ArticlePDF Available

Chronic maternal stress inhibits the capacity to up-regulate placental 11 -hydroxysteroid dehydrogenase type 2 activity

Authors:

Abstract

This study investigated the effects of acute and chronic restraint stress during the third week of pregnancy on placental 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) activity in rats. Acute exposure to stress on gestational day 20 immediately up-regulated placental 11beta-HSD2 activity by 160%, while chronic stress from day 14 to day 19 of pregnancy did not significantly alter basal 11beta-HSD2 activity. However, the latter reduced the capacity to up-regulate placental 11beta-HSD2 activity in the face of an acute stressor by 90%. Thus, immediate up-regulation of 11beta-HSD2, the feto-placental barrier to maternal corticosteroids, may protect the fetus against stress-induced high levels of maternal corticosteroids, but exposure to chronic stress greatly diminishes this protection.
RAPID COMMUNICATION
Chronic maternal stress inhibits the capacity to up-regulate
placental 11-hydroxysteroid dehydrogenase type 2 activity
Leonie A M Welberg, K V Thrivikraman and Paul M Plotsky
Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, WMB suite 4000, 101 Woodruff Cir, Atlanta, Georgia 30322, USA
(Requests for offprints should be addressed to L A M Welberg; Email: leonie.welberg@emory.edu)
Abstract
This study investigated the eects of acute and chronic
restraint stress during the third week of pregnancy on
placental 11-hydroxysteroid dehydrogenase type 2 (11-
HSD2) activity in rats. Acute exposure to stress on
gestational day 20 immediately up-regulated placental
11-HSD2 activity by 160%, while chronic stress from
day 14 to day 19 of pregnancy did not significantly alter
basal 11-HSD2 activity. However, the latter reduced the
capacity to up-regulate placental 11-HSD2 activity in
the face of an acute stressor by 90%. Thus, immediate
up-regulation of 11-HSD2, the feto-placental barrier to
maternal corticosteroids, may protect the fetus against
stress-induced high levels of maternal corticosteroids,
but exposure to chronic stress greatly diminishes this
protection.
Journal of Endocrinology (2005) 186, R7–R12
Introduction
Animal studies of prenatal stress, environmental enrich-
ment and maternal separation have shown that events early
in life can alter the set points of the hypothalamic–
pituitary–adrenal (HPA) and corticotropin-releasing factor
(CRF) systems with permanent behavioural and endocrine
consequences (Ladd et al. 2000, Welberg & Seckl 2001).
The mechanisms underlying these programming eects
are still unknown, but exposure to elevated levels of
glucocorticoids during a time of rapid brain development
may be a major factor (Welberg & Seckl 2001). Indeed,
exposing pregnant rats to the synthetic glucocorticoid
dexamethasone results in ospring with a hyperactive
HPA axis and elevated CRF expression in the amygdala
(Welberg et al. 2001). In addition, stress-induced eleva-
tions in maternal glucocorticoid levels have been shown to
underlie at least some of the eects of prenatal stress
(Barbazanges et al. 1996). However, access of maternal
corticosterone to the fetus is regulated, in part, by the
high-anity, high-eciency type 2 isoform of the placen-
tal enzyme 11-hydroxysteroid dehydrogenase (11-
HSD2), that rapidly converts corticosterone (in rats) and
cortisol (in humans) into their inactive metabolites (11-
dehydrocorticosterone and cortisone respectively) (Seckl
& Meaney 2004). Animal studies have shown that
pharmacological inhibition of placental 11-HSD2 during
pregnancy results in ospring with HPA hyperactivity
and an anxious phenotype with elevated CRF expression
in the amygdala (Welberg et al. 2000). Placental 11-
HSD2 activity varies significantly between individuals, in
both rats (Benediktsson et al. 1993) and humans (Stewart
et al. 1995). Since pregnant dams produce far more cortico-
sterone than fetuses, moderate decreases in 11-HSD2
activity would result in relatively large increases in
corticosterone reaching the fetal blood stream. Thus, in
order to understand the programming eects of endogen-
ous maternal corticosteroids it is crucial to understand the
regulation of their access to the fetus.
Few studies have addressed the in vivo regulation of
placental 11-HSD2, but data from recent in vitro studies
suggest that factors associated with stress may play a role:
Catecholamines reduced 11-HSD2 gene transcription
via activation of alpha-adrenergic receptors (Sarkar et al.
2001), while the synthetic glucocorticoid dexamethasone
increased 11-HSD2 activity and gene transcription (van
Beek et al. 2004) in cultured human trophoblasts. The
present study aimed to investigate the eects of acute and
chronic stress during the third week of pregnancy on
placental 11-HSD2 activity in rats.
Materials and Methods
Animals
Experiments were performed in accordance with NIH
Guidelines for the care and use of laboratory animals and
R7
Journal of Endocrinology (2005) 186, R7–R12
0022–0795/05/0186–R7 2005 Society for Endocrinology Printed in Great Britain
DOI: 10.1677/joe.1.06374
Online version via http://www.endocrinology-journals.org
all protocols were approved by the Emory University
Institutional Animal Care and Use Committee. Timed-
pregnant Long–Evans rats (Charles River Laboratories,
Inc, Wilmington, MA, USA) arrived in our facilities on
the morning of day 13 of pregnancy (day 1 being the day
after mating occurred). All animals used in Experiment 1
(see below) arrived in the same transport, as did all animals
used in Experiment 2. Rats were housed in standard (W
LH: 203215 cm) cages with corn-cob bedding.
Food and water were available ad libitum, and animals were
maintained on a 12:12 light:dark cycle (lights on 0700 h).
Experimental protocol
Experiment 1 was performed to establish whether acute
or chronic stress aected placental 11-HSD2 activity.
Pregnant rats were randomly assigned to one of three
treatment groups: (1) rats in the chronic-stress group (CS,
n=7) were restrained in flat-bottom rodent restrainers
(approximately 825 cm) for 45 min on day 14 of preg-
nancy, and then twice daily for 30 or 45 min, once in the
morning and once in the afternoon, until day 19 of
pregnancy. On day 20 of pregnancy CS rats were deeply
anesthetized with isoflurane, immediately after which
caesarean section was performed; (2) rats in the acute-stress
group (AS, n=6) were left undisturbed until day 20 of
pregnancy. On that day, AS rats were restrained for
45 min and immediately afterwards anesthetized with
isoflurane and subjected to caesarean surgery; (3) a control
group of unstressed rats (NS, n=7) was left undisturbed
until the morning of day 20 of pregnancy, when NS rats
were also anesthetized and subjected to caesarean section.
Experiment 2, carried out separately from Experiment 1,
was performed to establish whether chronic stress expo-
sure altered placental 11-HSD2 activity in response to an
acute stressor. Here, pregnant rats were weighed and then
assigned to one of two groups: (1) rats in the chronic +
acute stress group (CAS, n=6) were restrained from day
14 of pregnancy onwards as the CS rats in Experiment 1
described above, but underwent a final, acute, 45 min
restraint session on day 20 of pregnancy immediately prior
to anaesthesia, weighing and caesarean section; (2) a
control group of unstressed rats (NS, n=6) was left
undisturbed until the morning of day 20 of pregnancy
when they also underwent anaesthesia, weighing and
caesarean section.
