14, 36, 88]. A decrease in the level of polar- signal through heterotrimeric GTP-binding proteins (G
isation in the myocytes increases the chances of proteins). From here the signal is transmitted to phos-
depolarization and contraction at the latter stages of pholipase C (PLC) that triggers the well-known inositol
562 M. Thomson

phosphate system via the hydrolysis of the membrane hormones such as oxytocin and cortisol. At term, CRH
phospholipid phoshatidylinositol 4,5-bisphosphate by itself may not be able to increase contraction of the
(PIP2), which produces inositol triphosphate (IP3) and upper myometrium but may prime the tissue so that it
diacylglycerol (DAG). DAG activates protein kinase C becomes contractile when stimulated by another hor-
(PKC) that triggers the mitogen-activated protein kinase mone such as oxytocin or steroids and future research
(MAPK) cascade that acts to phosphorylate myosin is needed to test this theory.
light chain and stimulate contraction [30, 44]. Further- Indeed, when the time for labour approaches, there is
more, IP3 binds to the IP3 receptor (IP3R) in the sarco- a rapid proliferation in oxytocin receptors, which as
plasmic reticulum and allows calcium to exit the mentioned leads to a rise in the phosphorylation of
sarcoplasmic reticulum via the IP3R channel domain. myosin light chain and contraction of the smooth muscle
When calcium moves into the cytoplasm, it can attach to cells [37]. This is one stage in what is a complex
calmodulin, thereby activating it and allowing calmod- resetting of interacting intracellular messenger systems
ulin to bind to myosin light chain kinase (MLCK). The that previously held contraction in check and it is pos-
regulatory light chain of mysosin is able to interact with sible that as the time for childbirth draws near, CRH
actin after being phosphorylated by MLCK which receptors may have their coupling to cellular signalling
causes the smooth muscle cell to contract [2]. Waves pathways altered so that they switch to playing a role in
of contraction are paused when the stimulatory phos- stimulating contraction [95]. Urocortin II working
phate on actin is removed by a phosphatase [70]. through to CRH-R2 has been shown to stimulate the
For most of pregnancy, when the myometrium is in phosphorylation of myosin light chain in myometrial
its quiescent phase the contractile mechanisms are strips taken from women undergoing caesarean section
held in abeyance by a suite of molecular and cellular at term before labour. The CRH-R2 appears to be trig-
mechanisms including the export of cellular calcium gering the following cellular processes, CRH-R2 acti-
through the plasma membrane and compartmentaliz- vates a receptor associated heterotrimeric G protein
ing in the sarcoplasmic reticulum. During this time, which activates PKC that phosphorylates MEK1 which
CRH receptors inhibit PLC activation and other intra- is a mitogen-activated protein kinase kinase (MAPKK).
cellular signalling systems to prevent smooth muscle MEK1 then phosphorylates extracellular signal-related
contraction [30]. CRH-R1 molecules are coupled to G kinase 1 (ERK1) which is a MAPK. ERK1 then phos-
proteins, mainly to those which have a Gαs subunit. phorylates the myosin light chain resulting in height-
The Gαs subunit in an active and GTP bound G ened contraction. ERK1 and PKC may also
protein triggers the classic cAMP pathway whereby phosphorylate the small monomeric GTP-binding pro-
Gαs stimulates adenylate cyclise to convert ATP to tein RhoA which translocates to the plasma membrane
cAMP that binds to PKA and activates it by releasing after urocortin II stimulation of CRH-R2 and once it
inhibitory subunits. PKA then phosphorylates PLC at reaches the plasma membrane RhoA can activate Rho
an inhibitory phosphorylation site thus preventing kinase which may also phosphorylate myosin light
PLC from phosphorylating the myosin light chain chain thereby stimulating contraction [44].
and inhibiting myometrial contraction. If CRH recep- Recent studies have provided evidence that interplay
tors were simply decoupled from this system before between CRH-Rs and the large-conductance calcium-
labour it would be expected that CRH would be able activated potassium channel BKCa may also play a role
to attenuate myometrial tissue taken pre term, but in the switch from quiescence to contraction in the
would not be able to decrease contractions of myome- myometrium. BKCa is activated by voltage and calcium,
trial tissue at term, and instead may stimulate contrac- in early stages of pregnancy fluxes of calcium from the
tion of term myometrial tissue; however, this is not the sarcoplasmic reticulum into the cytoplasm activates
case. Exposure of spontaneously contracting myome- BKCa, which allows potassium to escape the cell thereby
trial strips taken both at pre-term and at term, to preventing the calcium from depolarizing the cell and in
increasing doses of CRH administered over 2.5 h has this way quiescence is maintained [47]. In the earlier
been shown to decrease the frequency and strength of stages of pregnancy and before labour, CRH increases
contractions [104]. It must be kept in mind that this in the expression of BKCa via CRH-R1 while dampening
vitro system does not completely recreate the situation BKCa synthesis via CRH-R2. At the time of parturition
in vivo where the myometrium is also exposed to other this relationship reverses and CRH-R1 attenuates BKCa
Roles of placental CRH in pregnancy and childbirth 563

expression while CRH-R2 increases BKCa production Cong and co-workers [19] used Western blot to show
[117]. So, at the time for childbirth, an increase in the that CRH-R1 but not CRH-R2 levels are lowered at
relative levels of CRH-R1 could be responsible for the the time of delivery in the upper section of the myo-
decreasing BKCa levels at the time of labour and this may metrium whereas CRH-R1 and CRH-R2 levels
help increase the chances of depolarization and contrac- remained constant in the lower section [19] (see
tion. It is not known at this point, however, whether Fig. 1). In contrast, Stevens and colleagues [91]
CRH-R1 and/or CRH-R2 switch roles in controlling reported that CRH-R1 mRNA increased in the myo-
BKCa levels by decoupling from their prelabour G pro- metrium at term and labour and this increase was due
teins and reattaching to different G proteins and intracel- to increased levels in the lower section of the uterus.
lular pathways at the time of myometrial transformation. Jin and colleagues [43] did not find any difference in
Furthermore, whether the myometrium increases its CRH-R1 and CRH-R2 mRNA and protein levels in
ratio of CRH-R1/CRH-R2 in order to allow contractions the myometrium of caesarean section biopsies taken
is unknown at present as experiments on the expression from women in labour as compared to those not in
of CRH-R1 and CRH-R2 before and approaching or labour; the distribution of CRH-R1 subtypes were
during labour have not given consistent and unequivocal found to be different, however, with CRH-R1f identi-
results and the question remains as to whether R1 and fied in all of the labour myometria samples while this
R2 subtype ratio shift plays a role in the myometrial molecule was only present in half of the non-labour
transition at the time for birth. The human non-pregnant myometria [43]. If it is possible for researchers in the
myometrium expresses the CRH receptor subtypes 1c future to take small biopsy samples from both the
and 2α but pre-labour the receptors 1α, 1β, 1c and 2α upper and lower portions of the uterus at the time of
are present [31]. The CRH affinity of these receptors caesarean delivery in women in labour and not in
may also alter as the time for parturition approaches and labour, a clearer understanding in the shift in CRH
the coupling efficiency to adenylate cyclase to produce receptor expression will be achieved.
cAMP may also reset [32]. Interactions between the
CRH receptors and prostaglandins may also play a role
in the timing of childbirth, prostaglandin E2 stimulates
Can CRH affect the psychology of the pregnant
cervical ripening and myometrial contractions and in
woman?
cultured chorion trophoblast prostaglandin E2 produc-
tion is accelerated by CRH and urocortin binding to
As discussed, there is evidence to show that the pregnant
CRH-R1 and not via CRH-R2 [26]. Whether the falling
woman’s pituitary is desensitized to CRH, thus causing
levels of CRH-R1 and CRH-R2 in the cervix during
an alteration to the normal functioning of the hypotha-
pregnancy have effects on the synthesis of prostaglandin
lamic–pituitary–adrenal axis during pregnancy. It has
E2 is unknown, and future studies are needed to deter-
mine whether changes in CRH-R1 to CRH-R2 ratios
can affect cervical ripening via the modulation of pros-
taglandin E2 [95]. As mentioned CRH receptors trigger
the cAMP pathway and cAMP regulates the expression
of cyclo-oxygenase-2 (COX-2) in human myometrial
cells and COX-2 is a key enzyme in the production of
prostaglandins [17]. It will be important to determine in
future studies whether CRH and related peptides have
an effect on prostacyclin (PGI2) as PGI2 stimulates the
production of contractile proteins and the gap junction
protein connexion 43 in cultured myometrial cells, and
increases the strength of oxytocins induced contractions
in myometrial strips [24]. Fig. 1 CRH-R1 expression in the upper segment (US) and
lower segment (LS) myometrium in non-pregnant (NP), term
More research is needed in the future to reliably non-labour (TNL) and term labour (TL) women, *P<0.05, **P<
determine how the profiles of CRH receptors change 0.01. Reproduced from [19] under the terms of the creative
in the myometrium during pregnancy and labour. commons attribution license
564 M. Thomson

