ArticlePDF Available

Activation of Placental mTOR Signaling and Amino Acid Transporters in Obese Women Giving Birth to Large Babies

Authors:

Abstract and Figures

Context: Babies of obese women are often large at birth, which is associated with perinatal complications and metabolic syndrome later in life. The mechanisms linking maternal obesity to fetal overgrowth are largely unknown. Objective: We tested the hypothesis that placental insulin/IGF-I and mammalian target of rapamycin (mTOR) signaling is activated and amino acid transporter activity is increased in large babies of obese women. Design and setting: Pregnant women were recruited prospectively for collection of placental tissue at a university hospital and academic biomedical center. Patients or other participants: Twenty-three Swedish pregnant women with first trimester body mass index ranging from 18.5 to 44.9 kg/m(2) and with uncomplicated pregnancies participated in the study. Interventions: There were no interventions. Main outcome measures: We determined the phosphorylation of key signaling molecules (including Akt, IRS-1, S6K1, 4EBP-1, RPS6, and AMPK) in the placental insulin/IGF-I, AMPK, and mTOR signaling pathways. The activity and protein expression of the amino acid transporter systems A and L were measured in syncytiotrophoblast microvillous plasma membranes. Results: Birth weights (range, 3025-4235 g) were positively correlated to maternal body mass index (P < 0.05). The activity of placental insulin/IGF-I and mTOR signaling was positively correlated (P < 0.001), whereas AMPK phosphorylation was inversely (P < 0.05) correlated to birth weight. Microvillous plasma membrane system A, but not system L, activity and protein expression of the system A isoform SNAT2 were positively correlated to birth weight (P < 0.001). Conclusions: Up-regulation of specific placental amino acid transporter isoforms may contribute to fetal overgrowth in maternal obesity. This effect may be mediated by activation of insulin/IGF-I and mTOR signaling pathways, which are positive regulators of placental amino acid transporters.
Content may be subject to copyright.
Activation of Placental mTOR Signaling and Amino
Acid Transporters in Obese Women Giving Birth to
Large Babies
Nina Jansson,* Fredrick J. Rosario,* Francesca Gaccioli, Susanne Lager,
Helen N. Jones, Sara Roos, Thomas Jansson, and Theresa L. Powell
Center for Pregnancy and Newborn Research (F.J.R., F.G., S.L., T.J., T.L.P.), Department of
Obstetrics/Gynecology, University of Texas Health Science Center, San Antonio, Texas 78229; Division of
Pediatric Surgery (H.N.J.), Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229; and
Institutes of Neuroscience and Physiology (N.J.) and Biomedicine (S.R.), Sahlgrenska Academy at University of
Gothenburg, S-40530 Gothenburg, Sweden
Context: Babies of obese women are often large at birth, which is associated with perinatal com-
plications and metabolic syndrome later in life. The mechanisms linking maternal obesity to fetal
overgrowth are largely unknown.
Objective: We tested the hypothesis that placental insulin/IGF-I and mammalian target of rapamycin
(mTOR) signaling is activated and amino acid transporter activity is increased in large babies of obese
women.
Design and Setting: Pregnant women were recruited prospectively for collection of placental tissue
at a university hospital and academic biomedical center.
Patients or Other Participants: Twenty-three Swedish pregnant women with first trimester body mass
index ranging from 18.5 to 44.9 kg/m
2
and with uncomplicated pregnancies participated in the study.
Interventions: There were no interventions.
Main Outcome Measures: We determined the phosphorylation of key signaling molecules (in-
cluding Akt, IRS-1, S6K1, 4EBP-1, RPS6, and AMPK) in the placental insulin/IGF-I, AMPK, and mTOR
signaling pathways. The activity and protein expression of the amino acid transporter systems A
and L were measured in syncytiotrophoblast microvillous plasma membranes.
Results: Birth weights (range, 3025– 4235 g) were positively correlated to maternal body mass index
(P0.05). The activity of placental insulin/IGF-I and mTOR signaling was positively correlated (P
0.001), whereas AMPK phosphorylation was inversely (P0.05) correlated to birth weight. Mi-
crovillous plasma membrane system A, but not system L, activity and protein expression of the
system A isoform SNAT2 were positively correlated to birth weight (P0.001).
Conclusions: Up-regulation of specific placental amino acid transporter isoforms may contribute
to fetal overgrowth in maternal obesity. This effect may be mediated by activation of insulin/IGF-I
and mTOR signaling pathways, which are positive regulators of placental amino acid transporters.
(J Clin Endocrinol Metab 98: 105–113, 2013)
ISSN Print 0021-972X ISSN Online 1945-7197
Printed in U.S.A.
Copyright © 2013 by The Endocrine Society
doi: 10.1210/jc.2012–2667 Received July 3, 2012. Accepted October 9, 2012.
First Published Online November 12, 2012
* N.J. and F.J.R. contributed equally to this work.
Abbreviations: AMPK, AMP-activated kinase; BMI, body mass index; 4E-BP1, eukaryotic
initiation factor 4E-binding protein 1; GDM, gestational diabetes mellitus; IRS-1, insulin
receptor substrate 1; IUGR, intrauterine growth restriction; LGA, large for gestational age;
MeAIB,
14
C-methyl-aminoisobutyric acid; mTOR, mammalian target of rapamycin;
mTORC, mTOR complex; MVM, microvillous plasma membrane; PKC
, protein kinase C-
;
RPS6, ribosomal protein S6; SGK1, serum and glucocorticoid-regulated kinase 1; S6K1, S6
kinase 1; SNAT, sodium-dependent neutral amino acid transporter.
ORIGINAL ARTICLE
Endocrine Care
J Clin Endocrinol Metab, January 2013, 98(1):105–113 jcem.endojournals.org 105
Women entering pregnancy overweight [body mass
index (BMI), 25–29.9 kg/m
2
] or obese (BMI 30
kg/m
2
) have an increased risk to deliver a large for gesta-
tional age (LGA) baby, often defined as a birth weight
above the 90th centile (1–3). Large babies have an in-
creased risk for shoulder dystocia and plexus injury at
delivery (1, 2) and are susceptible to develop obesity, di-
abetes, and hypertension in childhood and later in life (4).
The mechanisms underlying the relationship between ex-
cess maternal adiposity and fetal overgrowth are not well
established.
To grow appropriately, the fetus is critically dependent
on nutrient supply across the placenta, which is deter-
mined by numerous factors including placental and um-
bilical blood flows, transplacental concentration gradi-
ents, and placental metabolism. In addition, the type,
number, and activity of transporter proteins in the syncy-
tiotrophoblast plasma membranes constitute an impor-
tant determinant for the transplacental transport of nu-
trients such as glucose and amino acids. In pregnancies
complicated by intrauterine growth restriction (IUGR),
placental nutrient transporters for amino acids, such as the
sodium-dependent system A (5, 6), are down-regulated.
Women having type 1 diabetes or developing gestational
diabetes mellitus (GDM) are more likely to give birth to a
LGA baby (7), and an increased placental nutrient trans-
port capacity may be one important factor contributing to
fetal overgrowth in these pregnancy complications. For
example, placental leucine transport has been shown to be
increased in GDM/LGA (8), and system A activity was
increased in microvillous plasma membrane (MVM) iso-
lated from placentas obtained from pregnancies compli-
cated by diabetes (8). In contrast to these findings, a pre-
vious study showed that system A amino acid transporter
activity is reduced, and the activity of system L is unaltered
in MVM vesicles isolated from type 1 diabetes pregnancies
with LGA babies (9). Placental glucose transport and glu-
cose transporter protein 1 protein expression were re-
ported to be increased in type 1 diabetes (10, 11). In ad-
dition, emerging evidence suggests that fatty acid
transport to the fetus may be increased in diabetes with or
without obesity, providing one possible explanation for
the increased adiposity observed in babies of mothers with
diabetes. For example, the activity of placental lipoprotein
lipase has been shown to be increased in pregnancies with
type 1 diabetes and fetal overgrowth (12), and placental
expression of the fatty acid binding protein 4 (13) and
endothelial lipase (14) is elevated in pregnancies of obese
women with diabetes. Although it is well established that
high prepregnancy BMI is strongly associated to fetal
overgrowth (1–3), the effect of maternal overweight and
obesity on placental function in women without diabetes
remains largely unknown (15, 16).