Sample collection In both experiments, dams were
sacrificed on day 20 of pregnancy between 0900 and
1100 h. Once rats were anesthetized, the abdominal cavity
was opened, the uterus exposed and two (Experiment 1)
or four (Experiment 2) feto-placental units were quickly
dissected. Each feto-placental unit was weighed (Experi-
ment 2 only), after which fetus and placenta were separ-
ated. The placenta was then rapidly frozen on powdered
dry ice and stored at 70 C until further processing.
Tissue Processing To determine the eect of maternal
stress on placental 11-HSD2 activity, in both Experiment
1 and 2 two placentas per pregnancy were homogenized
together, after which enzyme activity was measured as
described below, yielding one value per pregnancy. To
determine the correlation between 11-HSD2 activity
and feto-placental weight, the additional two placentas per
pregnancy obtained in Experiment 2 were weighed and
then processed individually for enzyme activity measure-
ments. Placentas were homogenized using a Powergen
Model 125 homogenizer (Fisher Scientific, Atlanta, GA,
USA) in 1 ml buer (1 PBS containing 0·25 M sucrose)
per placenta. Protein concentrations of the samples
were determined using a BCA protein assay kit (Pierce,
Rockford, IL, USA).
11-HSD2 assay Enzyme activity was estimated using
a radiometric conversion assay according to protocols
described previously (Benediktsson et al. 1993), with small
variations. Briefly, 1 mg/ml protein of placental homoge-
nate was incubated with 500 nM NAD and 12 nM of
[1,2,6,7]
3
H-corticosterone (specific activity, 75·6 Ci/
mmol) in a final volume of 500 µl 1X PBS containing
0·25 M sucrose at 37 C for 30 min. The reaction was
terminated by the addition of 2 ml ethyl acetate (Fisher
Scientific). Steroids were extracted using ethyl acetate and
separated by means of thin-layer chromatography using
chloroform-ethanol (92:8) as solvent (Ethanol: Fisher
Scientific). Spots corresponding to corticosterone and
11-dehydrocorticosterone were visualized under ultra
violet light, cut out, transferred to vials containing liquid
scintillant, and their radioactivity was measured in a
beta-counter (1209 Rackbeta; LKB/Wallac/Perkin
Elmer, Boston, MA, USA). All samples were assayed in
duplicate. Blank controls were included in all assays, as
well as samples assayed as described above but with
addition of 40 µM carbenoxolone, an inhibitor of 11-
HSD. Activity of 11-HSD2 in each sample was estimated
by calculating the fractions of
3
H-dehydrocorticosterone
and
3
H-corticosterone (NEN/Perkin Elmer). Unless
otherwise specified, all reagents obtained from Sigma.
Data analysis Conversion levels are expressed as a
percentage of (unstressed) control values. Data from
Experiment 1 were analyzed using one-way ANOVA.
When the overall response was significant, post-hoc com-
parisons were performed using the Tukey HSD test. Data
from Experiment 2 were analyzed using unpaired t-tests.
Significance was set at P< 0·05.
Results
Experiment 1
Analysis of variance on conversion levels revealed a
significant eect of maternal stress on placental 11-HSD2
L A M WELBERG and others · Maternal stress and placental 11-HSD2 activityR8
www.endocrinology-journals.orgJournal of Endocrinology (2005) 186, R7–R12
activity (F=19·57, P< 0·0001). Post-hoc analysis showed
that acute stress increased 11-HSD2 activity by 160%
compared with activity in unstressed pregnant rats
(P< 0·0005), and by 84% compared with activity in
chronically-stressed pregnant rats (P< 0·001). Chronic
stress did not significantly aect placental 11-HSD2
activity (P=0·26) 16 h after the last exposure to stress
(Figure 1).
Experiment 2
As shown in Table 1, NS and CAS dams had similar body
weights on day 13 and day 20 of pregnancy, and their
percentage weight gain was not significantly dierent.
CAS reduced weights of whole feto-placental units on
gestational day 20, but not placental weights (Table 1).
A t-test showed that combined chronic and acute stress
increased placental 11-HSD2 activity by 16% compared
with activity in placentas from unstressed pregnancies
(t=2·33, P< 0·05) (Figure 2).
Correlations between placental 11-HSD2 activity and
weights of the feto-placental units as measured on gesta-
tional day 20 are shown in Table 2. Enzyme activity
correlated negatively with feto-placental weight (Figure 3)
and with frozen placental weight, although only the
Figure 1 Effect of acute or chronic exposure to stress during
pregnancy on placental 11-HSD2 activity. Stress took place
between days 14 and 20 of pregnancy; feto-placental units were
harvested on day 20 of gestation. NS=no stress (n=7); AS=acute
stress (n=6); CS=chronic stress (n=7); **P< 0·001; ***P< 0·0005
Table 1 Body weight gain and feto-placental weights in unstressed
dams (NS, n=6) and in dams exposed to chronic + acute stress
(CAS, n=6). Stress took place between days 14 and 20 of
pregnancy, feto-placental units of NS (n=23) and CAS (n=24)
pregnancies were harvested on day 20 of pregnancy. Frozen
placental weights were also measured in NS (n=12) and CAS
(n=12) pregnancies
NS CAS P-value
Body weight GD13 (g) 2608 2757 0·17
Body weight GD20 (g) 29810 3198 0·16
Weight gain (% start weight) 17·30·9 16·01·4 0·45
Feto-placental weight (g) 2·730·04 2·620·03 0·03*
Placental weight (mg) 42816 43119 0·90
NB: One feto-placental unit in the NS group was not weighted. *denotes a
significant difference. GD; Gestational day.
Figure 2 Effect of exposure to combined chronic and acute
exposure to stress during pregnancy on placental 11-HSD2
activity. Stress took place between days 14 and 20 of pregnancy;
feto-placental units were harvested on day 20 of gestation.
NS=no stress (n=6); CAS=chronic+acute stress (n=6); *P< 0·05
Table 2 Correlations (R-values) between placental 11-HSD2
activity and weights of feto-placental units. Stress took place
between days 14 and 20 of pregnancy; feto-placental units were
harvested on day 20 of gestation
Feto-placental weight Placental weight
All units (n=24) 0·47* 0·31
Unstressed (NS, n=12) 0·31 0·31
Stressed (CAS, n=12) 0·61* 0·57
*correlation significant at P< 0·05.
Figure 3 Correlation between feto-placental weight and placental
11-HSD2 activity. Feto-placental units were harvested on
gestational day 20 from unstressed (white circles, n=12) and CAS
pregnancies (black circles, n=12). R=-0·46, P=0·02.
Maternal stress and placental 11-HSD2 activity · L A M WELBERG and others R9
www.endocrinology-journals.org Journal of Endocrinology (2005) 186, R7–R12
former reached statistical significance. These correlations
disappeared when only values from unstressed pregnancies
were taken into account, but became stronger when
considering only values from stressed pregnancies (Table 2).