been known for some time that alterations of the hypo- research is, do higher than normal CRH levels in
thalamic–pituitary axis are associated with various pregnancy bring forward childbirth in depressed preg-
forms of psychological disturbance [4, 18, 69]. Patients nant women? In a group of 70 women, there was a
suffering from depression show a reduction in the significant correlation between severity of post-partum
ACTH response to an injection of CRH, a result indi- blues and the extent of CRH decrease in the 6-day
cating that their pituitary is desensitized to CRH [29, 40, period following childbirth [65]. After birth, the pitu-
98]. The desensitization of the pregnant woman’s pitu- itary may re-sensitise to CRH and the hypothalamus
itary to CRH may also cause a psychological disruption. may stop secreting higher amounts of CRH and other
Around about one third of women experience mood hormones such as vasopressin that were previously
disturbance during pregnancy and/or after childbirth needed to control the desensitized pituitary, and this
and this can range in severity from the common, ‘post- resetting of the hypothalamic–pituitary axis may be a
partum blues’ to more severe forms of depression and causal factor in the postpartum blues. The focus of
psychosis that are comparatively rare and need psychi- most research on CRH-R agonists and their role in the
atric care [9, 11, 71]. In the late 1980s, it was proposed psychology of the pregnant woman has been on CRH,
that alterations in the hypothalamus–pituitary–adrenal but because urocortins also interact with CRH-Rs and
axis caused by placental CRH could contribute to the are also implicated in mood control [81], it is high
mood disturbance encountered by some women before time for research to focus more intently on the possible
and after childbirth [99]. Common endocrine phenome- role of placental urocortins contributing to post-
na observed in both endogenous depression and preg- partum blues and other physiological roles in pregnan-
nancy include increased basal levels of cortisol in the cy. CRH-R blockers are under investigation as possi-
plasma and urine but still showing a diurnal rhythm, a ble therapeutics for psychological disturbances such as
blunted ability of the synthetic gluccocorticoid dexa- excess anxiety [28], whether they could be used to
methasone to reduce cortisol levels and a reduced in- counteract post-partum blues or other conditions
crease in plasma cortisol following an injection of CRH caused by excess placental production of CRH is
[21, 89]. A correlation was found between cortisol con- unknown at present.
centrations in the plasma at week 38 and post-partum
mood disorder [34, 86]. Women with post-partum de-
pression have a significantly blunted ACTH response to Can psychological stress trigger premature birth?
CRH injection, suggesting a pituitary desensitized by
placental CRH and this phenomenon persists up to It has been a long held popular belief that psychological
12 weeks after childbirth [58]. stress in the mother could bring about a premature birth.
Since the 1980s, evidence has accumulated to show The idea has some plausibility, and it has been proposed
a link between state of mind and an altered hypotha- that CRH released from the mother’s hypothalamus in
lamic–pituitary–adrenal axis during and after pregnan- response to stress would trigger ACTH and cortisol
cy. A group of 27 pregnant women suffering from release, and this could accelerate CRH synthesis and
major depression had significantly higher levels of release from the placenta [57]. Whether this kind of
circulating CRH in the second trimester as compared event could increase placental CRH output and plasma
to a control group of 38 euthymic pregnant women; CRH levels to the extent, and for long enough to accel-
cortisol levels measured in the evening were also erate the myometrial transition and bring forward partu-
elevated in the group with major depression [66]. rition, however, is unknown. Furthermore, the
The question remains — did the higher levels of desensitization of the pregnant woman’s pituitary by
CRH cause a hypothalamic–pituitary axis upset that placental CRH may severely blunt the mother’s ability
contributed to the depression or did the stress of the to increase cortisol output in response to stress [89, 94,
depression stimulate the maternal hypothalamus and 96, 99]. The research performed to date does not uni-
pituitary to stimulate glucocorticoid release from the versally support the hypothesis that psychological stress
adrenal which stimulates CRH release from the pla- increases CRH levels in the plasma of the pregnant
centa? The shorter pregnancy of the depressed women woman and increases the chances of pre-term birth.
was not significant but this may have been due to the Himes and Simhan [39] found no association between
small sample size and an important question for future perceived psychological stress and CRH and cortisol
Roles of placental CRH in pregnancy and childbirth 565