Placental nutrient transport is controlled by fetal, placen-
tal, and maternal factors. Placental mammalian target of
rapamycin (mTOR) constitutes a positive regulator of tro-
phoblast amino acid transporters (17, 18). In addition, in
vitro studies have demonstrated that hormones such as in-
sulin, IGF-I, and leptin, which are upstream regulators of
mTOR, stimulate placental transporters for amino acids
(19–22). Thus, placental growth and nutrient transport are
under the regulation of metabolic hormones (23–25). Obe-
sity in pregnancy is associated with perturbed maternal me-
tabolism and circulating hormone levels. For example, obese
pregnant women have higher serum levels of leptin, insulin,
and IL-6 in late pregnancy compared with pregnant women
with normal prepregnancy BMI (26). We recently extended
these observations and reported increased circulating levels
of leptin and insulin already in first trimester among over-
weight and obese women (27). Thus, it is possible that in-
creased levels of maternal hormones such as insulin, leptin,
and IGF-I provide a link between maternal obesity and fetal
overgrowth by up-regulation of placental nutrient transport
capacity. In the current study, we tested the hypothesis that
placental insulin/IGF-I and mTOR signaling is activated and
amino acid transporter activity is increased in large babies of
obese women.
Patients and Methods
Ethical approval
These studies conformed to the standards set by the latest
revision of the Declaration of Helsinki and were approved by the
Committee for Research Ethics at the University of Gothenburg.
Informed consent was obtained from subjects at recruitment.
After obtaining all the relevant clinical information, samples
were coded and deidentified. Some analyses were performed at
the University of Gothenburg, and deidentified samples were
subsequently transferred to the University of Texas Health Sci-
ence Center San Antonio for further studies.
Subjects
Pregnant women with an early pregnancy BMI [weight (ki-
lograms)/height (meters)
2
] ranging from 18.5 to 44.9 kg/m
2
were
enrolled in Gothenburg, Sweden, and placentas were collected
after term delivery. BMI was determined based on length and
weight measurements at the first prenatal visit at 8–12 wk ges-
tation. Estimated date of delivery was determined from the last
menstrual period and confirmed by ultrasound at 16–18 wk
gestation. When a large fetus was suspected based on clinical
signs, repeated ultrasounds were carried out to confirm acceler-
ation of fetal growth. Study subjects were recruited either im-
mediately before delivery (n 7) at the Sahlgrenska University
Hospital or in gestational wk 8–12 at the Lundby Prenatal Care
Center (n 16). Subjects recruited in early pregnancy were part
of a prospective cohort of 49 pregnant women described in detail
106 Jansson et al. Maternal Obesity and Placental Function J Clin Endocrinol Metab, January 2013, 98(1):105–113
elsewhere (27). The 16 subjects from this cohort included in the
current study represented the cases in which the placenta was ob-
tained immediately after delivery and therefore were a random sam-
ple of the larger cohort. The same inclusion and exclusion criteria
were used for all study subjects. The inclusion criteria were Scan-
dinavian heritage, good health, and age of at least 20 yr. The ex-
clusion criteria were smoking, vegetarianism, assisted reproduc-
tion, concurrent disease such as eating disorder, chronic
hypertension and diabetes, and development of pregnancy compli-
cations such as gestational diabetes, preeclampsia, or IUGR.
Preparation of placental homogenates and
syncytiotrophoblast microvillous membranes
Placentas were collected and weighed before trimming of the
cord and membranes. MVM vesicles were prepared as described
previously (8, 28). Briefly, placentas were immediately placed on
ice after delivery and dissected. The chorionic plate, amniotic
sac, and decidua were removed. Approximately 50 g of villous
tissue was cut into small pieces and rinsed with ice-cold physi-
ological saline. Tissue was placed in ice-cold buffer D [250 mM
sucrose, 0.7
Mpepstatin A, 1.6
Mantipain, 80
Maprotinin,
1mMEDTA, 10 mMHEPES-Tris (pH 7.4)] at 4 C and homog-
enized on ice using a polytron (Kinematika AG, Lucerne, Swit-
zerland). The homogenate was snap-frozen in liquid nitrogen
and stored at 80 C until analysis or further processing. To
prepare MVM vesicles, homogenates were thawed on ice and
then centrifuged twice at 10,000 gfor 15 min, and the resulting
supernatants were combined and centrifuged at 125,000 gfor
30 min. The pelleted crude membrane fraction was resuspended
in buffer D, and 12 mMMgCl
2
was added. The resulting sus-
pension was subjected to slow stirring on ice for 20 min. Sub-
sequently, the suspension was centrifuged for 10 min at 2500
g. The supernatant, which contained the MVM, was centrifuged
two times for 30 min at 125,000 g. Vesicles were aliquoted,
snap-frozen in liquid nitrogen, and stored at 80 C until use.
MVM enrichment was determined as the MVM/homogenate
ratio of alkaline phosphatase activity, which was assessed using
standard activity assays. Enrichment of alkaline phosphatase ac-
tivity in MVM was 30 6-fold and was independent of maternal
BMI. Protein content of the homogenates and MVM was deter-
mined by the method of Bradford.
Western blot
Protein expression of total and phosphorylated Akt (Thr-308
or Ser-473), insulin receptor substrate 1 (IRS-1; Tyr-612), AMP-
activated kinase (AMPK
; Thr-172), serum and glucocorticoid-
regulated kinase 1 (SGK1; Ser-422), protein kinase C-
(PKC
;
Ser-657), S6 kinase 1 (S6K1; Thr-389), eukaryotic initiation fac-
tor 4E-binding protein 1 (4E-BP1; Thr-37/46 or Thr-70), and
ribosomal protein S6 (RPS6; Ser-235/236) was analyzed in pla-
cental homogenates. The IRS-1 and PKC
antibodies were pur-
chased from Millipore (Billerica, MA), the SGK1 antibody was
obtained from Santa Cruz Biotechnology (Santa Cruz, CA), and
the remaining antibodies were purchased from Cell Signaling
Technology (Boston, MA). Protein expression of the three sys-
tem A amino acid transporter isoforms SNAT (sodium-depen-
dent neutral amino acid transporter) 1, -2, and -4 was determined
in MVM vesicles using Western blotting. A polyclonal SNAT2
antibody generated in rabbits (29) was a generous gift from Dr.
P. D. Prasad at the University of Georgia. Affinity-purified poly-
clonal anti-SNAT1 (raised against the peptide sequence
VPEDDNISNDSNDFT) and anti-SNAT4 antibodies (raised
against the peptide sequence YGEVEDELLHAYSKV) were gen-
erated in rabbits by Eurogentec (Seraing, Belgium). For negative
controls, the purified antigenic peptide was used in 15-fold ex-
cess to preabsorb antibody overnight at 4 C. Western blotting
was performed as described previously (30). Briefly, total protein
(10–20
g) from placental homogenate/MVM was loaded and
separated on Bis-Tris gels (7–12% acrylamide) and transferred
onto nitrocellulose membranes. Membrane blocking and anti-
body incubations were performed as described in the protocol
provided by the manufacturer. Subsequently, membranes were
incubated with the appropriate secondary peroxidase-labeled
antibodies for 1 h. After washing, bands were visualized using
enhanced chemiluminescence detection reagents (GE Health-
care, Piscataway, NJ). Because protein expression of
-actin in
placental samples was independent of BMI (Supplemental Fig. 1,
published on The Endocrine Society’s Journals Online web site
at http://jcem.endojournals.org), all blots were stripped and re-
probed for
-actin. Analysis of the blots was performed by den-
sitometry using
Imager (Alpha Innotech Corporation, Santa
Clara, CA). To account for variation in loading, the density of the
target band was divided by the corresponding
-actin band. For
each target, all values were expressed in relation to the highest tar-
get/
-actin ratio, which was arbitrarily assigned a value of 1.0.
Measurements of amino acid transporter activity
in MVM
The activity of the amino acid transporter systems A and L was
measured as previously described (8). In brief, MVM vesicles were
preloaded by incubation in 300 mMmannitol and 10 mMHEPES-
Tris (pH 7.4) overnight at 4 C. At time zero, 30
l of vesicles were
rapidly mixed (1:2) with the appropriate incubation buffer con-
taining
14
C-methyl-aminoisobutyric acid (MeAIB; 150
M)or
3
H-
L-leucine (0.375
M) at 37 C. Uptake of radiolabeled substrate was
terminated by the addition of 2 ml of ice-cold PBS after 30 sec
(MeAIB) or 8 sec (leucine) (8). Subsequently, vesicles were rapidly
separated from the substrate medium by filtration on mixed ester
filters (0.45-
m pore size; Millipore Corporation, Billerica, MA)
and washed with 3 2 ml of PBS. In studies of MeAIB transport,
150 mMNaCl and 150 mMKCl were used in incubation buffers to
assess total and sodium-independent uptake, respectively. In leucine
transport experiments, nonmediated flux was studied in the pres-
ence of 30 mMunlabeled L-leucine.
