You are on page 1of 15

Biology of Reproduction, 2018, 98(6), 795–809

doi:10.1093/biolre/ioy010
Research Article
Advance Access Publication Date: 18 January 2018

Research Article

Diet-induced obesity alters the maternal

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


metabolome and early placenta transcriptome
and decreases placenta vascularity in
the mouse†
Tami J. Stuart1 , Kathleen O’Neill2,‡ , David Condon3 , Issac Sasson2 ,
Payel Sen4 , Yunwei Xia5 and Rebecca A. Simmons1,∗
1
Department of Pediatrics, Division of Neonatology, Children’s Hospital of Philadelphia, University of Pennsylvania,
Philadelphia, Pennsylvania, USA; 2 Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia,
Pennsylvania, USA; 3 Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia,
Pennsylvania, USA; 4 Epigenetics Center, Department of Cell and Developmental Biology, University of Pennsylvania,
Philadelphia, Pennsylvania, USA and 5 College of Arts and Sciences, Cornell University, Ithaca, New York, USA

Correspondence: Center for Research on Reproduction and Women’s Health, Biomedical Research Building II/III 1308,
Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA. Children’s
Hospital of Philadelphia, Philadelphia, PA, USA. Email: rsimmons@pennmedicine.upenn.edu

Kathleen O’Neill is co-first author.

Grant Support: Funding for this research was provided by K12 HD000849-28 (KEO RSDP), T32 HD040135 (KEO),
T32HD060556 (TS), R01DK062965 (RAS).
Conference Presentation: Presented in part at the 2016 American Society for Reproductive Medicine, Salt Lake City, Utah,
the 2017 Pediatric Academic Societies Meeting, San Francisco, California, and the 2017 Society for Reproductive
Investigation Meeting, Orlando, Florida.
Edited by Dr. Sarah Kimmins, PhD, McGill University

Received 28 August 2017; Revised 19 December 2017; Accepted 17 January 2018

Abstract
Maternal obesity is associated with an increased risk of obesity and metabolic disease in offspring.
Increasing evidence suggests that the placenta plays an active role in fetal programming. In this
study, we used a mouse model of diet-induced obesity to demonstrate that the abnormal metabolic
milieu of maternal obesity sets the stage very early in pregnancy by altering the transcriptome of
placenta progenitor cells in the preimplantation (trophectoderm [TE]) and early postimplantation
(ectoplacental cone [EPC]) placenta precursors, which is associated with later changes in placenta
development and function. Sphingolipid metabolism was markedly altered in the plasma of obese
dams very early in pregnancy as was expression of genes related to sphingolipid processing in the
early placenta. Upregulation of these pathways inhibits angiogenesis and causes endothelial dys-
function. The expression of many other genes related to angiogenesis and vascular development
were disrupted in the TE and EPC. Other key changes in the maternal metabolome in obese dams
that are likely to influence placenta and fetal development include a marked decrease in myo and
chiro-inositol. These early metabolic and gene expression changes may contribute to phenotypic
changes in the placenta, as we found that exposure to a high-fat diet decreased placenta microves-
sel density at both mid and late gestation. This is the first study to demonstrate that maternal
obesity alters the transcriptome at the earliest stages of murine placenta development.

C The Author(s) 2018. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. 795
For permissions, please e-mail: journals.permissions@oup.com
796 T. J. Stuart, 2018, Vol. 98, No. 6

Summary Sentence
Obesity in a mouse model leads to alterations in the maternal metabolome and early placenta
transcriptome as well as changes in vascularity later in gestation which may provide a mechanism
for decreased fetal growth.
Key words: angiogenesis, developmental origins of health and disease, gene expression, metabolism, placenta,
trophectoderm.