levels in the plasma in women at 24 and 28 weeks of The mechanism of cortisol stimulation of placental
pregnancy and no effect of the perceived stress on CRH
preterm delivery. In a cohort of 282 women, there was
no correlation between levels of anxiety and plasma Additional work is needed to fully elucidate the mo-
CRH at 18–20 weeks of gestation; however, a modest lecular and cellular mechanism by which glucocorti-
correlation (p<0.05) was found at 28–30 weeks [59]. coids stimulate CRH synthesis in the placenta. Recent
Kramer and colleagues [51] found no correlation be- work has demonstrated the involvement of nuclear
tween psychological stress levels and circulating CRH transcription factor κB (NF-κB) [111]. NF-κB pro-
in a cohort of 635 women measured between 24 and teins are widely expressed in animal cells and often
26 weeks of gestation. More work is needed therefore to mediate the signals from pro-inflammatory signal mol-
determine whether psychological stress in the last tri- ecules and are also important signal molecules in
mester of pregnancy can hasten childbirth. development. Mammals usually express five NF-κB
In order to understand whether increased produc- proteins, NF-κB1 (also known as p50), NF-κB2
tion of hypothalamic CRH can significantly elevate (p52), RelA (p65), RelB, c-Rel, and these proteins
placental CRH, more research is required to under- can form a variety of homodimers and heterodimers
stand the cellular and molecular effects of cortisol on which can travel to the nucleus and stimulate the
the control of CRH production in the placenta as the in activation of various genes. In the cytosol, NF-κBs
vitro work done so far has not fully modelled the can be bound to an inhibitor, IκB. When IκB is
situation in vivo. It was in the late 1980s when it phosphorylated by IκB kinase kinase (IKK), it is
was discovered that glucocorticoids stimulated CRH marked for cellular destruction, thereby releasing
mRNA production in placental tissue using cytotro- NF-κB which can travel to the nucleus and stimu-
phoblast cultured in Dulbecco’s modified Eagle medi- late gene transcription [109]. IKK is a complex
um (DMEM) for 24 h with or without 1 μm of comprising of IKKα, IKKβ and the regulatory pro-
dexamethasone [78]. The control cell cultures that Rob- tein NF-κB essential modulator (NEMO). In the
inson and colleagues [78] used to show glucocorticoid classic or canonical pathway (see Fig. 3), the se-
stimulation of placental CRH were bathed in a static quence is triggered by Toll-like receptors (TLRs) that
bath of DMEM that contained no cortisol. This is a bind signal molecules such as TNFα and lipopolysac-
different situation to the placenta in vivo which is con- charides leading to the phosphorylation and activation
tinuously exposed to cortisol throughout pregnancy and of IKK, but the system can also be stimulated from
could be synthesizing CRH at the top of its abilities. within the cell by processes such as increased produc-
Whether a small and transient rise in cortisol due to tion of NF-κB proteins or via other kinases that phos-
psychological stress will have any effect on placental phorylate and activate IKK. In the non-canonical
CRH release in vivo is unknown. Pituitary cells that are pathway activated NF-κB inducing kinase (NIK)
superfused with a running stream of medium (as op- phosphorylates IκB kinase alpha (IKKα), which in
posed to a static bath in a culture dish) desensitize within turn phosphorylates the C-terminal portion of the
1 h to CRH stimulation [96] and if placental cells show a NF-κB2 precursor p100 and marks it for degradation
similar desensitization to continuous stimulation with to leave the N-terminal portion NF-κB2 that can
cortisol, additional doses of cortisol over background then dimerize with RelB and travel to the nucleus
may have a very limited ability to further stimulate the to stimulate target genes. Immunoprecipitation of
CRH gene in the placenta in vivo. The time is ripe for placental tissue has revealed RelB and NF-κB2 com-
new studies on the response of placental synthesis and plexes, with the CRH gene indicating that these
release of CRH and urocortins in response to a range of transcription factors can activate CRH transcription
cortisol levels using tissue that has been acclimated or via the non-canonical pathway. Additionally, glucco-
pre-exposed to cortisol levels that model those observed corticoid stimulation of CRH mRNA and protein
at various stages of pregnancy. If the placenta can re- production is inhibited by RNA interference blocking
spond to stress induced rises in cortisol over background of RelB and NF-κB2 [111]. It will be important to
levels, and can produce higher levels of CRH it would determine whether cortisol stimulates NIK synthesis
lend weight to the theory that stress could increase the and whether NIK activates NF-κB in human placen-
chances of premature birth. tal tissue.
566 M. Thomson

Glucose transfer glucose consumption in the foetus and causes hypo-


glycaemia and this condition has been implicated in
Glucose is a major source of energy in cells and is low birth weight [10]. Glucocorticoid receptor stimu-
used as a metabolite to produce numerous biological lation increases the expression of GLUT1 and GLUT3
molecules including fats and proteins. Glucose is in cultured human placental epithelial cells but
transported across plasma membranes by either a pas- decreases expression in cultured human trophoblast
sive facilitative process that allows passage of glucose cells [33, 48]. Further elucidation of the endocrine
to follow a concentration gradient driven transport or controls on GLUT expression in the different cells of
an active transport process that utilizes sodium gra- the placenta and how glucose supply to the placenta
dients to drive the transport of glucose across a plasma and foetus is regulated by CRH-R agonists from the
membrane against its concentration gradient. Active, placenta are likely to form an area of intense interest in
sodium-dependent transport of glucose only occurs in the coming years [87].
the epithelium of the small intestine and the nephron A mismatch of glucose supply to the foetus and
proximal convoluted tubule and this process utilizes placenta in relation to the metabolic needs of the
the sodium glucose transport protein (SGLT) family of tissues is thought to be one of the factors leading to
proteins. In other areas, the passive facilitative trans- intrauterine growth retardation and low birth weight.
port of glucose relies on the sugar transporter (GLUT) Intrauterine growth retardation increases infant mor-
family of proteins [118]. There are 13 proteins in the bidity and mortality and increases the chances of de-
GLUT family named GLUT 1–12 and H+-coupled veloping pathophysiologies including heart disease
myo-inositol transporter (HMIT) [113]. GLUT1 and hypertension and insulin intolerance later in life [7,
GLUT3 are essential for the transport of glucose from 115]. Indeed, real-time PCR studies have shown that
the maternal circulation through the placenta into the the CRH gene is translated in higher levels in postpar-
foetal circulation. In the rabbit placenta, GLUT1 is tum placentae from intrauterine growth restriction
localized at the periphery of outer trophoblasts sug- pregnancies [92, 105], and it will be vital to find out
gesting the role of the transporter in the uptake of whether inappropriately high levels of placental CRH
glucose from maternal blood to placenta unlike can cause inappropriate GLUT expression and disrupt
GLUT3 which is situated at the base of the inner healthy glucose transport in the placenta (Fig. 2). It
trophoblast and in foetal blood vessels indicating its will be interesting to see how future studies determine
role in transport to the foetal circulation [46]. what mechanisms are used by the foetus and placenta
GLUT1 and GLUT3 isoforms are present in villous to try and counteract conditions that cause low birth
syncytiotrophoblast, and their expression is regulated
by CRH although the regulation between CRH-R1 and
CRH-R2 is different. CRH working through CRH-R1
up-regulates GLUT1 but down-regulates GLUT3,
whereas CRH-R2 down-regulates both GLUT1 and
GLUT3 expression [27]. It will be important to find
out whether higher levels of CRH at the later stages of
pregnancy are working mainly through CRH-R1
receptors to increase GLUT1 expression and increase
glucose transport to the growing foetus and placenta.
The rapid growth of the placenta towards term neces-
sitates the increased transport of glucose via GLUTs so
the increased expression of GLUT1 and GLUT3 is
likely to be orchestrated via endocrine control includ- Fig. 2 CRH expression (normalized to hypoxanthine phosphor-
ing participation by CRH and related peptides. Gluco- ibosyl transferase) in placental tissue of intrauterine growth
corticoid receptors are expressed in the placenta and restricted (IUGR) neonates and in gestational age-matched con-
trols (GAMC) (n=22 matched pairs). Values are expressed as
glucocorticoids also play a role in the regulation of mean±SEM. There is a significant difference in CRH gene
glucose concentrations in the placenta and foetus [53]. expression between the two groups, *P < 0.05; **P < 0.01;
Over stimulation of glucocorticoid receptors increases ***P<0.001. Reproduced from [92] with permission
Roles of placental CRH in pregnancy and childbirth 567