Discussion
This is, to our knowledge, the first study that assessed
placental 11-HSD2 activity in response to maternal
stress. Placental 11-HSD2 is thought to act as a barrier
against maternal glucocorticoids and thus to protect the
fetus from potential harmful eects that the elevated levels
of these steroids can exert during development (Seckl &
Meaney 2004). The finding that placental 11-HSD2
activity increased in response to an acute stressor is
important since it shows that plasma corticosteroids pro-
duced by the dam in response to a single stressor are not
necessarily harmful to the fetus, provided that enzyme
activity is high enough to ‘inactivate’ them. Importantly,
however, the present study also showed that the capacity
to adapt placental 11-HSD2 activity in response to an
acute stressor was greatly reduced by previous exposure to
chronic stress.
In the present study CAS reduced feto-placental weight
on day 20 of gestation. Intrauterine growth retardation is a
common finding in prenatal stress studies (Patin et al.
2002, Lesage et al. 2004), as well as after administration of
synthetic glucocorticoids during pregnancy (Welberg et al.
2001). This indicates that stress-induced glucocorticoids
indeed reached the fetus in this study. Moreover, feto-
placental weight correlated negatively with 11-HSD2
activity, especially in the CAS group, suggesting that the
units with the largest growth retardation were exposed
to the highest levels of maternal corticosterone and up-
regulated their placental 11-HSD2 activity in an attempt
to prevent more corticosterone entering the fetal blood.
CAS-induced reduction in feto-placental weight
(around 100 mg) cannot be accounted for by the parallel
reduction in placental weight, as NS and CAS placentas
diered only by about 3 mg. Thus, although fetal weights
were not measured, it is likely that CAS reduced both
placental and fetal weights while slightly increasing
placental 11-HSD2 activity.
The combination of increased placental 11-HSD2
activity and decreased in utero growth appears to partly
contradict an earlier report in which placental 11-HSD2
activity correlated positively with birth weight and nega-
tively with placental weight (Benediktsson et al. 1993), but
several explanations are possible. In the present study, the
correlation between feto-placental weight and enzyme
activity is not necessarily a causal one, as the increased
placental 11-HSD2 activity in CAS pregnancies probably
resulted from both the acute stress exposure immediately
before harvesting and previous exposures, while the
placental (and likely, fetal) growth retardation in CAS was
due to the chronicity of the stressor. In contrast, in
Benediktsson et al. (1993), fetuses and placentas were
taken from unstressed pregnancies, thus linking ‘basal’
placental 11-HSD2 activity with birth weight. This
correlation probably does describe a causal relationship, as
artificially inhibiting placental 11-HSD2 activity without
stressing the pregnant dams also reduced intrauterine
growth (Welberg et al. 2000), as did chronic exposure
during pregnancy to synthetic glucocorticoids, which are
not metabolized by 11-HSD2 (Welberg et al. 2001).
Another important dierence between the present
study and that by Benediktsson et al. (1993) is that the
latter measured body weight at term, whereas here, feto-
placental weights were recorded on day 20 of gestation,
three days before expected delivery. Crucially, both
expression and activity of placental 11-HSD2 drop dra-
matically between gestational days 20 and 22 (Burton et al.
1996, Waddell et al. 1998) in the labyrinth zone of the
placenta, the site of maternal–fetal transfer, and this will
likely change existing correlations between weight and
enzyme activity on those days. Taken together, it is likely
that in the present study CAS-induced maternal cortico-
sterone reached the fetal blood stream in spite of increased
enzyme activity.
An alternative explanation for our finding that chronic
stress reduced the capacity to respond the acute stress with
an up-regulation of placental 11-HSD2 activity may be
that repetition of the restraint procedure caused it no
longer to be perceived as stressful. This interpretation
would be supported by the fact that CAS dams did not
show reduced weight gain during pregnancy, in contrast
to previous findings (Darnaudery et al. 2004). Since
maternal plasma corticosterone was not measured, it is
impossible to verify the stressfulness of repeated exposure
to restraint in this study. However, other studies have
shown that repeated restraint during pregnancy reliably
elevated maternal plasma corticosterone levels (Ward &
Weisz 1984, Barbazanges et al. 1996, Weinstock 2005)
and repeated restraint has been used many times as a
prenatal stressor with long-term eects on the ospring
(Barbazanges et al. 1996, Lesage et al. 2004). In addition,
as mentioned before, feto-placental weights from CAS
pregnancies were smaller than those from unstressed
pregnancies, confirming the common finding of fetal
growth retardation in prenatal stress paradigms (Patin et al.
2002, Lesage et al. 2004). It is important to note that dams
used in the present study arrived in our facilities on day 12
of pregnancy, and an eect of the stress of the transporta-
tion on basal placental 11-HSD2 activity or its response
to chronic or acute stress cannot be ruled out.
Although no other studies have investigated regulation
of placental 11-HSD2 by stress per se, it has been shown
that its gene expression in human trophoblast cells is
rapidly inhibited by catecholamines via activation of alpha-
adrenergic receptors (Sarkar et al. 2001). Moreover,
glucocorticoids can both up- and downregulate placental
L A M WELBERG and others · Maternal stress and placental 11-HSD2 activityR10
www.endocrinology-journals.orgJournal of Endocrinology (2005) 186, R7–R12
11-HSD2 mRNA expression, depending on species,
timing and mode of administration (Kerzner et al. 2002,
Ma et al. 2003, van Beek et al. 2004). Another study
reported a reduction in ovine placental 11-HSD2 activity
in response to chronically elevated glucocorticoid levels
(Clarke et al. 2002). Importantly, none of the above studies
investigated immediate regulation of placental 11-HSD2
activity in response to glucocorticoids, although this pre-
sumably would be the most ecient way to inactivate
stress-induced corticosteroids from maternal blood.
A recent investigation showed that activity of the renal
11-HSD2 enzyme, which is identical to that in the
placenta, is up-regulated within at least two hours (the
earliest time point studied) by corticosterone injections,
but also by stress in adrenalectomized rats (Zallocchi et al.
2004). This indicates that 11-HSD2 activity can be
regulated by adrenal steroids as well as by extra-adrenal
factors, although the exact mechanism remains to be
determined. Thus, the capacity of placental 11-HSD2
activity to rapidly increase in response to stress in combi-
nation with an attenuated maternal HPA reactivity during
pregnancy (Neumann et al. 1998) may function to control
access of corticosteroids to the fetal blood stream, ensuring
the proper level of glucocorticoids necessary for normal
growth and maturation.
In conclusion, this study showed that immediate up-
regulation of 11-HSD2, the feto-placental barrier to
maternal corticosteroids, may protect the fetus against
stress-induced elevations of maternal corticosteroids,
but exposure to chronic stress greatly diminishes this
protection.
Funding
This work was supported by a Young Investigator Award
from the National Alliance for Research on Schizophrenia
and Depression (LAMW). The authors declare that they
have no conflict of interest that would preclude their
impartiality of this work.