In all uptake experiments, each condition was studied in trip-
licate for each vesicle preparation. Filters were dissolved in 2 ml
of liquid scintillation fluid and counted, and uptakes were ex-
pressed as picomoles per milligram of protein. Na
-dependent
uptake of MeAIB (corresponding to system A activity) was cal-
culated by subtracting Na
-independent from total uptakes.
Mediated leucine uptake, which in isolated MVM almost en-
tirely represents system L activity (31), was calculated by sub-
tracting nonmediated transport from total uptake. Uptakes were
expressed as picomoles per minute per 30 sec (system A) or pi-
comoles per minute per 8 sec (system L).
Data presentation and statistics
Summary data are presented as means SEM. Variables were
analyzed as continuous across the range of BMI and birth
weights, and linear relationships between variables were deter-
mined using bivariate analysis and Pearson’s correlation coeffi-
cients. A P0.05 value (two-tailed) was considered significant.
J Clin Endocrinol Metab, January 2013, 98(1):105–113 jcem.endojournals.org 107
Results
Clinical data
Table 1 shows selected clinical data for the study sub-
jects, divided into Normal BMI (18.5–24.9 kg/m
2
) and
High BMI (25– 44.9 kg/m
2
) groups according to measure-
ments of body weight and length in early pregnancy. There
were no statistical differences between BMI groups with
regard to gestational weight gain or gestational age. There
was a trend toward higher birth weights and placental
weights in the High BMI group; however, these differences
failed to reach statistical significance. However, when an-
alyzed across the BMI range of all subjects, placental
weights (r 0.43; P0.05) and birth weights (r 0.46;
P0.05) were positively correlated to maternal BMI.
Activity of placental insulin/IGF-I signaling
Placental insulin/IGF-I signaling activity was assessed
by determining phosphorylation of IRS-1 at Tyr-612 and
Akt at Thr-308 (Fig. 1). IRS-1 phosphorylation was pos-
itively correlated to BMI (P0.05; Fig. 1B). In addition,
phosphorylation of both IRS-1 and Akt was positively
correlated to birth weight (P0.01; Fig. 1, C and D).
There was no significant correlation between BMI or birth
weight and total IRS-1 or Akt expression.
Activity of placental AMPK signaling
As shown in Fig. 2, placental AMPK activity, as deter-
mined by Thr-172 phosphorylation, was inversely corre-
lated to maternal BMI (P0.05) and birth weight (P
0.05). There was no significant correlation between BMI
or birth weight and total AMPK expression.
Activity of placental mTOR complex (mTORC)-1
signaling
mTOR is a ubiquitously expressed serine/threonine ki-
nase that exists as two complexes, mTORC1 and -2, with
distinct regulation and function (32). S6K1, RPS6, and
FIG. 1. Placental insulin/IGF-I signaling in relation to BMI and birth
weight. A, Representative Western blots for total and
phosphorylated IRS-1 (Tyr-612) and Akt (Thr-308) in homogenates
of placentas from pregnancies with varying maternal BMI and birth
weights. There was no significant correlation between BMI or birth
weight and total IRS-1 or Akt. B, Relationship between BMI and
phosphorylation of placental IRS-1. C and D, Relationship between
birth weight and phosphorylation of placental IRS-1 (C) or Akt (D).
n17; r Pearson’s correlation coefficient.
TABLE 1. Selected clinical data
Normal BMI
(18.5–24.9)
High BMI
(25.044.9)
n1112
BMI (kg/m
2
) 21.7 0.6 38.8 2.3
a
Gestational weight gain (kg) 11.3 1.1 13.1 1.4
Gestational age (wk) 40.6 0.4 40.4 0.5
Birth weight (g) 3423 67 3635 151
Placental weight (g) 599 45 679 28
Data are expressed as means SEM.
a
P0.0001 vs. Normal BMI, Student’s ttest.
FIG. 2. Placental AMPK signaling in relation to BMI and birth weight. A,
Representative Western blots for total and phosphorylated AMPK
(Thr-
172) in homogenates of placentas from pregnancies with varying BMI and
birth weights. There was no significant correlation between BMI or birth
weight and total AMPK
. B and C, Relationship between BMI (B) or birth
weight (C) and phosphorylation of placental AMPK
.n17; r
Pearson’s correlation coefficient.
108 Jansson et al. Maternal Obesity and Placental Function J Clin Endocrinol Metab, January 2013, 98(1):105–113
4E-BP1 are key downstream targets of mTORC1. Placen-
tal 4E-BP1 phosphorylation (both at Thr-37/46 and Thr-
70) was positively correlated to early pregnancy BMI (P
0.01; Fig. 3, B and C). Phosphorylation of S6K1 (Thr-389;
P0.01), RPS6 (Ser235/236; P0.05), and 4E-BP1 (Thr
37/46, P0.05; and Thr-70, P0.001) was positively
correlated to birth weight (Fig. 3, D–G). There was no
significant correlation between BMI or birth weight and
total S6K1, RPS6, or 4E-BP1 expression.
Activity of placental mTORC2 signaling
mTORC2 phosphorylates Akt (Ser-473), SGK1 (Ser-
422), and PKC
(Ser-657). Phosphorylation of placental
SGK1 was positively correlated to maternal early preg-
FIG. 3. Placental mTORC1 signaling in relation to BMI and birth weight. A, Representative Western blots for total and phosphorylated S6K1 (Thr-
389), 4E-BP1 (Thr-37/46 or Thr-70), and RPS6 (Ser-235/236) in homogenates of placentas from pregnancies with varying BMI and birth weights.
There was no significant correlation between BMI or birth weight and total S6K1, 4E-BP1, or RPS6. B and C, Relationship between BMI and
phosphorylation of placental 4EBP-1 (Thr-37/46) (A) or 4E-BP1 (Thr-70) (B). D–G, Relationship between birth weight and phosphorylation of
placental S6K1 (D), RPS6 (E), 4E-BP1 (Thr-37/46) (F), or 4EBP-1 (Thr-70) (G). n 17; r Pearson’s correlation coefficient.
J Clin Endocrinol Metab, January 2013, 98(1):105–113 jcem.endojournals.org 109
nancy BMI (P0.05; Fig. 4B). In addition, phosphory-
lation of all three mTORC2 targets was positively corre-
lated to birth weight (Fig. 4, C–E). The expression of total
SGK1 and PKC
was not influenced by maternal BMI or
birth weight.
MVM system A and L transport activity
The activity of system A and system L in MVM isolated
from women with normal BMI and appropriate fetal
growth were similar to what we have reported previously
(8). MVM system A activity in the High BMI group
(69.7 9.3 pmol/mg 30 sec) was not significantly dif-
ferent from the normal BMI group (57.5 9.4 pmol/mg
30 sec). Similarly, MVM system L activity was not altered
in the High BMI group (0.055 0.013 pmol/mg 8 sec)
compared with the Normal BMI group (0.069 0.013
pmol/mg 8 sec). However, MVM system A transport
activity (Fig. 5A), but not system L activity (data not
shown), showed a strong positive correlation to birth
weight (r 0.60; P0.01), but not to maternal BMI (data
not shown).
Protein expression of system A amino acid
transporter isoforms in MVM
MVM SNAT1 or SNAT4 protein expression was not
significantly correlated to birth weight or BMI (data not
shown). In contrast, SNAT2 expression was positively
correlated to maternal early pregnancy BMI (P0.05;
Fig. 5C) and birth weight (P0.01; Fig. 5D).
Discussion
To the best of our knowledge, this is the first report to
study placental signaling and amino acid transport in
women with high BMI without pregnancy complications
such as gestational diabetes. We demonstrate that the ac-
tivity of the insulin/IGF-I and mTOR signaling pathways,
system A amino acid transporter activity, and protein ex-
pression of the SNAT2 isoform are increased in placentas
of obese women giving birth to large babies. We propose
that up-regulation of specific placental amino acid trans-
porter isoforms may contribute to fetal overgrowth in
obese women. This effect may be mediated by activation
of insulin and mTOR signaling pathways, which are pos-
itive regulators of placental amino acid transporters.