weight. For example, in conditions of hypoxia tropho- normal levels signal to the foetus that the maternal
blast respond by increasing the expression of GLUT1, environment is becoming unsafe, and the foetus
perhaps to allow more glucose to reach the foetus to responds by speeding up its development and trigger-
cope with the physiological stress [35]. Hypoxia also ing endocrine controls that will cause the date of
increases expression of urocortins II and III, but parturition to be brought forward in time [83]. It is
whether these trigger CRH-Rs to alter GLUT expres- not known at this time, however, whether stress in
sion and modulate glucose transport in the foetus and pregnant women can elevate cortisol significantly
placenta is unknown at present [41]. above background levels and for long enough to in-
crease expression of the placental CRH gene and
accelerate the triggering of parturition. This will be
Placental CRH and foetal programming important to know as acceleration of development
and/or delivery comes at a cost to premature born
Foetal programming occurs when events such as stress babies, the consequences include low birth weight
and overexposure to hormones has a lasting effect on the and lower levels of brain development accompanied
physiology and psychology of the foetus that persists by stunted neuromuscular coordination and emotional
postpartum and sometimes on into adult life [64]. Ex- resilience. As mentioned, it will be important to un-
posure of the foetus to hypothalamic–pituitary–adrenal derstand how placental CRH-R agonists control
axis hormones especially cortisol may have a bell- GLUT expression in the placenta and whether defi-
shaped curve in relation to the effects on the ongoing ciencies in this system can lead to interuterine growth
health of the infant. For example, exposure to cortisol is restriction with consequent health implications such as
needed for healthy foetal nervous system, heart and lung type 2 diabetes, obesity, hypertension and atheroscle-
development; in higher doses, however, cortisol expo- rosis that can manifest at various times for the rest of
sure in the foetus can lead to an increased susceptibility the newborn’s life [7].
to cardiovascular disease later in life and chronically
elevated levels of CRH can be neurotoxic to developing
brain cells [83]. It has been known for some time that Infection and the inflammation
glucocorticoid treatment during pregnancy elevates the
risk of a growth restricted foetus (perhaps by altering the It is well established that infection of the amniotic cavity
physiology and development of the placenta) and can cause premature labour [80], but the mechanism by
increases the likelihood of problems with the nervous, which this occurs is not yet fully characterised. Micro-
endocrine and cardiovascular systems later in life [23, bial invasion of the amniotic cavity can cause lesions in
60, 116]. Whether unusually elevated endogenous glu- both the maternal and foetal portions of the placenta
cocorticoids can have the same effect is not known. with increases in inflammatory cytokine and prostanoid
Furthermore, high levels of cortisol can cause a lower- production, ruptures in membranes or poor placental
ing of bone-derived neurotrophic factor (BDNF), a mol- perfusion [50, 79, 80]. These phenomena may be linked
ecule involved in the survival and development of to the production of placental CRH-R agonists, placenta
neurones, and this may contribute to the toxic effect of obtained from women who had premature rupture of
cortisol on cultured rat hippocampus cells [63]. In rats, membranes with chorioamnionitis had higher levels of
foetal growth retardation induced by administration of CRH, urocortin2 and CRH-R1 mRNA than women
dexamethasone is accompanied by increases in GLUT1 with preterm labour or premature rupture of membranes
and GLUT3 expression, which may be a mechanism without chorioamnionitis, indicating that infection of
whereby the placenta tries to increase glucose transport the uterus and foetal membranes can trigger the release
to stressed tissues [52]. The potential risks of overactive of these components of the stress response, a conclusion
placental CRH and gluccorticoid production mean that supported by the finding that exposing trophoblast tis-
it is vital to find out whether stressors and infection can sue to lipopolysaccharide to mimic the infective process,
amplify the production of these molecules. resulted in an increase in the in vitro expression CRH,
In humans and several animal species, there is urocortin2 and CRH-R1 mRNA [102].
evidence to suggest that hypothalamic–pituitary–adre- A clear pathway by which intrauterine infection
nal hormones that are elevated substantially over could stimulate placental CRH is apparent (Fig. 3).
568 M. Thomson

TLR LPS

MyD88
IRAK-4
IRAK-1 Uev1A Ubc13
TRAF6 TRAF6
Iκ B TAB1 TAB2
NEMO TAK1
NF κ B
IKKα IKKβ

NF κ B
transcription
NF κ B

CRH CRH gene


promoter

Fig. 3 Possible pathway used by LPS to stimulate CRH expres- consequence, TAK1 is activated and phosphorylates and acti-
sion via MyD88 and the canonical NFκB pathway. LPS inter- vates the IKK complex that is comprised of NEMO, IKKα and
acts with TLR that can use adapter MyD88 to attract IRAK-4, IKKβ. IKKβ then phosphorylates IκB, which marks IκB for
IRAK-1 and TRAF6. TRAF6 is activated as a consequence, ubiquitylation and proteolysis in the proteosomes. The liberated
disassociates and complexes with TAB1, TAB2 and TAK1, NFκB then travels to the nucleus where it binds to the CRH
which then add Uev1A and Ubc13 to the complex. As a promoter and stimulates transcription (original figure by author)

Lipopolysaccharide is a component of the Gram (−) are needed, however, to determine whether MyD88 is
bacterial cell wall and has been found to cause a rise in exerting its effects on the placental CRH gene via this
trophoblast CRH synthesis [106, 107]. Lipopolysac- pathway.
charide induces its action via a TLR that activates the The increased levels of CRH due to infection
myeloid differentiation primary response gene product may help switch the uterus from quiescent to con-
(MyD88) that causes a rise in the nuclear factor NF- tractive mode. Reduced oxygen levels to placental
κB migrating to the nucleus which stimulates tran- tissues caused by infection and reduced placental
scription of the CRH gene [106]. In numerous tissues, perfusion may also trigger placental CRH-R agonist
MyD88 acts as an adaptor protein that binds to the production as lowered oxygen levels caused
cytoplasmic side of the TLR and when activated increases in Ucn2 and Ucn3 mRNA production in
attracts interluekin-1 receptor-associated kinase 1 cultures of early first term villous explants and term
(IRAK-1), IRAK-4 and TNF receptor associated cultured placental cells [41]. There may be inter-
factor-6 (TRAF6). IRAK-1 is phosphorylated by play between placental CRH-R agonists and the
IRAK-4 and it and TRAF6 dissociate from the TLR, immune system that are especially significant dur-
TRAF6 then forms a complex with TGF-beta activat- ing infection. Urocortin has been found to exert the
ed kinase-1 (TAK1), TAK1 binding protein-1 (TAB1) following effects on cultured placental cells: (a) it
and TAB2. This complex is later joined by ubiquitin- attenuates the release of the inflammatory cytokine
conjugating enzyme-13 (Ubc13) and ubiquitin- tumour necrosis factor alpha (TNF-α) stimulated by
conjugating enzyme E2 variant 1 isoform A (Uev1A) lipopolysaccharide, and (b) it stimulates the release
which activate TAK1. TAK1 phosphorylates and acti- of interleukin-4 and interleukin-10, both effects are
vates the IKK complex. The IKK complex phosphor- mediated by CRH-R2 [103]. Urocortin may push
ylates IκB complexed with NF-κB, causing the IκB to the immune status of the tissue from an inflamma-
be ubiquitylated and degraded in the proteasomes, tory state (Th2) to an anti-inflammatory state (Th1)
thus releasing NF-κB to travel into the nucleus where and thereby serve to limit the inflammation of the
it can activate target genes [93]. Future experiments uterine and placental tissues [103].
Roles of placental CRH in pregnancy and childbirth 569