References
Barbazanges A, Piazza PV, Le MoalM&Maccari S 1996 Maternal
glucocorticoid secretion mediates long-term eects of prenatal stress.
Journal of Neuroscience 16 3943–3949.
vanBeekJP,GuanH,JulanL&YangK2004 Glucocorticoids
stimulate the expression of 11 beta-hydroxysteroid dehydrogenase
type 2 in cultured human placental trophoblast cells. Journal of
Clinical Endocrinology and Metabolism 89 5614–5621.
Benediktsson R, Lindsay RS, Noble J, Seckl JR & Edwards CR 1993
Glucocorticoid exposure in utero: new model for adult
hypertension. Lancet 341 339–341.
Burton PJ, Smith RE, Krozowski ZS & Waddell BJ 1996 Zonal
distribution of 11 beta-hydroxysteroid dehydrogenase types 1
and 2 messenger ribonucleic acid expression in the rat placenta
and
decidua during late pregnancy. Biology of Reproduction 55
1023–1028.
Clarke KA, Ward JW, Forhead AJ, Giussani DA & Fowden AL 2002
Regulation of 11 beta-hydroxysteroid dehydrogenase type 2 activity
in ovine placenta by fetal cortisol. Journal of Endocrinology 172
527–534.
Darnaudery M, Dutriez I, Viltart O, Morley-FletcherS&Maccari S
2004 Stress during gestation induces lasting eects on emotional
reactivity of the dam rat. Behavioural Brain Research 153
211–216.
Kerzner LS, Stonestreet BS, Wu KY, SadowskaG&Malee MP 2002
Antenatal dexamethasone: eect on ovine placental 11 beta-
hydroxysteroid dehydrogenase type 2 expression and fetal growth.
Pediatric Research 52 706–712.
Ladd CO, Huot RL, Thrivikraman KV, Nemero CB, Meaney MJ
& Plotsky PM 2000 Long-term behavioral and neuroendocrine
adaptations to adverse early experience. Progress in Brain Research
122 81–103.
Lesage J, Del-Favero F, Leonhardt M, Louvart H, Maccari S, Vieau D
& Darnaudery M 2004 Prenatal stress induces intrauterine growth
restriction and programmes glucose intolerance and feeding
behaviour disturbances in the aged rat. Journal of Endocrinology 181
291–296.
Ma XH, Wu WX & Nathanielsz PW 2003 Gestation-related and
betamethasone-induced changes in 11 beta-hydroxysteroid
dehydrogenase types 1 and 2 in the baboon placenta. American
Journal of Obstetrics and Gynecology 188 13–21.
Neumann ID, Johnstone HA, Hatzinger M, Liebsch G, Shipston M,
Russell JA, Landgraf R, Douglas AJ 1998 Attenuated
neuroendocrine responses to emotional and physical stressors in
pregnant rats involve adenohypophysial changes. Journal of Physiology
508 289–300.
Patin V, Lordi B, Vincent A, Thoumas JL, VaudryH&Caston J
2002 Eects of prenatal stress on maternal behavior in the rat. Brain
Research. Developmental Brain Research 139 1–8.
Sarkar S, Tsai SW, Nguyen TT, Plevyak M, Padbury JF & Rubin LP
2001 Inhibition of placental 11 beta-hydroxysteroid dehydrogenase
type 2 by catecholamines via alpha-adrenergic signaling. American
Journal of Physiology Regulatory, Integrative and Comparative
Physiology 281 R1966–R1974.
Seckl JR & Meaney MJ 2004 Glucocorticoid programming. Annals of
the New York Academy of Sciences 1032 63–84.
Stewart PM, Rogerson FM & Mason JI 1995 Type 2 11 beta-
hydroxysteroid dehydrogenase messenger ribonucleic acid and
activity in human placenta and fetal membranes: its relationship to
birth weight and putative role in fetal adrenal steroidogenesis.
Journal of Clinical Endocrinology and Metabolism 80 885–890.
Waddell BJ, Benediktsson R, Brown RW & Seckl JR 1998 Tissue-
specific messenger ribonucleic acid expression of 11 beta-
hydroxysteroid dehydrogenase types 1 and 2 and the glucocorticoid
receptor within rat placenta suggests exquisite local control of
glucocorticoid action. Endocrinology 139 1517–1523.
Ward IL & Weisz J 1984 Dierential eects of maternal stress on
circulating levels of corticosterone, progesterone, and testosterone in
male and female rat fetuses and their mothers. Endocrinology 114
1635–1644.
Weinstock M 2005 The potential influence of maternal stress
hormones on development and mental health of the ospring.
Brain, Behavior, and Immunity 19 296–308.
Welberg LAM & Seckl JR 2001 Prenatal stress, glucocorticoids and
the programming of the brain. Journal of Neuroendocrinology 13
113–128.
Welberg LAM, Seckl JR & Holmes MC 2000 Inhibition of 11
beta-hydroxysteroid dehydrogenase, the foeto-placental barrier to
maternal glucocorticoids, permanently programs amygdala GR
mRNA expression and anxiety-like behaviour in the ospring.
European Journal of Neuroscience 12 1047–1054.
Welberg LAM, Seckl JR & Holmes MC 2001 Prenatal gluco-
corticoid programming of brain corticosteroid receptors and
Maternal stress and placental 11-HSD2 activity · L A M WELBERG and others R11
www.endocrinology-journals.org Journal of Endocrinology (2005) 186, R7–R12
corticotrophin- releasing hormone: possible implications for
behaviour. Neuroscience 104 71–79.
Zallocchi ML, Matkovic L, Calvo JC & Damasco MC 2004 Adrenal
gland involvement in the regulation of renal 11 beta-hydroxysteroid
dehydrogenase 2. Journal of Cellular Biochemistry 92 591–602.
Received 15 June 2005
Accepted 27 July 2005
Made available online as an
Accepted Preprint 27 July 2005
L A M WELBERG and others · Maternal stress and placental 11-HSD2 activityR12
www.endocrinology-journals.orgJournal of Endocrinology (2005) 186, R7–R12
... The influence of ACEs may occur much earlier as a form of chronic stress exposure from childhood. Animal models have shown a link between chronic stress exposure and an impaired ability to upregulate placental 11β-HSD2 in response to acute stress, diminishing the innate protective mechanism against fetal steroid overexposure [20]. While human studies to validate this same response are needed, this maladaptation may have exponential effects when considering the increased likelihood of ACE exposure, co-existing mood disorders, and an impaired ability to regulate placental 11β-HSD2 in response to acute stress. ...
Article
Full-text available
Adverse childhood experiences (ACEs) are extremely prevalent in the United States population. Although ACEs occurs in childhood, exposure to them has been associated with adverse future pregnancy outcomes and an increased risk of poorer social determinants of health, which further drive the risk of negative pregnancy outcomes. In addition, maternal ACE exposure has been linked to poor infant and child outcomes, highlighting the intergenerational transmission of risk from mother to child. While alterations along the Maternal–Placental–Fetal Hypothalamic–pituitary–adrenal (HPA) axis is hypothesized to be involved, the exact biological pathway underlying this intergenerational passage of risk is mostly unknown. This present work will highlight what is known about pregnancy-related stress hormone physiology, discuss the potential mechanisms of action of ACEs on cortisol regulation, and suggest opportunities for further clinical and translational studies.