The microvillous plasma membrane of the syncytiotro-
phoblast, which is bathed in maternal blood, expresses a
number of hormone receptors, including receptors for in-
sulin (33) and IGF-I (34), suggesting that trophoblast
function is regulated by maternal hormones. Maternal cir-
culating IGF-I concentrations are positively correlated to
fetal growth in normal pregnancy (35), and maternal se-
rum concentrations of IGF-I have consistently been shown
to be decreased in IUGR (36). These findings are in agree-
ment with reports of inhibition of placental insulin/IGF-I
signaling in IUGR (37, 38). In addition, fasting insulin is
increased in obese pregnant women (26, 27). These ob-
servations are consistent with the increased phosphoryla-
tion of IRS-1 and Akt that we found in the placenta of high
BMI women giving birth to large babies, indicating acti-
vation of insulin/IGF-I signaling.
mTOR signaling constitutes a master regulator of pro-
tein translation, thereby controlling cell growth and me-
tabolism in response to a large number of upstream reg-
ulators, including growth factors, nutrient, oxygen, and
energy levels (32). We found that both placental mTORC1
and mTORC2 signaling pathways were activated in as-
sociation to high BMI and increased fetal growth. AMPK
FIG. 4. Placental mTORC2 signaling in relation to BMI and birth
weight. A, Representative Western blots for phosphorylated Akt (Ser-
473), total and phosphorylated SGK1 (Ser-422), and PKC
(Ser-657) in
homogenates of placentas from pregnancies with varying BMI and
birth weights. There was no significant correlation between BMI or
birth weight and total SGK1 or PKC
. B, Relationship between BMI
and phosphorylation of placental SGK1. C–E, Relationship between
birth weight and phosphorylation of placental Akt (Ser-473) (C), SGK1
(D), or PKC
(E). n 17; r Pearson’s correlation coefficient.
110 Jansson et al. Maternal Obesity and Placental Function J Clin Endocrinol Metab, January 2013, 98(1):105–113
is the primary cellular energy sensor and is phosphorylated
at Thr-172 in response to increased AMP/ATP ratio as-
sociated with energy deprivation. In this study we dem-
onstrated that the activity of placental AMPK, which in-
hibits mTORC1, was decreased and that IGF-I/insulin
signaling, which stimulates mTORC1 and -2, was acti-
vated in association to increased BMI and fetal growth.
The observed changes in AMPK and insulin/IGF-I signal-
ing are therefore likely to contribute to mTOR activation
in placentas of obese women giving birth to large babies.
System A is a sodium-dependent transporter mediating
the uptake of nonessential neutral amino acids into the
cell. System A activity establishes the high intracellular
concentration of amino acids like glycine, which is used to
exchange for extracellular essential amino acids via system
L. Thus, system A activity is critical for placental transport
of both nonessential and essential amino acids. System L
is a sodium-independent exchanger mediating cellular up-
take of essential amino acids including leucine. We found
that system A activity is increased in MVM isolated from
large babies of obese women, which may contribute to
increased fetal amino acid availability and fetal growth.
We reported previously that MVM system A activity was
increased in pregnancies complicated by GDM or type 1
diabetes independently of fetal overgrowth. However,
MVM system A activity was unaffected in lean nondia-
betic women giving birth to LGA fe-
tuses (8). This is consistent with the
possibility that obesity and diabetes in
pregnancy have common underlying
metabolic disturbances that can result
in increased placental nutrient trans-
port and fetal growth.
It is well established that IGF-I, in-
sulin, and mTOR signaling stimulate
placental amino acid transport (17–19,
21, 22). The activation of insulin/IGF-I
and mTOR signaling that we observed
in placentas of obese women giving
birth to large babies is likely to contrib-
ute to the observed increase in system A
activity. However, system A amino acid
transporter activity is also regulated by
other signaling pathways, such as leptin,
that we have not directly addressed in the
present study. This is relevant because
leptin stimulates trophoblast system A
amino acid transport in vitro (19) and ma-
ternal leptin levels are elevated in pregnant
women with high BMI (27). All three
known isoforms of system A, SNAT1
(SLC38A1), SNAT2 (SLC38A2), and
SNAT4 (SLC38A4), are expressed in the
placenta (39). The effect of obesity on system A was isoform-
specific because protein expression of SNAT2, but not
SNAT1 and SNAT4, was up-regulated in MVM isolated
from placentas of large babies of obese women. Little is
known with respect to regulation of placental SNATs; how-
ever, SNAT4 appears to be gestationally regulated in the
human placenta (39), and SNAT1 and -2 expression has been
reported to be differentially regulated in response to amino
acid deprivation in BeWo cells (40). Furthermore, placental
IGF-II has been shown to specifically regulate SNAT4 gene
expression in mice (41).
Despite a small sample size, birth weight was positively
correlated to maternal BMI, in agreement with the liter-
ature (1–3). Some of the outcome variables in our study
(phosphorylation of IRS-1, 4EBP-1, AMPK and SGK1,
and SNAT2 protein expression) were significantly corre-
lated to both birth weight and maternal early pregnancy
BMI, whereas others (phosphorylation of Akt, S6K1,
RPS6, and PKC
, and system A activity) were significantly
correlated to birth weight only. This may reflect that fac-
tors unrelated to BMI regulate placental signaling and
amino acid transport and contribute to increased fetal
growth. Alternatively, these findings may be explained by
the small sample size, which is a limitation of our study.
Our results should be confirmed in larger studies, which
FIG. 5. MVM system A activity and SNAT 2 expression in relation to BMI and birth weight. A,
Relationship between birth weight and system A amino acid transport activity was measured
in vitro in MVM isolated from placentas of pregnancies with varying BMI and birth weights
(n 23). B, Representative Western blots for MVM expression of SNAT1, -2, and -4 isoforms
of the system A amino acid transporter. C and D, Relationship between BMI (C) or birth
weight (D) and MVM SNAT 2 expression. n 22; r Pearson’s correlation coefficient.
J Clin Endocrinol Metab, January 2013, 98(1):105–113 jcem.endojournals.org 111
will have the power to allow multiple regression modeling.
Furthermore, our sample included a wide range of BMIs,
and it cannot be excluded that a few subjects with a BMI
over 40 kg/m
2
may have skewed results in the obese group.
Future studies may therefore benefit from stratifying study
subjects by BMI category. Placental system A and L trans-
port activity in pregnancies complicated by diabetes and
fetal overgrowth has been reported to be different in a
Swedish (8) and a British population (9). This suggests that
there may be ethnic or population differences in the pla-
cental response to maternal metabolic disease. Thus, ex-
ploration of the impact of maternal obesity on placental
signaling and transport in populations other than the one
studied in the current report appears warranted. This in-
formation will increase our understanding of the mecha-
nisms linking maternal obesity to large size at birth and
may facilitate the development of novel intervention strat-
egies to alleviate fetal overgrowth.
Acknowledgments
In memory of Margareta Wennergren (1948–2011), to whom
we are profoundly indebted for her support and inspiration over
many years.
We thank the midwives at Lundby Prenatal Care Center for
helping us to recruit pregnant women to our study and Ellen
Samuelsson and staff at O
¨stra KKO
¨and Mo¨ lndals hospitals who
made it possible for us to collect the placentas.
Address all correspondence and requests for reprints to:
Theresa L. Powell, Ph.D., Center for Pregnancy and Newborn
Research, Department of Obstetrics and Gynecology, Univer-
sity of Texas Health Science Center San Antonio, Mail Code
7836, 7703 Floyd Curl Drive, San Antonio, Texas 78229-
3900. E-mail: powellt3@uthscsa.edu.
This work was supported by grants from the Swedish Re-
search Council (10838 to T.J. and 14555 to T.L.P.) and the
National Institutes of Health (HD68370 to T.J. and DK89989 to
T.L.P.).
Disclosure Summary: The authors declare that there are no
conflicts of interest in relation to this study.
References
1. Sebire NJ, Jolly M, Harris JP, Wadsworth J, Joffe M, Beard RW,
Regan L, Robinson S 2001 Maternal obesity and pregnancy out-
come: a study of 287,213 pregnancies in London. Int J Obes Relat
Metab Disord 25:1175–1182
2. Baeten JM, Bukusi EA, Lambe M 2001 Pregnancy complications
and outcomes among overweight and obese nulliparous women.