The finding that NF-κB stimulates the CRH gene function in depression with early life stress — a systematic
suggests that cellular pathways that interact with the review. Acta Neuropsychiatr 24:4–15
5. Bale TL, Chen A (2012) Minireview: CRF and Wylie
pro-inflammatory NF-κB could change the timing of Vale: a story of 41 amino acids and a Texan with grit.
childbirth. For example, oxidised cholesterol metabolites Endocrinology 153:2556–2561
(oxysterols) are produced in increasing concentrations 6. Bale TL, Vale WW (2004) CRF and CRF receptors: role in
during pregnancy and have recently been found to in- stress responsivity and other behaviors. Annu Rev Phar-
macol 44:525–557
crease the phosphorylation of NF-κB and translocation 7. Barker DJP (2006) Adult consequences of fetal growth
to the nucleus in cultured trophoblast [3]. In many cell restriction. Clin Obstet Gynecol 49:270–283
types other than placenta, cellular stressors such as hyp- 8. Bauer ME (2005) Stress, glucocorticoids and ageing of the
oxia are known to activate IKK leading to the activation immune system. Stress 8:69–83
9. Beck CT (2008) State of the science on postpartum de-
of NF-κB [54, 61]. It will be important to determine pression: what nurse researchers have contributed — part
whether this mechanism is working in the placenta as it 1. MCN Am J Matern Child Nurs 33:121–126
would yield further evidence to suggest that when the 10. Bloom SL, Sheffield JS, McIntire DD, Leveno KJ (2001)
uterine environment becomes stressed, the response is to Antenatal dexamethasone and decreased birth weight.
Obstet Gynecol 97:485–490
activate mechanisms that increase CRH production and 11. Bortolotti B, Menchetti M, Bellini F, Montaguti MB,
bring forward parturition. Berardi D (2008) Psychological interventions for major
depression in primary care: a meta-analytic review of
randomized controlled trials. Gen Hosp Psychiat 30:293–
302
Conclusion 12. Breuiller-Fouche M, Germain G (2006) Gene and protein
expression in the myometrium in pregnancy and labor.
Reproduction 131:837–850
The possibility of reducing low birth weights, prevent- 13. Campbell EA, Linton EA, Wolfe CDA, Scraggs PR, Jones
ing foetal programming and helping women cope with MT, Lowry PJ (1987) Plasma corticotropin-releasing hor-
psychological distress due to hypothalamic–pituitary– mone concentrations during pregnancy and parturition. J
adrenal axis disruption is a powerful incentive to re- Clin Endocr Metab 64:1054–1059
14. Challis JRG, Matthews SG, Gibb W, Lye SJ (2000) Endo-
search groups around the globe to continue the push to crine and paracrine regulation of birth at term and preterm.
understand how placental urocortins and CRH regulate Endocr Rev 21:514–550
the physiology and psychology of the pregnant woman 15. Chang CL, Hsu SY (2004) Ancient evolution of stress-
and the physiology and health of her offspring. The regulating peptides in vertebrates. Peptides 25:1681–1688
16. Chen R, Lewis KA, Perrin MH, Vale WW (1993) Expres-
possibility of using pharmaceuticals such as CRH-R sion cloning of a human corticotropin-releasing-factor re-
blockers to counteract problems caused by CRH pro- ceptor. Proc Natl Acad Sci USA 90:8967–8971
duction outside the healthy range is one of the drivers 17. Chen L, Sooranna SR, Lei K, Kandola M, Bennett PR,
for concentrated research focus in this field. Liang Z, Grammatopoulos D, Johnson MR (2012) Cyclic
AMP increases COX-2 expression via mitogen-activated
kinase in human myometrial cells. J Cell Mol Med
16:1447–1460
18. Claes SJ (2004) Corticotropin-releasing hormone (CRH)
References in psychiatry: from stress to psychopathology. Ann Med
36:50–61
19. Cong B, Zhang L, Gao L, Ni X (2009) Reduced expression
1. Abou-Seif C, Shipman KL, Allars M, Norris MH, Chen of CRH receptor type 1 in upper segment human myome-
YX, Smith R, Nicholson RC (2012) Tissue specific epige- trium during labour. Reprod Biol Endocrinol 7:43
netic differences in CRH gene expression. Front Biosci 20. Coufalik AH, Monder C (1981) Stimulation of gluconeo-
17:713–725 genesis by cortisol in fetal rat liver in organ culture. En-
2. Aguilar HN, Mitchell BF (2010) Physiological pathways docrinology 108:1132–1137
and molecular mechanisms regulating uterine contractility. 21. DeBold CR, DeCherney GS, Jackson RV, Sheldon WR,
Hum Reprod Update 16:725–744 Alexander AN, Island DP, Rivier J, Vale W, Orth DN
3. Aye ILMH, Waddell BJ, Mark PJ, Keelan JA (2012) Oxy- (1983) Effect of synthetic ovine corticotropin-releasing
sterols exert proinflammatory effects in placental tropho- factor: prolonged duration of action and biphasic response
blasts via TLR4-dependent, cholesterol-sensitive of plasma adrenocorticotropin and cortisol. J Clin Endocr
activation of NF-κB. Mol Hum Reprod 18:341–353 Metab 57:294–298
4. Baes CVW, Tofoli SMC, Martins CMS, Juruena MF (2012) 22. Dores RM, Baron AJ (2011) Evolution of POMC: origin,
Assessment of the hypothalamic–pituitary–adrenal axis ac- phylogeny, posttranslational processing, and the melano-
tivity: glucocorticoid receptor and mineralocorticoid receptor cortins. Ann NY Acad Sci 1220:34–48
570 M. Thomson