... The study highlights stress as a critical environmental factor impacting fetal development, notably increasing the risk of cleft lip and palate [2,27,28]. ...
Article
Full-text available
Introduction: Cleft lip with or without cleft palate (CL/P) stands as the most common congenital facial anomaly, stemming from multifactorial causes. Objective: Our study aimed to ascertain the prevalence and characteristics of cleft palates, identify associated risk factors to inform prevention and prenatal detection for early intervention, and assess postoperative rehabilitation protocols for cleft palates. Design: This study employs a retrospective descriptive and clinical approach. Patients: The study includes 103 children with cleft palates treated at the Department of Head and Neck Surgery Clinic for Children and Young Adults, Department of Clinical Pediatrics, University of Warmia and Mazury. Methods: We conducted a thorough evaluation of records, considering variables such as sex, cleft type, maternal occupation, parental education, and family history of clefts. Data analysis was carried out using R software version GPL-3 and ordinal logistic regression analyses. Results: Notably, children born to mothers who experienced significant stress during pregnancy exhibited a 9.4-fold increase in the odds of having bilateral cleft palates. Conversely, no substantial evidence was found to support the influence of the child’s sex, birth order, body mass, maternal exposure to workplace toxins, infections, or drug toxicity on the dependent variable. Conclusions: Our findings suggest that children with parents who have a history of clefts and those with less educated mothers are more likely to develop bilateral cleft palates. Additionally, children born to mothers experiencing stress during pregnancy face an increased risk of bilateral cleft palates. It is important to note that there is a paucity of literature on rehabilitation following various cleft palate surgical techniques in children.
... [27] [28]. Enzym 11-beta-hydroksysteroid dehydrogenazy typu 2 (11beta HSD2), odpowiedzialny za regulację przepuszczalności bariery łożyskowej, jest regulowany w dół w przypadku stresu [29]. Ponadto w wczesnych etapach ciążykrytycznym momencie dla formowania masy twarzowejten enzym jest mniej reprezentowany. ...
Preprint
Full-text available
Introduction: Cleft lip with or without cleft palate (CL/P) is the most common congenital face anomaly in the world of multifactorial etiology. Objective: To determine the prevalence and characteristics of cleft palate. To identify associated risk factors to guide prevention and/or prenatal detection for early intervention and better health outcomes. To assess the rehabilitation protocols after operations in cleft palate. Design: Retrospective descriptive and clinical study. Patients: One hundred and three children with cleft palate treated in the Department of Head and Neck Surgery Clinic for Children and Young Adults, Department of Clinical Pediatrics, University of Warmia and Mazury, were included in the study. Methods: Records evaluation was conducted regarding sex, cleft type, mother occupation, parents education, histories of cleft in any of parents. The data were analyzed using R (R core team 2021) and the ordinal logistic regression analyses. Results: Children of stressed mothers during pregnancy had 9.4 times higher odds of having bilateral cleft. While there was no evidence that children sex, order of birth, and body mass as well as risk of maternal toxicity at work or reporting infections or drugs toxicity had a significant effect on the dependent variable. Conclusions: Children of parents with cleft history and having low educated mothers had higher probability of bilateral cleft palate. Children of stressed mothers during pregnancy had higher odds of having bilateral cleft. In literature, there is a lack of reports about rehabilitation in different operations methods in children with cleft palates.
... Sperm and egg being affected by the salient environmental experience and the consequent biological inheritance of its influences (Rodgers et al. 2013(Rodgers et al. , 2015Gapp et al. 2014aGapp et al. , 2020Radford et al. 2014;Milekic et al. 2015;Sharma et al. 2016;Dickson et al. 2018). Being exposed to salient environmental experiences as a fetus gestating in utero and the consequent alteration of development (Welberg et al. 2005;Mueller and Bale 2008;Bronson and Bale 2016). Post-natal salient environmental experiences disrupting caregiving and future generations being the recipients of that sub-optimal caregiving (Kaffman and Meaney 2007;Baram et al. 2012;Monk et al. 2012;Ward et al. 2013;O'Donnell et al. 2014;Kundakovic and Champagne 2015;Bell et al. 2016;Chen and Baram 2016). ...
Article
Full-text available
Evidence for parental environments profoundly influencing the physiology, biology and neurobiology of future generations has been accumulating in the literature. Recent efforts to understand this phenomenon and its underlying mechanisms have sought to use species like rodents and insects to model multi-generational legacies of parental experiences like stress and nutritional exposures. From these studies, we have come to appreciate that parental exposure to salient environmental experiences impact the cadence of brain development, hormonal responses to stress, and the expression of genes that govern cellular responses to stress, in offspring. Recent studies using chemosensory exposure have emerged as a powerful tool to shed new light on how future generations come to be influenced by environments to which parents are exposed. With a specific focus on studies that have leveraged such use of salient chemosensory experiences, this review synthesizes our current understanding of the concept, causes and consequences of the inheritance of chemosensory legacies by future generations and how this field of inquiry informs the larger picture of how parental experiences can influence offspring biology.
Chapter
The “Developmental Origins of Health and Disease” (DOHaD) hypothesis postulates that specific environmental influences during critical pre- and early postnatal phases "shape" or "programme" the formation of an individual's functional phenotype. Exposure to suboptimal nutritional conditions, substance abuse or maternal stress and infections may have a negative impact on development and thus increase the risk for different diseases and disorders at later life. On the other hand, favourable environmental conditions at early development are essential to induce adaptive processes that may help an individual to life successfully and survive in its later life environment. Increasing evidence reveals that these programming mechanisms are mediated by epigenetic mechanisms, thus epigenetics provide the crucial interface between genetic predispositions and the programming influences of the environment. Epigenetically mediated (re-)programming processes are also a hallmark of functional brain maturation, since most neurodevelopmental trajectories start during gestation and continue into early postnatal life and thus are particularly sensitive to environmental influences. Deleterious programming effects that end up in dysfunctional brain circuits increase the risk for mental disorders, psychopathologies or cognitive deficits. However, depending on the type, duration and developmental time-point of the programming environmental influence, “healthy” adaptive processes in the brain may occur promoting resilience and improved stress coping. This book article explains the main concepts of early-life programming and summarizes major findings with respect to the environmentally driven development of functional brain circuits and its underlying epigenetic mechanisms.