Am J Public Health 91:436–440
3. Ehrenberg HM, Mercer BM, Catalano PM 2004 The influence of
obesity and diabetes on the prevalence of macrosomia. Am J Obstet
Gynecol 191:964–968
4. Boney CM, Verma A, Tucker R, Vohr BR 2005 Metabolic syn-
drome in childhood: association with birth weight, maternal obesity,
and gestational diabetes. Pediatrics 115:e290–e296
5. Mahendran D, Donnai P, Glazier JD, D’Souza SW, Boyd RD, Sibley
CP 1993 Amino acid (system A) transporter activity in microvillous
membrane vesicles from the placentas of appropriate and small for
gestational age babies. Pediatr Res 34:661–665
6. Glazier JD, Cetin I, Perugino G, Ronzoni S, Grey AM, Mahendran
D, Marconi AM, Pardi G, Sibley CP 1997 Association between the
activity of the system A amino acid transporter in the microvillous
plasma membrane of the human placenta and severity of fetal com-
promise in intrauterine growth restriction. Pediatr Res 42:514 –519
7. Evers IM, de Valk HW, Mol BW, ter Braak EW, Visser GH 2002
Macrosomia despite good glycaemic control in type I diabetic preg-
nancy; results of a nationwide study in The Netherlands. Diabeto-
logia 45:1484–1489
8. Jansson T, Ekstrand Y, Bjo¨ rn C, Wennergren M, Powell TL 2002
Alterations in the activity of placental amino acid transporters in
pregnancies complicated by diabetes. Diabetes 51:2214–2219
9. Kuruvilla AG, D’Souza SW, Glazier JD, Mahendran D, Maresh MJ,
Sibley CP 1994 Altered activity of the system A amino acid trans-
porter in microvillous membrane vesicles from placentas of macro-
somic babies born to diabetic women. J Clin Invest 94:689–695
10. Jansson T, Wennergren M, Powell TL 1999 Placental glucose trans-
port and GLUT 1 expression in insulin dependent diabetes. Am J
Obstet Gynecol 180:163–168
11. Gaither K, Quraishi AN, Illsley NP 1999 Diabetes alters the expres-
sion and activity of the human placental GLUT1 glucose trans-
porter. J Clin Endocrinol Metab 84:695–701
12. Magnusson AL, Waterman IJ, Wennergren M, Jansson T, Powell
TL 2004 Triglyceride hydrolase activities and expression of fatty
acid binding proteins in human placenta in pregnancies complicated
by IUGR and diabetes. J Clin Endocrinol Metab 89:4607–4614
13. Scifres CM, Chen B, Nelson DM, Sadovsky Y 2011 Fatty acid bind-
ing protein 4 regulates intracellular lipid accumulation in human
trophoblasts. J Clin Endocrinol Metab 96:E1083–E1091
14. Gauster M, Hiden U, van Poppel M, Frank S, Wadsack C, Hau-
guel-de Mouzon S, Desoye G 2011 Dysregulation of placental en-
dothelial lipase in obese women with gestational diabetes mellitus.
Diabetes 60:2457–2464
15. Higgins L, Greenwood SL, Wareing M, Sibley CP, Mills TA 2011
Obesity and the placenta: a consideration of nutrient exchange
mechanisms in relation to aberrant fetal growth. Placenta 32:1–7
16. Dube´ E, Gravel A, Martin C, Desparois G, Moussa I, Ethier-Chi-
asson M, Forest JC, Gigue`re Y, Masse A, Lafond J 2012 Modulation
of fatty acid transport and metabolism by obesity in the human
full-term placenta. Biol Reprod 87:14, 1–11
17. Roos S, Kanai Y, Prasad PD, Powell TL, Jansson T 2009 Regulation
of placental amino acid transporter activity by mammalian target of
rapamycin. Am J Physiol Cell Physiol 296:C142–C150
18. Roos S, Jansson N, Palmberg I, Sa¨ ljo¨ K, Powell TL, Jansson T 2007
Mammalian target of rapamycin in the human placenta regulates
leucine transport and is down-regulated in restricted foetal growth.
J Physiol 582:449–459
19. Jansson N, Greenwood SL, Johansson BR, Powell TL, Jansson T
2003 Leptin stimulates the activity of the system A amino acid trans-
porter in human placental villous fragments. J Clin Endocrinol
Metab 88:1205–1211
20. Kniss DA, Shubert PJ, Zimmerman PD, Landon MB, Gabbe SG
1994 Insulin growth factors: their regulation of glucose and amino
acid transport in placental trophoblasts isolated from first-trimester
chorionic villi. J Reprod Med 39:249–256
21. Karl PI, Alpy KL, Fisher SE 1992 Amino acid transport by the cul-
tured human placental trophoblast: effect of insulin on AIB trans-
port. Am J Physiol 262:C834–C839
22. Karl PI 1995 Insulin-like growth factor-1 stimulates amino acid
uptake by the cultured human placental trophoblast. J Cell Physiol
165:83–88
112 Jansson et al. Maternal Obesity and Placental Function J Clin Endocrinol Metab, January 2013, 98(1):105–113
23. Jones HN, Powell TL, Jansson T 2007 Regulation of placental nu-
trient transport. A review. Placenta 28:763–774
24. Jansson T, Powell TL 2006 IFPA 2005 Award in Placentology
Lecture. Human placental transport in altered fetal growth: does
the placenta function as a nutrient sensor? A review. Placenta
27(Suppl A):S91–S97
25. Sferruzzi-Perri AN, Owens JA, Pringle KG, Robinson JS, Roberts
CT 2006 Maternal insulin-like growth factors I and II act via dif-
ferent pathways to promote fetal growth. Endocrinology 147:
3344–3355
26. Ramsay JE, Ferrell WR, Crawford L, Wallace AM, Greer IA, Sattar
N2002 Maternal obesity is associated with dysregulation of met-
abolic, vascular and inflammatory pathways. J Clin Endocrinol
Metab 87:4231–4237
27. Jansson N, Nilsfelt A, Gellerstedt M, Wennergren M, Rossander-
Hulthe´n L, Powell TL, Jansson T 2008 Maternal hormones linking
maternal body mass index and dietary intake to birth weight. Am J
Clin Nutr 87:1743–1749
28. Illsley NP, Wang ZQ, Gray A, Sellers MC, Jacobs MM 1990 Si-
multaneous preparation of paired, syncytial, microvillous and basal
membranes from human placenta. Biochim Biophys Acta 1029:
218–226
29. Ling R, Bridges CC, Sugawara M, Fujita T, Leibach FH, Prasad PD,
Ganapathy V 2001 Involvement of transporter recruitment as well
as gene expression in the substrate-induced adaptive regulation of
amino acid transport system A. Biochim Biophys Acta 1512:15–21
30. Rosario FJ, Jansson N, Kanai Y, Prasad PD, Powell TL, Jansson T
2011 Maternal protein restriction in the rat inhibits placental insu-
lin, mTOR, and STAT3 signaling and down-regulates placental
amino acid transporters. Endocrinology 152:1119–1129
31. Jansson T, Scholtbach V, Powell TL 1998 Placental transport of
leucine and lysine is reduced in intrauterine growth restriction. Pe-
diatr Res 44:532–537
32. Alessi DR, Pearce LR, García-Martínez JM 2009 New insights into
mTOR signaling: mTORC2 and beyond. Sci Signal 2:pe27
33. Desoye G, Hartmann M, Blaschitz A, Dohr G, Hahn T, Kohnen G,
Kaufmann P 1994 Insulin receptors in syncytiotrophoblast and fetal
endothelium of human placenta. Immunohistochemical evidence
for developmental changes in distribution pattern. Histochemistry
101:277–285
34. Fang J, Furesz TC, Lurent RS, Smith CH, Fant ME 1997 Spatial
polarization of insulin-like growth factor receptors on the human
syncytiotrophoblast. Pediatr Res 41:258–265
35. Chellakooty M, Vangsgaard K, Larsen T, Scheike T, Falck-Larsen
J, Legarth J, Andersson AM, Main KM, Skakkebaek NE, Juul A
2004 A longitudinal study of intrauterine growth and the placental
growth hormone (GH)-insulin-like growth factor I axis in maternal
circulation: association between placental GH and fetal growth.