23. Drake AJ, Seckl JR (2011) Transmission of programming 37. Hillhouse EW, Grammatopoulos DK (2002) Role of stress
effects across generations. Pediatr Endocrinol Rev 9:566– peptides during human pregnancy and labour. Reproduc-
578 tion 124:323–329
24. Fetalvero KM, Zhang P, Shyu M, Young BT, Hwa J, 38. Hillhouse EW, Grammatopoulos DK (2006) The molecu-
Young RC, Martin KA (2008) Prostacyclin primes preg- lar mechanisms underlying the regulation of the biological
nant human myometrium for an enhanced contractile re- activity of corticotropin-releasing hormone receptors:
sponse in parturition. J Clin Invest 118:3966–3979 implications for physiology and pathophysiology. Endocr
25. Florio P, Lowry PJ, Benedetto C, Galleri L, Torricelli M, Rev 27:260–286
Giovannelli A, Battista R, Reis FM, Petraglia F (2007) 39. Himes KP, Simhan HN (2011) Plasma corticotropin-
Maternal plasma corticotropin-releasing factor (CRF) and releasing hormone and cortisol concentrations and per-
CRF-binding protein (CRF-BP) levels in post-term preg- ceived stress among pregnant women with preterm and
nancy: effect of prostaglandin administration. Eur J Endo- term birth. Am J Perinatol 28:443–448
crinol 157:279–284 40. Holsboer F, Vonbardeleben U, Gerken A, Stalla GK, Muller
26. Gao L, Lu C, Xu C, Tao Y, Cong B, Ni X (2008) Differ- OA (1984) Blunted corticotropin and normal cortisol re-
ential regulation of prostaglandin production mediated by sponse to human corticotropin-releasing factor in depression.
corticotropin-releasing hormone receptor type 1 and type 2 New Engl J Med 311:1127
in cultured human placental trophoblasts. Endocrinology 41. Imperatore A, Rolfo A, Petraglia F, Challis JRG, Caniggia
149:2866–2876 I (2010) Hypoxia and preeclampsia: increased expression
27. Gao L, Lv C, Xu C, Li Y, Cui X, Gu H, Ni X (2012) of Urocortin 2 and Urocortin 3. Reprod Sci 17:833–843
Differential regulation of glucose transporters mediated by 42. Ishimoto H, Jaffe RB (2011) Development and function of
CRH receptor type 1 and type 2 in human placental troph- the human fetal adrenal cortex: a key component in the
oblasts. Endocrinology 153:1464–1471 feto-placental unit. Endocr Rev 32:317–355
28. Gilpin NW (2012) Corticotropin-releasing factor (CRF) 43. Jin D, He P, You X, Zhu X, Dai L, He Q, Liu C, Hui N,
and neuropeptide Y (NPY): effects on inhibitory transmis- Sha J, Ni X (2007) Expression of corticotropin-releasing
sion in central amygdala, and anxiety- and alcohol-related hormone receptor type 1 and type 2 in human pregnant
behaviors. Alcohol 46:329–337 myometrium. Reprod Sci 14:568–577
29. Gold PW, Loriaux DL, Roy A, Kling MA, Calabrese JR, 44. Karteris E, Hillhouse EW, Grammatopoulos D (2004)
Kellner CH, Nieman LK, Post RM, Pickar D, Gallucci W, Urocortin II is expressed in human pregnant myometrial
Avgerinos P, Paul S, Oldfield EH, Cutler GB, Chrousos cells and regulates myosin light chain phosphorylation:
GP (1986) Responses to corticotropin-releasing hormone potential role of the type-2 corticotropin-releasing hor-
in the hypercortisolism of depression and Cushing's dis- mone receptor in the control of myometrial contractility.
ease. New Engl J Med 314:1329–1335 Endocrinology 145:890–900
30. Grammatopoulos DK (2007) The role of CRH receptors 45. Karteris E, Markovic D, Chen J, Hillhouse EW,
and their agonists in myometrial contractility and quies- Grammatopoulos DK (2010) Identification of a novel
cence during pregnancy and labour. Front Biosci 12:561– corticotropin-releasing hormone type 1β-like receptor vari-
571 ant lacking exon 13 in human pregnant myometrium regu-
31. Grammatopoulos D, Dai Y, Chen J, Karteris E, Papadopoulou lated by estradiol-17β and progesterone. Endocrinology
N, Easton AJ, Hillhouse EW (1998) Human corticotropin- 151:4959–4968
releasing hormone receptor: differences in subtype expression 46. Khan H, Kusakabe KT, Wakitani S, Hiyama M, Kiso Y
between pregnant and nonpregnant myometria. J Clin Endocr (2011) Quantitative expression and immunohistochemical
Metab 83:2539–2544 detection of glucose transporters, GLUT1 and GLUT3 in
32. Grammatopoulos D, Stirrat GM, Williams SA, Hillhouse the rabbit placenta during successful pregnancy. J Vet Med
EW (1996) The biological activity of the corticotropin- Sci 73:1177–1183
releasing hormone receptor–adenylate cyclase complex in 47. Khan RN, Matharoo-Ball B, Arulkumaran S, Ashford MLJ
human myometrium is reduced at the end of pregnancy. J (2001) Potassium channels in the human myometrium.
Clin Endocr Metab 81:745–751 Exp Physiol 86:255–264
33. Hahn T, Barth S, Graf R, Engelmann M, Beslagic D, Reul 48. Kipmen-Korgun D, Ozmen A, Unek G, Simsek M, Demir
JMHM, Holsboer F, Dohr G, Desoye G (1999) Placental R, Korgun ET (2012) Triamcinolone up-regulates GLUT 1
glucose transporter expression is regulated by glucocorti- and GLUT 3 expression in cultured human placental en-
coids. J Clin Endocr Metab 84:1445–1452 dothelial cells. Cell Biochem Funct 30:47–53
34. Handley SL, Dunn TL, Waldron G, Baker JM (1980) 49. Korebrits C, Ramirez MM, Watson L, Brinkman E, Bocking
Tryptophan, cortisol and puerperal mood. Brit J Psychiat AD, Challis JRG (1998) Maternal corticotropin-releasing
136:498–508 hormone is increased with impending preterm birth. J Clin
35. Hayashi M, Sakata M, Takeda T, Yamamoto T, Okamoto Endocr Metab 83:1585–1591
Y, Sawada K, Kimura A, Minekawa R, Tahara M, Tasaka 50. Kovo M, Schreiber L, Ben-Haroush A, Asalee L, Seadia S,
K, Murata Y (2004) Induction of glucose transporter 1 Golan A, Bar J (2011) The placental factor in spontaneous
expression through hypoxia-inducible factor 1a under hyp- preterm labor with and without premature rupture of mem-
oxic conditions in trophoblast-derived cells. J Endocrinol branes. J Perinat Med 39:423–429
183:145–154 51. Kramer MS, Lydon J, Seguin L, Goulet L, Kahn SR,
36. Hertelendy F, Zakar T (2004) Regulation of myometrial McNamara H, Genest J, Dassa C, Chen MF, Sharma S,
smooth muscle functions. Curr Pharm Design 10:2499–2517 Meaney MJ, Thomson S, Van Uum S, Koren G, Dahhou
Roles of placental CRH in pregnancy and childbirth 571