Article
Objective This study aimed to test the hypothesis that being pregnant and delivering during the coronavirus disease 2019 (COVID-19) pandemic was associated with changes in gestational weight gain (GWG) or frequency of small- (SGA) or large-for-gestational-age (LGA) neonates. Study Design Secondary analysis of a multicenter observational cohort comparing pregnant people who delivered during the COVID-19 pandemic (June–December 2020) to people who delivered prior to the pandemic (March–December 2019). Those with multiple gestations, fetuses with major congenital anomalies, implausible GWG values, unavailable body mass index (BMI), or who were severe acute respiratory syndrome coronavirus-2-positive were excluded. The primary outcome was frequency of optimal recommended GWG based on prepregnancy BMI. Neonatal outcomes included birth weight, ponderal index, and frequency of SGA, LGA, and small head circumference for live births. Multivariable regression analysis was used to assess associations between exposure to the pandemic and outcomes. Results A total of 10,717 pregnant people were included in our analysis. A total of 4,225 pregnant people were exposed to the pandemic and 6,492 pregnant people delivered prior to the COVID-19 pandemic. Pregnant people exposed to the pandemic were older and more likely to have gestational diabetes. The frequency of appropriate GWG was 28.0% during the pandemic and 27.6% before the pandemic (adjusted odds ratio [aOR]: 1.02, 95% confidence interval [CI]: 0.93–1.11). Excessive GWG was more likely (54.9 vs. 53.1%; aOR: 1.08, 95% CI: 1.001–1.17), and inadequate GWG was less likely during the pandemic (17.0 vs. 19.3%; aOR: 0.86, 95% CI: 0.77–0.95). The frequency of SGA was 5.4% during the pandemic and 6.1% before the pandemic (aOR: 0.90, 95% CI: 0.76–1.06), and the frequency of LGA was 16.0% during the pandemic versus 15.0% before the pandemic (aOR: 1.06, 95% CI: 0.95–1.18). Other neonatal outcomes including birth weight percentile (62.1 [35.8–83.2] vs. 60.2 [34.4–82.2]; adjusted mean difference (aMD) = 1.50, 95% CI: −0.28 to 3.29), ponderal index (2.6 g/cm3 [2.4–2.8] in both groups; aMD = 0.01, 95% CI: 0.00–0.02), and small head circumference for livebirths (<10th percentile [8.2 vs. 8.1%; aOR: 1.03, 95% CI: 0.89–1.19], <3rd percentile [3.5 vs. 3.1%; aOR: 1.16, 95% CI: 0.93–1.44]) were similar between groups as well. Conclusion Being pregnant and delivering during the COVID-19 pandemic was associated with a higher likelihood of excessive GWG and a lower likelihood of inadequate GWG. Key Points
Article
Abstract Background The placenta acts as a buffer to regulate the degree of fetal exposure to maternal cortisol through the 11-Beta Hydroxysteroid Dehydrogenase isoenzyme type 2 (11-β HSD2) enzyme. We conducted a systematic review and meta-analysis to assess the effect of prenatal psychological distress (PPD) on placental 11-β HSD2 gene expression and explore the related mechanistic pathways involved in fetal neurodevelopment. Methods We searched PubMed, Embase, Scopus, APA PsycInfo®, and ProQuest Dissertations for observational studies assessing the association between PPD and 11-β HSD2 expression in human placentas. Adjusted regression coefficients (β) and corresponding 95% confidence intervals (CIs) were pooled based on three contextual PPD exposure groups: prenatal depression, anxiety symptoms, and perceived stress. Results Of 3159 retrieved records, sixteen longitudinal studies involving 1,869 participants across seven countries were included. Overall, exposure to PPD disorders showed weak negative associations with the placental 11-β HSD2 gene expression as follows: prenatal depression (β -0.01, 95% CI 0.05 -0.02, I2=0%), anxiety symptoms (β -0.02, 95% CI 0.06 - 0.01, I2=0%), and perceived stress (β -0.01 95% CI 0.06 - 0.04, I2=62.8%). Third-trimester PPD exposure was more frequently associated with lower placental 11-β HSD2 levels. PPD and placental 11-β HSD2 were associated with changes in cortisol reactivity and the development of adverse health outcomes in mothers and children. Female-offspring were more vulnerable to PPD exposures. Conclusion The study presents evidence of a modest role of prenatal psychological distress in regulating placental 11-β HSD2 gene expression. Future prospective cohorts utilizing larger sample sizes or advanced statistical methods to enhance the detection of small effect sizes should be planned. Additionally, controlling for key predictors such as the mother’s ethnicity, trimester of PPD exposure, mode of delivery, and infant sex is crucial for valid exploration of PPD effects on fetal programming.
Article
Full-text available
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo–pituitary–adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Article
This study examines the interplay between maternal depression/anxiety and infant temperament's developmental trajectory in 1687 Swedish‐speaking mother–infant dyads from Uppsala County (2009–2019), Sweden. The sample includes a high proportion of university‐educated individuals and a low share of foreign‐born participants. Maternal depressive and anxiety symptoms were assessed using the Edinburgh Postnatal Depression Scale during gestational weeks 17 and 32 and postpartum at week 6. Multinomial regression explored associations between maternal variables and infant temperament trajectories at 6 weeks, 12 months, and 18 months. Prenatal anxiety is associated with the high‐rising infant difficult temperament trajectory, while prenatal depression/anhedonia is associated with the stable‐medium trajectory, attenuated postpartum. Associations between infant temperament and maternal mood depended on timing (pre/postpartum) and symptom type (depression/anhedonia vs. anxiety).
Article
Full-text available
There is growing evidence that stressors occurring during pregnancy can impair biological and behavioral adaptation to stress in the adult offspring. Mechanisms by which stress in the pregnant rat can influence development of the offspring are still unknown. In the present study, we investigated the involvement of maternal corticosterone secretion during pregnancy on the hypothalamo-pituitary-adrenal axis activity of adult offspring. We investigated stress-induced corticosterone secretion and hippocampal type I and type II corticosteroid receptors in male adult rats submitted to prenatal stress born to either mothers with intact corticosterone secretion or mothers in which stress-induced corticosterone secretion was blocked by adrenalectomy with substitutive corticosterone therapy. Repeated restraint during the last week of pregnancy was used as prenatal stressor. Furthermore, the specific role of an injection of corticosterone before the restraint stress on adrenalectomized mothers with substitutive corticosterone treatment was also studied. We report here that blockade of the mother's stress-induced glucocorticoid secretion suppresses the prolonged stress-induced corticosteroid response and the decrease in type I hippocampal corticosteroid receptors usually observed in prenatally stressed adults. Conversely, corticosterone administered during stress, to mothers in which corticosterone secretion is blocked, reinstates the effects of prenatal stress. These results suggest for the first time that stress-induced increases in maternal glucocorticoids may be a mechanism by which prenatal stress impairs the development of the adult offspring's glucocorticoid response.
Article
Full-text available
There is growing evidence that stressors occurring during pregnancy can impair biological and behavioral adaptation to stress in the adult offspring. Mechanisms by which stress in the pregnant rat can influence development of the offspring are still unknown. In the present study, we investigated the involvement of maternal corticosterone secretion during pregnancy on the hypothalamo-pituitary-adrenal axis activity of adult offspring. We investigated stress-induced corticosterone secretion and hippocampal type I and type II corticosteroid receptors in male adult rats submitted to prenatal stress born to either mothers with intact corticosterone secretion or mothers in which stress-induced corticosterone secretion was blocked by adrenalectomy with substitutive corticosterone therapy. Repeated restraint during the last week of pregnancy was used as prenatal stressor. Furthermore, the specific role of an injection of corticosterone before the restraint stress on adrenalectomized mothers with substitutive corticosterone treatment was also studied. We report here that blockade of the mother's stress-induced glucocorticoid secretion suppresses the prolonged stress-induced corticosteroid response and the decrease in type I hippocampal corticosteroid receptors usually observed in prenatally stressed adults. Conversely, corticosterone administered during stress, to mothers in which corticosterone secretion is blocked, reinstates the effects of prenatal stress. These results suggest for the first time that stress-induced increases in maternal glucocorticoids may be a mechanism by which prenatal stress impairs the development of the adult offspring's glucocorticoid response.