J Clin Endocrinol Metab 89:384–391
36. Bhatia S, Faessen GH, Carland G, Balise RL, Gargosky SE, Druzin
M, El-Sayed Y, Wilson DM, Giudice LC 2002 A longitudinal anal-
ysis of maternal serum insulin-like growth factor I (IGF-I) and total
and nonphosphorylated IGF-binding protein-1 in human pregnan-
cies complicated by intrauterine growth restriction. J Clin Endocri-
nol Metab 87:1864–1870
37. Laviola L, Perrini S, Belsanti G, Natalicchio A, Montrone C, Leon-
ardini A, Vimercati A, Scioscia M, Selvaggi L, Giorgino R, Greco P,
Giorgino F 2005 Intrauterine growth restriction in humans is asso-
ciated with abnormalities in placental insulin-like growth factor sig-
naling. Endocrinology 146:1498–1505
38. Yung HW, Calabrese S, Hynx D, Hemmings BA, Cetin I, Charnock-
Jones DS, Burton GJ 2008 Evidence of translation inhibition and
endoplasmic reticulum stress in the etiology of human intrauterine
growth restriction. Am J Pathol 173:451–462
39. Desforges M, Lacey HA, Glazier JD, Greenwood SL, Mynett KJ,
Speake PF, Sibley CP 2006 The SNAT4 isoform of the system A
amino acid transporter is expressed in human placenta. Am J Physiol
Cell Physiol 290:C305–C312
40. Jones HN, Ashworth CJ, Page KR, McArdle HJ 2006 Expression
and adaptive regulation of amino acid transport system A in a pla-
cental cell line under amino acid restriction. Reproduction 131:951–
960
41. Constaˆ ncia M, Angiolini E, Sandovici I, Smith P, Smith R, Kelsey G,
Dean W, Ferguson-Smith A, Sibley CP, Reik W, Fowden A 2005
Adaptation of nutrient supply to fetal demand in the mouse involves
interaction between the Igf2 gene and placental transporter systems.
Proc Natl Acad Sci USA 102:19219–19224
J Clin Endocrinol Metab, January 2013, 98(1):105–113 jcem.endojournals.org 113
... Specifically, the activity of System A and L amino acid transporters has been reported to be decreased in human IUGR [9][10][11][12] . In addition, various studies have reported an upregulation of placental System A and L transport activity in fetal overgrowth [13][14][15] . In primate and rodent models of IUGR, down-regulation of placental System A and L transporter activity precedes fetal growth restriction [16][17][18] , consistent with the possibility that altered placental amino acid transport is mechanistically linked to abnormal fetal growth. ...
... Western blot analysis was carried out as described 14,18,32,43 . Briefly, 10-15 μg of placental homogenates or 5-10 μg of TPM were loaded onto a NuPAGE Bis-Tris Gels (Thermo Fisher Scientific), and electrophoresis was performed using MOPS-SDS running buffer at a constant 100 V for 1 h. ...
... Following the washing step in TBS plus 0.1% Tween 20, the bands were visualized using enhanced chemiluminescence detection reagents from Thermo Fisher Scientific (Waltham, MA, USA). Blots were stripped 14,18,43 and reprobed for β-actin. Image J software from the National Institutes of Health (Bethesda, MD) was used to perform densitometry analysis of the Immunoblots. ...
Preprint
Abnormal fetal growth is associated with perinatal complications and adult disease. The placental mechanistic target of rapamycin (mTOR) signaling activity is positively correlated to placental nutrient transport and fetal growth. However, if this association represents a mechanistic link, it remains unknown. We hypothesized that trophoblast-specific mTOR knockdown in late pregnant mice decreases placental nutrient transport and inhibits fetal growth. PiggyBac Transposase-Enhanced Pronuclear Injection was performed to generate transgenic mice containing a trophoblast-specific Cyp19I.1 promoter-driven, doxycycline-inducible luciferase reporter transgene with a Mtor shRNAmir sequence in its 3’ untranslated region (UTR). We induced mTOR knockdown by administration of doxycycline starting at E14.5. Dams were euthanized at E 17.5, and trophoblast-specific gene targeting was confirmed. Placental mTOR protein expression was reduced by ∼68% in these animals, which was associated with a marked inhibition of mTORC1 and mTORC2 signaling activity. Moreover, we observed a decreased expression of System A amino acid transporter isoform SNAT2 and the System L amino acid transporter isoform LAT1 in isolated trophoblast plasma membranes and lower fetal but not placental weight. Inhibition of trophoblast mTOR signaling in late pregnancy is mechanistically linked to decreased placental nutrient transport and reduced fetal growth. Modulating placental mTOR signaling may represent a novel intervention in pregnancies with abnormal fetal growth.
... Other studies have also provided evidence in support of a connection between metabolism and epigenetics. The dysregulation of adenosine monophosphate-activated protein kinase (AMPK) and the mammalian target of rapamycin (mTOR) homeostasis are linked to gestational obesity [81]. AMPK activation occurs in response to reduced energy levels suggested by high and low ATP concentrations. ...
Preprint
Full-text available
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by trans-generationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for pregnancy-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish the exposure threshold to the fetus due to nutrient deficiencies or other environmental factors that can lead to clinically relevant epigenetic changes that modulate disease risks. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development.
... All experimental protocols were approved by the Institutional Review Board of the University of Colorado Anschutz Medical Campus. We used a well-established mouse model of maternal obesity that shows extensive similarities to the human condition, including elevated levels of maternal leptin, glucose intolerance, activation of placental insulin and mTOR signaling, increased placental nutrient transport, and fetal overgrowth [26][27][28]. In brief, 12-week-old C57BL/6J female mice were fed either a control diet (D12489; Research Diets, New Brunswick, NJ, USA) or an obesogenic diet (Western Diet D12089B; Research Diets) consisting of pellets containing 10% and 40% calories from fat, respectively, as described previously [29]. ...
Article
Full-text available
Background Children born to obese mothers are at increased risk of developing mood disorders and cognitive impairment. Experimental studies have reported structural changes in the brain such as the gliovascular unit as well as activation of neuroinflammatory cells as a part of neuroinflammation processing in aged offspring of obese mothers. However, the molecular mechanisms linking maternal obesity to poor neurodevelopmental outcomes are not well established. The ephrin system plays a major role in a variety of cellular processes including cell–cell interaction, synaptic plasticity, and long-term potentiation. Therefore, in this study we determined the impact of maternal obesity in pregnancy on cortical, hippocampal development, vasculature and ephrin-A3/EphA4-signaling, in the adult offspring in mice. Methods Maternal obesity was induced in mice by a high fat/high sugar Western type of diet (HF/HS). We collected brain tissue (prefrontal cortex and hippocampus) from 6-month-old offspring of obese and lean (control) dams. Hippocampal volume, cortical thickness, myelination of white matter, density of astrocytes and microglia in relation to their activity were analyzed using 3-D stereological quantification. mRNA expression of ephrin-A3, EphA4 and synaptic markers were measured by qPCR in the brain tissue. Moreover, expression of gap junction protein connexin-43, lipocalin-2, and vascular CD31/Aquaporin 4 were determined in the hippocampus by immunohistochemistry. Results Volume of hippocampus and cortical thickness were significantly smaller, and myelination impaired, while mRNA levels of hippocampal EphA4 and post-synaptic density (PSD) 95 were significantly lower in the hippocampus in the offspring of obese dams as compared to offspring of controls. Further analysis of the hippocampal gliovascular unit indicated higher coverage of capillaries by astrocytic end-feet, expression of connexin-43 and lipocalin-2 in endothelial cells in the offspring of obese dams. In addition, offspring of obese dams demonstrated activation of microglia together with higher density of cells, while astrocyte cell density was lower. Conclusion Maternal obesity affects brain size, impairs myelination, disrupts the hippocampal gliovascular unit and decreases the mRNA expression of EphA4 and PSD-95 in the hippocampus of adult offspring. These results indicate that the vasculature–glia cross-talk may be an important mediator of altered synaptic plasticity, which could be a link between maternal obesity and neurodevelopmental/neuropsychiatric disorders in the offspring. Graphical Abstract
... For example, the capacity of the placenta to transport amino acids is positively associated with fetal growth in several pregnancy complications including obesity, diabetes, and intrauterine fetal growth restriction. [1][2][3] We have reported that mechanistic target of rapamycin (mTOR) is a positive regulator of placental functions, including transport of amino acids, 4 folate, 5 and mitochondrial respiration. 6 mTOR is a serine/threonine kinase that, in response to nutrients and growth factor signaling, regulates an array of cellular growth related processes such as protein translation, cytoskeleton reorganization, and cell migration. ...