M, Lamoureux J, Platt RW (2009) Stress pathways to of depressed pregnant women: a pilot study. J Affect Disor-
spontaneous preterm birth: the role of stressors, psycho- ders 130:300–305
logical distress, and stress hormones. Am J Epidemiol 67. Orth DN, Mount CD (1987) Specific high-affinity binding
169:1319–1326 protein for human corticotropin-releasing hormone in nor-
52. Langdown ML, Sugden MC (2001) Enhanced placental mal human plasma. Biochem Biophys Res Commun
GLUT1 and GLUT3 expression in dexamethasone- 143:411–417
induced fetal growth retardation. Mol Cell Endocrinol 68. Paiva P, Whitehead C, Saglam B, Palmer K, Tong S (2011)
185:109–117 Measurement of mRNA transcripts of very high placental
53. Lee M-J, Wang Z, Yee H, Ma Y, Swenson N, Yang L, expression in maternal blood as biomarkers of preeclamp-
Kadner SS, Baergen RN, Logan SK, Garabedian MJ, sia. J Clin Endocr Metab 96:E1807–E1815
Guller S (2005) Expression and regulation of glucocorti- 69. Palazidou E (2012) The neurobiology of depression. Brit
coid receptor in human placental villous fibroblasts. En- Med Bull 101:127–145
docrinology 146:4619–4626 70. Pato MD, Adelstein RS (1980) Dephosphorylation of the
54. Ling J, Kumar R (2012) Crosstalk between NFkB and 20,000-dalton light chain of myosin by two different phos-
glucocorticoid signaling: a potential target of breast cancer phatases from smooth muscle. J Biol Chem 255:6535–
therapy. Cancer Lett 322:119–126 6538
55. Linton EA, Perkins AV, Woods RJ, Eben F, Wolfe CDA, 71. Pearlstein T (2008) Perinatal depression: treatment options
Behan DP, Potter E, Vale WW, Lowry PJ (1993) Cortico- and dilemmas. J Psychiat Neurosci 33:302–318
tropin releasing hormone-binding protein (CRH-BP): plas- 72. Peckett AJ, Wright DC, Riddell MC (2011) The effects of
ma levels decrease during the 3rd trimester of normal glucocorticoids on adipose tissue lipid metabolism. Me-
human pregnancy. J Clin Endocr Metab 76:260–262 tabolism 60:1500–1510
56. Linton EA, Wolfe CDA, Behan DP, Lowry PJ (1988) A 73. Perrin M, Donaldson C, Chen R, Blount A, Berggren T,
specific carrier substance for human corticotrophin releasing Bilezikjian L, Sawchenko P, Vale W (1995) Identification
factor in late gestational maternal plasma which could mask of a second corticotropin-releasing factor receptor gene
the ACTH-releasing activity. Clin Endocrinol 28:315–324 and characterization of a cDNA expressed in heart. P Natl
57. Lockwood CJ (1999) Stress-associated preterm delivery: Acad Sci USA 92:2969–2973
the role of corticotropin-releasing hormone. Am J Obstet 74. Petraglia F, Sawchenko PE, Rivier J, Vale W (1987) Evi-
Gynecol 180:S264–S266 dence for local stimulation of ACTH secretion by
58. Magiakou MA, Mastorakos G, Rabin D, Dubbert B, Gold corticotropin-releasing factor in human placenta. Nature
PW, Chrousos GP (1996) Hypothalamic corticotropin- 328:717–719
releasing hormone suppression during the postpartum pe- 75. Power ML, Williams LE, Gibson SV, Schulkin J, Helfers J,
riod: implications for the increase in psychiatric manifes- Zorrilla EP (2010) Pattern of maternal circulating CRH in
tations at this time. J Clin Endocr Metab 81:1912–1917 laboratory-housed squirrel and owl monkeys. Am J Prima-
59. Mancuso RA, Schetter CD, Rini CM, Roesch SC, Hobel tol 72:1004–1012
CJ (2004) Maternal prenatal anxiety and corticotropin- 76. Rivier C, Brownstein M, Spiess J, Rivier J, Vale W (1982)
releasing hormone associated with timing of delivery. Psy- In vivo corticotropin-releasing factor-induced secretion of
chosom Med 66:762–769 adrenocorticotropin, β-endorphin, and corticosterone. En-
60. Mandl M, Ghaffari-Tabrizi N, Haas J, Nöhammer G, Desoye docrinology 110:272–278
G (2006) Differential glucocorticoid effects on proliferation 77. Robinson BG, Arbiser JL, Emanuel RL, Majzoub JA
and invasion of human trophoblast cell lines. Reproduction (1989) Species-specific placental corticotropin releasing
132:159–167 hormone messenger RNA and peptide expression. Mol
61. Melvin A, Mudie S, Rocha S (2011) Mechanism of Cell Endocrinol 62:337–341
hypoxia-induced NFkB. Cell Cycle 10:879–882 78. Robinson BG, Emanuel RL, Frim DM, Majzoub JA (1988)
62. Myers B, McKlveen JM, Herman JP (2012) Neural regu- Glucocorticoid stimulates expression of corticotropin-
lation of the stress response: the many faces of feedback. releasing hormone gene in human placenta. P Natl Acad
Cell Mol Neurobiol 32:683–694 Sci USA 85:5244–5248
63. Nitta A, Ohmiya M, Sometani A, Itoh M, Nomoto H, 79. Romero R, Espinoza J, Kusanovic JP, Gotsch F, Hassan S,
Furukawa Y, Furukawa S (1999) Brain-derived neurotro- Erez O, Chaiworapongsa T, Mazor M (2006) The preterm
phic factor prevents neuronal cell death induced by corti- parturition syndrome. BJOG-Int J Obstet Gy 113:17–42
costerone. J Neurosci Res 57:227–235 80. Romero R, Gómez R, Chaiworapongsa T, Conoscenti G,
64. Nugent BM, Tobet SA, Lara HE, Lucion AB, Wilson ME, Cheol Kim J, Mee Kim Y (2001) The role of infection in
Recabarren SE, Paredes AH (2012) Hormonal program- preterm labour and delivery. Paediatr Perinat Epidemiol
ming across the lifespan. Horm Metab Res 44:577–586 15:41–56
65. O’Keane V, Lightman S, Patrick K, Marsh M, Papadopoulos 81. Ryabinin AE, Tsoory MM, Kozicz T, Thiele TE, Neufeld-
AS, Pawlby S, Seneviratne G, Taylor A, Moore R (2011) Cohen A, Chen A, Lowery-Gionta EG, Giardino WJ, Kaur
Changes in the maternal hypothalamic–pituitary–adrenal ax- S (2012) Urocortins: CRF's siblings and their potential role
is during the early puerperium may be related to the postpar- in anxiety, depression and alcohol drinking behavior. Al-
tum ‘blues’. J Neuroendocrinol 23:1149–1155 cohol 46:349–357
66. O'Keane V, Lightman S, Marsh M, Pawlby S, Papadopoulos 82. Saijonmaa O, Laatikainen T, Wahlström T (1988)
AS, Taylor A, Moore R, Patrick K (2011) Increased pitui- Corticotrophin-releasing factor in human placenta: localiza-
tary–adrenal activation and shortened gestation in a sample tion, concentration and release in vitro. Placenta 9:373–385
572 M. Thomson