Article
Placental 11beta-hydroxysteroid dehydrogenase (11beta-HSD) regulates transplacental passage of maternal glucocorticoids to the fetus and is thus a key determinant of fetal glucocorticoid levels. It has also been proposed that placental 11beta-HSD expression may influence local glucocorticoid actions by regulating access of corticosterone to the glucocorticoid receptor (GR) or mineralocorticoid receptor (MR). Therefore, the present study used a rat model to assess whether the GR or MR are coexpressed with the two forms of 11beta-HSD (types 1 and 2) in the placental labyrinth zone, the major site of maternal-fetal transfer, and in the basal zone, the primary site of placental hormone synthesis. In situ hybridization analysis was used to assess messenger RNA (mRNA) expression for the GR, MR, 11beta-HSD-1, and 11beta-HSD-2 in the two placental zones on days 16, 19 and 22 of pregnancy (term = day 23). Whereas expression of the GR appeared relatively unchanged in both zones at these three stages of pregnancy, that of 11beta-HSD-1 clearly increased in the labyrinth zone but fell in basal zone, whereas the opposite pattern of expression was observed for 11beta-HSD-2. MR expression was not detected at any stage. The pattern of placental 11beta-HSD-2 mRNA expression over days 16, 19, and 22 of pregnancy was paralleled by changes in 11beta-HSD-2-specific bioactivity, but despite clear expression of 11beta-HSD-1 mRNA, no bioactivity attributable to this enzyme was measurable in either placental zone. To assess the role of fetal adrenal maturation on these changes in 11beta-HSD, two experimental models, maternal adrenalectomy and fetectomy, were employed. Maternal adrenalectomy on day 13 advanced maturation of the fetal adrenal cortex but had no effect on 11beta-HSD-2 bioactivity in either of the placental zones at day 19. Placental 11beta-HSD-2 bioactivity on day 22 was also unaffected by fetectomy 3 or 6 days earlier. In conclusion, the consistent expression of the GR in the two placental zones late in pregnancy suggests that concomitant and marked changes in 11beta-HSD-1 and 11beta-HSD-2 expression could have a major influence on glucocorticoid action in the placenta at this time. Moreover, the changes in 11beta-HSD expression appear to be unrelated to development of the fetal adrenal cortex and are likely to reduce the placental glucocorticoid barrier near the end of pregnancy.
Article
Testosterone, progesterone, and corticosterone titers were measured by RIA in plasma of stressed and control pregnant rats and their male and female fetuses on days 17, 18, 19, and 21 of gestation and on the day of birth. The regimen of stress used (three 45-min periods of restraint under intense illumination daily from days 14-21 of pregnancy) causes failure of masculinization and defeminization of behavioral potentials in male offspring. In fetuses of both sexes, corticosterone titers increased sharply between days 17 and 18 postconception (pc) to a peak that was maintained through day 19 and then declined. This pattern resembled that obtained for testosterone in control male fetuses in which the levels of testosterone also rose sharply between days 17 and 18 pc. Corticosterone titers were elevated in samples obtained during the middle of the stress session from both the mothers (serum) and their male and female fetuses (plasma). Increased corticosterone levels were no longer evident in samples obtained from fetuses 75-165 min after the end of a stress session. Testosterone titers were altered by stress only in male fetuses. Their testosterone levels were elevated on day 17 pc, and the surge on days 18 and 19 pc, characteristic of control males, was absent in samples obtained 75-165 min after termination of stress. Progesterone titers were not affected by stress in either mothers or their fetuses. In both stressed and control groups, progesterone concentrations were identical in male and female fetuses, were higher in mothers than in fetuses, and declined in both fetuses and mothers between days 19 and 21 pc. Thus, a persistent effect of stress was observed only on testosterone and only in males.
Article
Two isoforms of 11 beta-hydroxysteroid dehydrogenase (11 beta HSD) have been described which catalyze the interconversion of cortisol (F) to cortisone (E). 11 beta HSD activity has previously been reported in placenta and fetal membranes, where its role may be to protect the developing fetus from glucocorticoid excess. Furthermore, in the rat, an association between placental 11 beta HSD activity and the subsequent development of hypertension in the offspring has been reported. We have characterized the isoforms of 11 beta HSD in human fetal membranes and dissected placental tissue at term and investigated the relationship between placental 11 beta HSD activity and fetal and placental weights. 11 beta HSD activity studies in the presence of 0.1 mumol/L F and NAD (indicative of type 2 isoform activity) revealed high levels of activity in trophoblast dissected free of vessels (561 +/- 87 pmol E/h.mg protein; n = 4) > undissected placenta > cotyledenous vessels dissected away from trophoblast > placental and reflected amnion. In contrast, in the presence of 2.5 mumol/L F and NADP (indicative of type 1 isoform activity), only decidua and chorion demonstrated significant levels of 11 beta HSD activity. Type 1 11 beta HSD activity in chorion was probably due to decidual contamination, in that it was absent in decidua-free fused chorion obtained from a twin pregnancy. In keeping with these data, type 1 11 beta HSD messenger ribonucleic acid (1.5 kilobases) was detected in decidua, but in no other tissue, and high levels of type 2 11 beta HSD messenger ribonucleic acid (1.9 kilobases) were found in undissected placenta and trophoblast. In 27 term placentas, 11 beta HSD activity varied from 194-448 pmol E/h.mg protein. There was a weak, but significant, positive correlation between term placental 11 beta HSD activity and fetal weight (r = 0.408; P = 0.034), but no correlation with placental weight. Thus, in man, the reported association of a small fetus and a large placenta predisposing to adult hypertension cannot be explained on the basis of defective 11 beta HSD activity. However, the placenta offers an immense reservoir for F clearance (1.73-7.95 mumol/min.placenta) and may be a principal factor driving fetal ACTH secretion and, hence, fetal adrenal steroidogenesis.
Article
Hypertension is strongly predicted by the combination of low birthweight and a large placenta. This association could be due to increased fetal exposure to maternal glucocorticoids. Fetal protection is normally effected by placental 11 beta-hydroxysteroid dehydrogenase (11 beta-OHSD), which converts physiological glucocorticoids to inactive products. We found that rat placental 11 beta-OHSD activity correlated positively with term fetal weight and negatively with placental weight. Offspring of rats treated during pregnancy with dexamethasone (which is not metabolised by 11 beta-OHSD) had lower birthweights and higher blood pressure when adult than did offspring of control rats. Increased fetal glucocorticoid exposure secondary to attenuated placental 11 beta-OHSD activity may link low birthweight and high placental weight with hypertension.