Article
Full-text available
Normal fetal development is critically dependent on optimal nutrient supply by the placenta, and placental amino acid transport has been demonstrated to be positively associated with fetal growth. Mechanistic target of rapamycin (mTOR) is a positive regulator of placental amino acid transporters, such as System A. Oleic acid (OA) has been previously shown to have a stimulatory role on placental mTOR signaling and System A amino acid uptake in primary human trophoblast (PHT) cells. We investigated the mechanistic link between OA and System A activity in PHT. We found that inhibition of mTOR complex 1 or 2, using small interfering RNA to knock down raptor or rictor, prevented OA‐stimulated System A amino acid transport indicating the interaction of OA with mTOR. Phosphatidic acid (PA) is a key intermediary for phospholipid biosynthesis and a known regulator of the mTOR pathway; however, phospholipid biosynthetic pathways have not been extensively studied in placenta. We identified placental isoforms of acyl transferase enzymes involved in de novo phospholipid synthesis. Silencing of 1‐acylglycerol‐3‐phosphate‐O‐acyltransferase‐4, an enzyme in this pathway, prevented OA mediated stimulation of mTOR and System A amino acid transport. These data indicate that OA stimulates mTOR and amino acid transport in PHT cells mediated through de novo synthesis of PA. We speculate that fatty acids in the maternal circulation, such as OA, regulate placental functions critical for fetal growth by interaction with mTOR and that late pregnancy hyperlipidemia may be critical for increasing nutrient transfer to the fetus.
... In particular, placental inflammation has been observed in pregnancies complicated by obesity [13] and gestational diabetes mellitus (GDM) [14] and may play a central role in determining the fetal environment in these pregnancies. These changes in placental function caused by altered inflammatory profiles can lead to the co-morbidities observed in these pregnancies [15][16][17][18][19]. So, obesity in pregnancy and GDM have been linked to the disruption in several inflammatory mediators in the maternal and fetal compartments. ...
Article
Full-text available
Objective: In pregnancies complicated by maternal obesity and diabetes, a disruption in inflammatory mediators occurs, resulting in endothelial microvascular dysfunction, oxidative stress, tissue damage, and maternal and feto-neonatal complications. To outline this proinflammatory status, an innovative approach is represented by the measurement of proinflammatory cytokines. Among these biomarkers, B-cell-activating factor (BAFF) and platelet-activating factor (PAF) play a key role in metabolic regulation, immune response to infections, tissue homeostasis, and "food-related inflammation." The aim of the present study is to investigate the blood expression of BAFF and PAF in a cohort of pregnant women affected by obesity and diabetes compared with a control group of healthy pregnant women. Methods: A prospective longitudinal cohort study has been conducted on pregnant women referred to Fondazione Policlinico Universitario Gemelli IRCCS in Rome. For each pregnant woman, a capillary sample was collected with a swab in three different consecutive evaluations carried out in the three trimesters of pregnancy. Results: A total of 77 pregnant women have been enrolled. No significant differences in BAFF and PAF levels were longitudinally observed between groups. Focusing on the exposed group, in the third trimester of pregnancy, both PAF and BAFF levels were lower than the basal time. Among the selected group of patients who developed Gestational Diabetes, only PAF values were longitudinally lower when compared to other groups. The multivariate analysis showed that BAFF levels were positively correlated with thyroid-stimulating hormone levels. No macrosomia, no shoulder dystocia, no major perineal lacerations at birth, and no intrauterine growth restriction were observed in the whole population. Conclusions: This study supports the involvement of metabolic and proinflammatory biomarkers in the mechanisms related to pregnancy complications. Improving a good metabolic environment for obese and diabetic pregnant women could break the vicious cycle connecting inflammation, oxidative stress, and metabolic disorders.
Article
Abnormal fetal growth, i.e., intrauterine growth restriction (IUGR) or fetal growth restriction (FGR) and fetal overgrowth, is associated with increased perinatal morbidity and mortality and is strongly linked to the development of metabolic and cardiovascular disease in childhood and later in life. Emerging evidence suggests that changes in placental amino acid transport may contribute to abnormal fetal growth. This review is focused on amino acid transport in the human placenta, however, relevant animal models will be discussed to add mechanistic insights. At least 25 distinct amino acid transporters with different characteristics and substrate preferences have been identified in the human placenta. Of these, System A, transporting neutral nonessential amino acids, and System L, mediating the transport of essential amino acids, have been studied in some detail. Importantly, decreased placental Systems A and L transporter activity is strongly associated with IUGR and increased placental activity of these two amino acid transporters has been linked to fetal overgrowth in human pregnancy. An array of factors in the maternal circulation, including insulin, IGF‐1, and adiponectin, and placental signaling pathways such as mTOR, have been identified as key regulators of placental Systems A and L. Studies using trophoblast‐specific gene targeting in mice have provided compelling evidence that changes in placental Systems A and L are mechanistically linked to altered fetal growth. It is possible that targeting specific placental amino acid transporters or their upstream regulators represents a novel intervention to alleviate the short‐ and long‐term consequences of abnormal fetal growth in the future.
Article
Gestational diabetes mellitus (GDM) is a common disorder in the clinic, which may lead to severe detrimental outcomes both for mothers and infants. However, the underlying mechanisms for GDM are still not clear. In the present study, we performed label-free proteomics using placentas from GDM patients and normal controls. Vitronectin caused our attention among differentially expressed proteins due to its potential role in the pathological progression of GDM. Vitronectin was increased in the placentas of GDM patients, which was confirmed by Western blot analysis. Vitronectin represses insulin signal transduction in trophoblast cells, whereas the knockdown of vitronectin further potentiates insulin-evoked events. Neutralization of CD51/61 abolishes the repressed insulin signal transduction in vitronectin-treated trophoblast cells. Moreover, vitronectin activates JNK in a CD51/61-depedent manner. Inhibition of JNK rescues impaired insulin signal transduction induced by vitronectin. Overall, our data indicate that vitronectin binds CD51/61 in trophoblast cells to activate JNK, and thus induces insulin resistance. In this regard, increased expression of vitronectin is likely a risk factor for the pathological progression of GDM. Moreover, blockade of vitronectin production or its receptors (CD51/61) may have therapeutic potential for dealing with GDM.
Article
Objective Maternal obesity affects 39.7% of reproductive‐age women in the United States. Emerging research has suggested that in utero exposure to maternal obesity is associated with adverse neurodevelopmental outcomes, but knowledge of underlying mechanisms in human samples is lacking. Methods A matched case–control study was performed in women with singleton fetuses who were undergoing elective pregnancy termination at gestational ages 15 to 21 weeks. Maternal adiponectin levels from plasma were measured using ELISA kits. RNA was extracted from fetal brain tissue using RNeasy Mini Kit (QIAGEN). mRNA expression from ADIPOR1 , ADIPOR2 , MTOR , ATG5 , ATG7 , BECN1 , and MAP1LC3B was quantified through the ΔΔCt method and using GAPDH as a housekeeping gene. Results We have identified transcription patterns associated with inhibition of autophagy in male fetal brain tissue exposed to maternal obesity (↑ MTOR , ↓ATG5 , ↓ATG7 , and ↓MAP1LC3B ), with female fetuses demonstrating either no change in transcription or nonsignificant changes associated with increased autophagy. There was significant downregulation of the autophagy‐associated gene BECN1 in both male and female individuals who were exposed to obesity in utero. Conclusions We present novel evidence suggesting that in utero exposure to maternal obesity in humans may significantly affect neurodevelopment, especially in male fetuses, through alterations in normal autophagy molecular mechanisms and with adiponectin as a potential mediator.
Article
Full-text available
Gestational diabetes (GDM) changes the maternal metabolic and uterine environment, thus increasing the risk of short- and long-term adverse outcomes for both mother and child. Children of mothers who have GDM during their pregnancy are more likely to develop Type 2 Diabetes (T2D), early-onset cardiovascular disease and GDM when they themselves become pregnant, perpetuating a multigenerational increased risk of metabolic disease. The negative effect of GDM is exacerbated by maternal obesity, which induces a greater derangement of fetal adipogenesis and growth. Multiple factors, including genetic, epigenetic and metabolic, which interact with lifestyle factors and the environment, are likely to contribute to the development of GDM. Genetic factors are particularly important, with 30% of women with GDM having at least one parent with T2D. Fetal epigenetic modifications occur in response to maternal GDM, and may mediate both multi- and transgenerational risk. Changes to the maternal metabolome in GDM are primarily related to fatty acid oxidation, inflammation and insulin resistance. These might be effective early biomarkers allowing the identification of women at risk of GDM prior to the development of hyperglycaemia. The impact of the intra-uterine environment on the developing fetus, “developmental programming”, has a multisystem effect, but its influence on adipogenesis is particularly important as it will determine baseline insulin sensitivity, and the response to future metabolic challenges. Identifying the critical window of metabolic development and developing effective interventions are key to our ability to improve population metabolic health.