83. Sandman CA, Davis EP, Buss C, Glynn LM (2011) Pre- 99. Thomson M, Smith R (1989) The action of hypothalamic
natal programming of human neurological function. Int J and placental corticotropin releasing factor on the cortico-
Pept 2011:837596 trope. Mol Cell Endocrinol 62:1–12
84. Shibasaki T, Odagiri E, Shizume K, Ling N (1982) 100. Tizabi Y, Aguilera G (1992) Desensitization of the hypo-
Corticotropin-releasing factor-like activity in human pla- thalamic–pituitary–adrenal axis following prolonged ad-
cental extracts. J Clin Endocr Metab 55:384–386 ministration of corticotropin-releasing hormone or
85. Sibai B, Meis PJ, Klebanoff M, Dombrowski MP, Weiner vasopressin. Neuroendocrinology 56:611–618
SJ, Moawad AH, Northen A, Iams JD, Varner MW, Caritis 101. Torricelli M, Ignacchiti E, Giovannelli A, Merola A, Scarpetti
SN, O'Sullivan MJ, Miodovnik M, Leveno KJ, Conway D, E, Calonaci G, Picciolini E, Florio P, Reis FM, Linton EA,
Wapner RJ, Carpenter M, Mercer B, Ramin SM, Thorp Petraglia F (2006) Maternal plasma corticotrophin-releasing
JM, Peaceman AM, Gabbe S (2005) Plasma CRH mea- factor and urocortin levels in post-term pregnancies. Eur J
surement at 16 to 20 weeks' gestation does not predict Endocrinol 154:281–285
preterm delivery in women at high-risk for preterm deliv- 102. Torricelli M, Novembri R, Bloise E, De Bonis M, Challis
ery. Am J Obstet Gynecol 193:1181–1186 JR, Petraglia F (2011) Changes in placental CRH, urocor-
86. Smith R, Cubis J, Brinsmead M, Lewin T, Singh B, Owens tins, and CRH-receptor mRNA expression associated with
P, Chan EC, Hall C, Adler R, Lovelock M, Hurt D, Rowley preterm delivery and chorioamnionitis. J Clin Endocr
M, Nolan M (1990) Mood changes, obstetric experience Metab 96:534–540
and alterations in plasma cortisol, beta-endorphin and cor- 103. Torricelli M, Voltolini C, Bloise E, Biliotti G, Giovannelli
ticotropin releasing hormone during pregnancy and the A, De Bonis M, Imperatore A, Petraglia F (2009) Urocor-
puerperium. J Psychosom Res 34:53–69 tin increases IL-4 and IL-10 secretion and reverses LPS-
87. Smith R, Maiti K (2012) The placenta, a transducer linking induced TNF-α release from human trophoblast primary
maternal nutrition to adult disease in the offspring? Endo- cells. Am J Reprod Immunol 62:224–231
crinology 153:1572–1574 104. Tyson EK, Smith R, Read M (2009) Evidence that
88. Smith R, Nicholson RC (2007) Corticotrophin releasing corticotropin-releasing hormone modulates myometrial
hormone and the timing of birth. Front Biosci 12:912–918 contractility during human pregnancy. Endocrinology
89. Smith R, Thomson M (1991) Neuroendocrinology of the 150:5617–5625
hypothalamo–pituitary–adrenal axis in pregnancy and the 105. Tzschoppe AA, Struwe E, Dörr HG, Goecke TW, Beckmann
puerperium. Baillière Clinical Endoc 5:167–186 MW, Schild RL, Dötsch J (2010) Differences in gene ex-
90. Stalla GK, Hartwimmer J, von Werder K, Muller OA pression dependent on sampling site in placental tissue of
(1984) Ovine (o) and human (h) corticotrophin releasing fetuses with intrauterine growth restriction. Placenta 31:178–
factor (CRF) in man: CRF-stimulation and CRF- 185
immunoreactivity. Acta Endocrinol (Copenh) 106:289– 106. Uh A, Nicholson RC, Gonzalez GV, Simmons CF, Gombart
297 A, Smith R, Equils O (2008) Lipopolysaccharide stimulation
91. Stevens MY, Challis JRG, Lye SJ (1998) Corticotropin- of trophoblasts induces corticotropin-releasing hormone ex-
releasing hormone receptor subtype 1 is significantly up- pression through MyD88. Am J Obstet Gynecol
regulated at the time of labor in the human myometrium. J 199:317.e1–317.e6
Clin Endocr Metab 83:4107–4115 107. Uh A, Simmons CF, Bresee C, Khoury N, Gombart AF,
92. Struwe E, Berzl G, Schild R, Blessing H, Drexel L, Hauck Nicholson RC, Kocak H, Equils O (2009) MyD88 and
B, Tzschoppe A, Weidinger M, Sachs M, Scheler C, TRIF mediate the cyclic adenosine monophosphate
Schleussner E, Dötsch J (2010) Microarray analysis of (cAMP) induced corticotropin releasing hormone (CRH)
placental tissue in intrauterine growth restriction. Clin expression in JEG3 choriocarcinoma cell line. Reprod Biol
Endocrinol 72:241–247 Endocrinol 7:74
93. Takeda K, Akira S (2004) TLR signaling pathways. Semin 108. Vale W, Spiess J, Rivier C, Rivier J (1981) Characteriza-
Immunol 16:3–9 tion of a 41-residue ovine hypothalamic peptide that stim-
94. Thomson M (1998) Does the CRH binding protein shield ulates secretion of corticotropin and beta-endorphin.
the anterior pituitary from placental CRH? Endocrine Science 213:1394–1397
9:221–226 109. Vallabhapurap S, Karin M (2009) Regulation and function
95. Thomson M (2008) The effects of placental corticotrophin of NF-κB transcription factors in the immune system. Ann
releasing hormone on the physiology and psychology of Rev Immunol 27:693–733
the pregnant woman. Curr Women Health Rev 4:270–279 110. Vaughan J, Donaldson C, Bittencourt J, Perrin MH,
96. Thomson M, Chan EC, Falconer J, Madsen G, Geraghty S, Lewis K, Sutton S, Chan R, Turnbull AV, Lovejoy D,
Curryer N, Boulton J, Smith R (1990) Desensitization of Rivier C, Rivier J, Sawchenko PE, Vale W (1995)
superfused isolated ovine anterior pituitary cells to human Urocortin, a mammalian neuropeptide related to fish
corticotropin-releasing factor. J Neuroendocrinol 2:181–187 urotensin I and to corticotropin-releasing factor. Nature
97. Thomson M, Chan EC, Falconer J, Madsen G, Smith R 378:287–292
(1988) Secretion of corticotropin-releasing hormone by 111. Wang B, Parobchak N, Rosen T (2012) RelB/NF-κB2
superfused human placental fragments. Gynecol Endocri- regulates corticotropin-releasing hormone in the human
nol 2:87–100 placenta. Mol Endocrinol 26:1356–1369
98. Thomson F, Craighead M (2008) Innovative approaches 112. Weiser MJ, Osterlund C, Spencer RL (2011) Inhibitory
for the treatment of depression: targeting the HPA axis. effects of corticosterone in the hypothalamic paraventricu-
Neurochem Res 33:691–707 lar nucleus (PVN) on stress-induced adrenocorticotrophic
Roles of placental CRH in pregnancy and childbirth 573

hormone secretion and gene expression in the PVN and 116. Wyrwoll CS, Kerrigan D, Holmes MC, Seckl JR,
anterior pituitary. J Neuroendocrinol 23:1231–1240 Drake AJ (2012) Altered placental methyl donor trans-
113. Wood IS, Trayhurn P (2003) Glucose transporters (GLUT port in the dexamethasone programmed rat. Placenta
and SGLT): expanded families of sugar transport proteins. 33:220–223
Br J Nutr 89:3–9 117. Xu C, Gao L, You X, Dai L, Li Y, Gu H, Slater DM,
114. Woods RJ, Grossman A, Saphier P, Kennedy K, Ur E, Olson DM, Ni X (2011) CRH acts on CRH-R1 and -
Behan D, Potter E, Vale W, Lowry PJ (1994) Association R2 to differentially modulate the expression of large-
of human corticotropin-releasing hormone to its binding conductance calcium-activated potassium channels in
protein in blood may trigger clearance of the complex. J human pregnant myometrium. Endocrinology 152:4406–
Clin Endocr Metab 78:73–76 4417
115. Wu G, Imhoff-Kunsch B, Girard AW (2012) Biological 118. Zhao FQ, Keating AF (2007) Functional properties and
mechanisms for nutritional regulation of maternal health genomics of glucose transporters. Curr Genomics 8:113–
and fetal development. Paediatr Perinat Epidemiol 26:4–26 128

You might also like