Article
Glucocorticoid action in several target tissues is dependent on expression of 11 beta-hydroxysteroid dehydrogenase (11 beta-HSD), and in the placenta 11 beta-HSD is thought to regulate transfer of active glucocorticoid to the fetus. This study compared expression of the two recognized 11 beta-HSD enzymes, types 1 and 2, in the rat placenta and decidua on Days 16 and 22 of gestation (term = Day 23). According to S1 nuclease protection analysis, although mRNA for 11 beta-HSD-1 was only just detectable in the labyrinth zone on Day 16, by Day 22 this expression had increased almost 20-fold. There was also an increase (approximately 2-fold) in 11 beta-HSD-1 mRNA in the basal zone between Days 16 and 22. In Day 16 decidua, 11 beta-HSD-1 mRNA was also highly expressed, but insufficient tissue was available for analysis on Day 22. Western blot analysis showed that immunoreactive 11 beta-HSD-1 (molecular mass 34 kDa) was present in those tissues with the highest 11 beta-HSD-1 mRNA expression (Day 16 decidua and Day 22 labyrinth zone). With respect to mRNA for 11 beta-HSD-2, high expression was observed in the decidua and labyrinth zone at Day 16, but in the latter this expression then declined 90% by Day 22. In contrast, expression of mRNA for 11 beta-HSD-2 increased more than 3-fold in the basal zone over the same period. Consistent with coexpression of the two 11 beta-HSD enzymes, both 11-oxoreductase and 11 beta-dehydrogenase bioactivity were clearly evident in all tissues, and each varied with stage of gestation. Specifically, 11 beta-dehydrogenase activity in the basal zone increased from 38 +/- 2% (mean +/- SEM) on Day 16 to 56 +/- 2% on Day 22, while 11-oxoreductase activity fell from 55 +/- 3% to 43 +/- 2% over the same period. In contrast, 11 beta-dehydrogenase activity in the labyrinth zone fell with advancing pregnancy (Day 16: 63 +/- 2%; Day 22: 48 +/- 2%). Both 11-oxoreductase (58 +/- 3%) and 11 beta-dehydrogenase (38 +/- 4%) activities were also evident in decidua at Day 16. In conclusion, this study shows that expression of 11 beta-HSD-1 and -2 is zone-specific in the placenta and maternal decidua. Moreover, opposite changes in the expression of the two enzymes occur in the basal and labyrinth zones of the placenta over the last days of pregnancy, indicative of distinct regulatory mechanisms and functional significance for the enzymes in the two placental zones.
Article
The responsiveness of the rat hypothalamo‐pituitary‐adrenal (HPA) axis and hypothalamo‐neurohypophysial system (HNS) to emotional (elevated plus‐maze) and physical (forced swimming) stressors and to administration of synthetic corticotrophin‐releasing hormone (CRH) was investigated during pregnancy and lactation. In addition to pregnancy‐related adaptations at the adenohypophysial level, behavioural responses accompanying the neuroendocrine changes were studied. Whereas basal (a.m.) plasma corticosterone, but not corticotrophin (adrenocorticotrophic hormone; ACTH), levels were increased on the last day (i.e. on day 22) of pregnancy, the stress‐induced rise in both plasma hormone concentrations was increasingly attenuated with the progression of pregnancy beginning on day 15 and reaching a minimum on day 21 compared with virgin control rats. A similar attenuation of responses to both emotional and physical stressors was found in lactating rats. Although the basal plasma oxytocin concentration was elevated in late pregnancy, the stress‐induced rise in oxytocin secretion was slightly lower in day 21 pregnant rats. In contrast to vasopressin, oxytocin secretion was increased by forced swimming in virgin and early pregnant rats indicating a differential stress response of these neurohypophysial hormones. The blunted HPA response to stressful stimuli is partly due to alterations at the level of corticotrophs in the adenohypophysis, as ACTH secretion in response to CRH in vivo (40 ng kg ⁻¹ , i.v.) was reduced with the progression of pregnancy and during lactation. In vitro measurement of cAMP levels in pituitary segments demonstrated reduced basal levels of cAMP and a lower increase after CRH stimulation (10 nM, 10 min) in day 21 pregnant compared with virgin rats, further indicating reduced corticotroph responsiveness to CRH in pregnancy. The reduced pituitary response to CRH in late pregnancy is likely to be a consequence of a reduction in CRH receptor binding as revealed by receptor autoradiography. [ ¹²⁵ I] CRH binding in the anterior pituitary was significantly reduced in day 11, 17 and 22 pregnant rats compared with virgin controls. Anxiety‐related behaviour of the animals as revealed by the time on and entries into the open arms of the elevated plus‐maze was different between virgin and pregnant rats with decreased number of entries indicating increased anxiety with the progression of pregnancy (except on pregnancy day 18). The emotional behaviour, however, was not correlated with the neuroendocrine responses. The results indicate that the reduced response of the HPA axis to stressors described previously during lactation is already manifested around day 15 of pregnancy in the rat and involves physiological adaptations at the adenohypophysial level. However, alterations in stressor perception at higher brain levels with the progression of pregnancy may also be involved.
Article
Glucocorticoids may underlie the association between prenatal stress, low birth weight and adult stress-associated disorders, e.g. hypertension and type 2 diabetes, increased hypothalamic-pituitary-adrenal (HPA) activity and affective dysfunction. Normally, 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) rapidly inactivates glucocorticoids in placenta and many foetal tissues, thus acting as a 'barrier' to maternal steroids. We investigated the effect of inhibiting foeto-placental 11beta-HSD in rats, using carbenoxolone (CBX), on subsequent HPA activity and regulation and stress-induced behaviour in adult offspring. Pregnant Wistar rats were injected with CBX (12.5 mg s.c.) or vehicle daily throughout pregnancy. CBX treatment reduced birth weight. Adult offspring of CBX-treated dams had persistently reduced body weight, increased basal corticosterone (CORT) levels, increased corticotropin-releasing hormone (CRH) and reduced glucocorticoid receptor (GR) mRNA in the hypothalamic paraventricular nucleus, though hippocampal GR and mineralocorticoid receptor (MR) mRNA expression were unaltered. In addition, these animals showed less grooming and rearing in an open field and reduced immobility in a forced swim test, and had increased GR mRNA expression in the basolateral (BLA), central (CEA) and medial (MEA) nuclei of the amygdala, with unaltered MR mRNA. These data suggest that disturbance of the foeto-placental enzymatic barrier to maternal glucocorticoids reduces birth and body weight, and produces permanent alterations of the HPA axis and anxiety-like behaviour in aversive situations. The behavioural and HPA effects may reflect GR gene programming in amygdala and hypothalamus, respectively. Foetal overexposure to endogenous glucocorticoids (prenatal stress or reduced activity of foeto-placental 11beta-HSD) may represent a common link between the prenatal environment, foetal growth and adult neuroendocrine and affective disorders.