Article
Full-text available
Knowledge of the consequences of maternal obesity in human placental fatty acids (FA) transport and metabolism is limited. Animal studies suggest that placental uptake of maternal FA is altered by maternal overnutrition. We hypothesized that high maternal body mass index (BMI) affects human placental FA transport by modifying expression of key transporters. Full-term placentas were obtained by vaginal delivery from normal weight (BMI, 18.5-24.9 kg/m(2)) and obese (BMI > 30 kg/m(2)) women. Blood samples were collected from the mother at each trimester and from cord blood at delivery. mRNA and protein expression levels were evaluated with real-time RT-PCR and Western blotting. Lipoprotein lipase (LPL) activity was evaluated using enzyme fluorescence. In vitro linoleic acid transport was studied with isolated trophoblasts. Our results demonstrated that maternal obesity is associated with increased placental weight, decreased gestational age, decreased maternal high-density lipoprotein (HDL) levels during the first and third trimesters, increased maternal triglyceride levels during the second and third trimesters, and increased maternal T3 levels during all trimesters, and decreased maternal cholesterol (CHOL) and low-density lipoprotein (LDL) levels during the third trimester; and increased newborn CHOL, LDL, apolipoprotein B100, and T3 levels. Increases in placental CD36 mRNA and protein expression levels, decreased SLC27A4 and FABP1 mRNA and protein and FABP3 protein expression, and increased LPL activity and decreased villus cytotrophoblast linoleic acid transport were also observed. No changes were seen in expression of PPARA, PPARD, or PPARG mRNA and protein. Overall this study demonstrated that maternal obesity impacts placental FA uptake without affecting fetal growth. These changes, however, could modify the fetus metabolism and its predisposition to develop diseases later in life.
Article
Full-text available
This study addressed the hypothesis that placental endothelial lipase (EL) expression is affected by pregnancies complicated by obesity and gestational diabetes mellitus (GDM). EL expression in placental tissues from pregnancies complicated by obesity, GDM, or obesity combined with GDM (obese-GDM) was analyzed by quantitative RT-PCR. Moreover, primary placental cells were isolated and treated with insulin, glucose, leptin, or tumor necrosis factor (TNF)-α, and EL expression was measured. Inhibitors of nuclear factor (NF)-κB or mitogen-activated protein kinase (MAPK) signaling were used to detect potential pathways of EL regulation in primary placental endothelial cells (ECs). In placentas from obese-GDM pregnancies, EL expression was upregulated by 1.9-fold (P < 0.05) compared with lean pregnancies, whereas obesity or GDM alone had no significant effect. Analyses of metabolic parameters in maternal venous and umbilical venous plasma revealed significantly increased insulin and leptin as well as slightly increased glucose and TNF-α values in the obese and obese-GDM groups. Cell culture experiments identified TNF-α and leptin, but not glucose or insulin, as regulators of EL expression in ECs. Induction of EL expression by these mediators occurred in a para/endocrine manner, since only leptin and TNF-α receptors, but not the cytokines themselves, were expressed in ECs. Inhibitor experiments suggested that TNF-α and leptin-mediated upregulation of EL may occur via two different routes. Whereas TNF-α induced EL upregulation in ECs by activation of the NF-κB pathway, leptin did not stimulate NF-κB or MAPK signaling pathways in these cells. Metabolic inflammation with high leptin and locally increased TNF-α concentrations at the fetal-placental interface regulates placental EL expression.
Article
OBJECTIVE: To examine the maternal and foetal risks of adverse pregnancy outcome in relation to maternal obesity, expressed as body mass index (BMI, kg=m 2) in a large unselected geographical population. DESIGN: Retrospective analysis of data from a validated maternity database system which includes all but one of the maternity units in the North West Thames Region. A comparison of pregnancy outcomes was made on the basis of maternal BMI at booking. SUBJECTS: A total of 287 213 completed singleton pregnancies were studied including 176 923 (61.6%) normal weight (BMI 20 – 24.9), 79 014 (27.5%) moderately obese (BMI 25 – 29.9) and 31 276 (10.9%) very obese (BMI ! 30) women. MEASUREMENTS: Antenatal complications, intervention in labour, maternal morbidity and neonatal outcome were examined and data presented as raw frequencies and adjusted odds ratios with 99% confidence intervals following logistic regression analysis to account for confounding variables. RESULTS: Compared to women with normal BMI, the following outcomes were significantly more common in obese pregnant women (odds ratio (99% confidence interval) for BMI 25 – 30 and BMI ! 30 respectively): gestational diabetes mellitus (.71)). However, delivery before 32 weeks' gestation (0.73 (0.65 – 0.82), 0.81 (0.69 – 0.95)) and breastfeeding at discharge (0.86 (0.84 – 0.88), 0.58 (0.56 – 0.60)) were significantly less likely in the overweight groups. In all cases, increasing maternal BMI was associated with increased magnitude of risk. CONCLUSION: Maternal obesity carries significant risks for the mother and foetus. The risk increases with the degree of obesity and persists after accounting for other confounding demographic factors. The basis of many of the complications is likely to be related to the altered metabolic state associated with morbid obesity.
Article
Although a number of causes of poor fetal growth are known, the involvement of placental transport proteins in the etiology of growth retardation is not understood. The aim of this study was to investigate the activity of the system A amino acid transporter and the Na+/H+ exchanger in vesicles isolated from the microvillous membrane of the syncytiotrophoblast of placentas of appropriate and small for gestational age babies. There were no biochemical differences between the membranes from the two groups of placentas, and there nas no difference in the activity of the Na+/H+ exchanger. The initial rate of uptake of methylaminoisobutyric acid (a nonmetabolizable amino acid analogue) was 63% lower in vesicles from placentas of small for gestational age babies. Kinetic analysis of the system A transporter (utilized by methylaminoisobutyric acid) showed that the V-max in the vesicles from placentas of small for gestational age babies (0.24 +/- 0.03 nmol/mg protein)30 s, n = 5) wits significantly lower than that in vesicles from placentas of appropriate for gestational age babies (0.64 +/- 0.09 nmol)mg protein)30 s, n = 4, p < 0.001), whereas the Km was not different between the two groups. It is concluded that there is an abnormality of system A amino acid transporter function in placentas of small for gestational age babies.
Article
The localisation of insulin receptors (IR) was investigated on cryosections of human non-pathologic first trimester and full term placentae by indirect immunohistochemistry with three different monoclonal antibodies (MABS). In placentae from 6 to 10 weeks post-menstruation (p-m.), only syncytiotrophoblast was stained, predominantly that of mesenchymal villi and syncytial sprouts, which are areas of high proliferative activity. In placentae from 11 to 14 weeks p-m., endothelial cells commenced to react with the IR MABS and the syncytiotrophoblast was less intensely labelled than at weeks 6 to 10 p-m. In term placentae, the microvillous membrane of the syncytiotrophoblast showed only patches of weak immunoreactivity. In contrast, the endothelial cells in the placenta but not in the umbilical cord were strongly stained. The amniotic epithelium in the chorionic plate and fibroblasts in the stroma were conspicuously labelled. The data indicate: (1) the receptor density on villous syncytiotrophoblast decreases and that of fetal endothelium increases throughout gestation; (2) syncytiotrophoblast of human term placentae expresses a low level per unit area of surface IR; and (3) the majority of IR in human term placentae is located in fetal endothelium. Apart from yet unknown functional effects of maternal and fetal insulin at the placental barrier, the results suggest a growth promoting effect on the trophoblast of maternal insulin in first trimester as well as developmental effects of fetal insulin on the feto-placental vessels at term.
Article
We investigated the molecular mechanism involved in the adaptive regulation of the amino acid transport system A, a process in which amino acid starvation induces the transport activity. These studies were done with rat C6 glioma cells. System A activity in these cells is mediated exclusively by the system A subtype, amino acid transporter A2 (ATA2). The other two known system A subtypes, ATA1 and ATA3, are not expressed in these cells. Exposure of these cells to an amino acid-free medium induces system A activity. This process consists of an acute phase and a chronic phase. Laser-scanning confocal microscopic immunolocalization of ATA2 reveals that the acute phase is associated with recruitment of preformed ATA2 from an intracellular pool to the plasma membrane. In contrast, the chronic phase is associated with an induction of ata2 gene expression as evidenced from the increase in the steady-state levels of ATA2 mRNA, restoration of the intracellular pool of ATA2 protein, and blockade of the induction by cycloheximide and actinomycin D. The increase in system A activity induced by amino acid starvation is blocked specifically by system A substrates, including the non-metabolizable α-(methylamino)isobutyric acid.