Introduction derstand the way in which the condition alters normal metabolic and

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


physiologic processes resulting in adverse outcomes [14]. Integration
The rate of obesity has more than doubled in the past 35 years, with
of “-omic” data may identify potential causal pathways underlying
one-third of adults in the Unites States now classified as obese [1].
abnormal placenta function and later development of obesity and
Not surprisingly, this overall increase in the prevalence of obesity
metabolic disorders in the offspring. Therefore, we hypothesized that
has been accompanied by an equivalent rise in the number of obese
obesity prior to pregnancy and during very early pregnancy alters the
pregnant women. While obese women are more likely to give birth
maternal metabolome which in turn adversely impacts placenta de-
to large for gestational age babies [2], they may also give birth to
velopment as evidenced by changes in the transcriptome, and leads
infants that are small for gestational age (SGA) [3], and recent stud-
to downstream effects on placenta development.
ies show that SGA infants born to obese women face higher rates
of neonatal morbidity than appropriately grown infants [4]. More-
over, higher maternal weight before pregnancy is associated with the
development of obesity, insulin resistance, and dyslipidemia in the
Materials and Methods
offspring [5]. Animals and Diet
The mechanisms underlying the development of these diseases All experimental procedures involving the use of live animals were
later in life remain to be fully elucidated. However, it is becoming approved by the Institutional Animal Care and Use Committee re-
more evident that the placenta is playing an active role in the pro- view boards of the Children’s Hospital of Philadelphia and the Uni-
gramming of offspring of obese mothers. Survival and growth of the versity of Pennsylvania. Female C57BL/6J mice (JAX stock #000664)
fetus are critically dependent on the placenta, which acts as the in- at 4 weeks were housed (five animals per cage) and given free ac-
terface between the maternal and fetal circulation to maintain fetal cess to feed and water. Animals were randomly selected to be fed
homeostasis and supply nutrients for optimal fetal growth. Impor- one of two diets: either standard control diet (Lab Diet 5015) or
tantly, nutrient transport and hormonal secretion can be altered by a high-fat diet (HFD; Harlan TD.06414). The control diet is 4.7
the placenta in response to alterations in the maternal environment kcal/gm, with calories provided by 19.8% protein, 54.1% carbo-
such as obesity, hypoxia, and inflammation [6]. hydrates, and 26.1% fat (∼37% saturated, 40% monounsaturated,
The window of time during which a fetus is exposed to an obese and 23% polyunsaturated). The HFD is 5.1 kcal/gm, with calo-
milieu is wide, from conception through birth. Therefore, attempts to ries provided by 18.4% protein, 21.3% carbohydrates, and 60.3%
elucidate the impact of maternal obesity at different developmental fat (37% saturated, 47% monounsaturated, and 16% polyunsat-
stages are paramount to understanding this complex process. While urated). Female animals were permitted to feed ad libitum for at
the mouse model of diet-induced obesity using a high-fat diet does least 12 weeks prior to mating and were continued on control or
not directly mimic a Western diet, it is the most common animal HFD throughout gestation. Females were mated with control male
model used to mimic morbid obesity in humans and results in very C57BL/6J mice, and mating was confirmed via presence of a vaginal
similar features of human obesity, i.e. insulin and leptin resistance plug (e0.5).
and hyperlipidemia [7–10]. Using this mouse model of diet-induced
obesity, we previously showed that marked maternal obesity is asso-
ciated with fetal growth restriction and abnormal placentation and Transcriptome analysis of trophectoderm (e4.5)
ultimately leads to the development of obesity and impaired glucose Four days after mating, blastocysts were harvested. The uterine horn
tolerance in the offspring, similar to what has been observed in hu- of the pregnant mouse was removed from the peritoneal cavity
man pregnancies complicated by morbid obesity [11]. Further, in and placed in cold calcium and magnesium-free phosphate buffered
embryo transfer studies, we found that exposure to maternal obesity saline (PBS). A 30 G needle was attached to a 1 mL syringe filled
prior to implantation was sufficient to result in placental dysfunction with PBS, and the needle was inserted into the end of the uter-
and fetal growth restriction, suggesting that the early embryonic pe- ine horn. Blastocysts were flushed from the uterine horn into a 60
riod represents a critical window of susceptibility [11]. At embryonic mm dish filled with cold PBS. Under a dissecting microscope, mi-
day 6.5 (e6.5), implantation is fully established and the ectoplacental croneedles were used to dissect the trophectoderm (TE) mechani-
cone (EPC) is formed, but its cells are still pluripotent. Therefore, cally from the abembryonic pole. Trophectoderm from five blasto-
the window between implantation at e4.5 and formation of the EPC cysts were pooled and preserved in RNAlater Stabilization Solution
at e6.5 provides an ideal time to investigate early alterations of the (Ambion) to produce a single sample. Trophectoderm cDNA from
placenta that are direct consequences of maternal exposures. four replicates of either control or HFD-derived blastocysts was uti-
The mechanisms underlying abnormal placenta function and de- lized for gene expression profiling. Total RNA was isolated from TE
velopment in maternal obesity have been extensively studied [6, 11, (PicoPure RNA Isolation Kit, Arcturus KIT0202). Complementary
12], yet the precise mechanisms are not fully understood. Maternal DNA was generated and linearly amplified (Ovation PICO WTA
metabolic function is altered in the obese state [13], which is likely to System, Nugen). A subsequent round of linear amplification was
have a major impact on the developing conceptus. Complex diseases, performed (TransPlex WTA Kit, Rubicon) to generate an adequate
such as obesity, require the use of integrated approaches to fully un- sample for microarray hybridization. Samples were hybridized to
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 797

the GeneChip Mouse Exon 1.0ST array (Affymetrix 901168) and clipping (∼0.3 mL/mouse). The blood was collected in EDTA tubes,
scanned (GeneChip Scanner 3000 7G System, Affymetrix). Probe and samples were centrifuged for 10 min at 1500× g. Plasma from
intensity data were normalized and summarized to transcript cluster 10 animals on each diet (n = 20) was isolated and stored at –80◦ C
ID level using RMA as implemented in the Partek Genomics Suite. until pregnancy was confirmed by delivery of pups. Samples were
Absence of embryonic tissue in samples was confirmed by lack of sent to Metabolon Inc. (Durham, NC, USA) for analysis and were
expression of octamer-binding transcription factor 4 (Oct4) using extracted and prepared using Metabolon’s standard solvent extrac-
quantitative PCR. Pairwise comparisons were made between groups tion method. The extracted samples were split into equal parts for
and false discovery rates (FDR) were calculated along with adjusted analysis on complementary GC/MS (gas chromatography mass spec-
P values by the Benjamini and Hochberg step-up method. Ingenuity trometry) and LC/MS (liquid chromatography mass spectrometry)
Pathway Analysis (IPA) software was used to evaluate functional platforms. Following normalization to volume extracted (plasma),

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


pathways. log transformation and imputation of missing values, if any, with
the minimum observed value for each compound, Welch two-sample
t-test and ANOVA contrasts were used to identify biochemicals that
Transcriptome analysis of EPC (e6.5)
differed significantly between experimental groups. Analysis by two-
Postimplantation embryos were isolated at embryonic day 6.5 (e6.5).
way ANOVA identified biochemicals exhibiting significant interac-
The uterine horn of the pregnant mouse was removed from the peri-
tion and main effects for experimental parameters of diet. An esti-
toneal cavity and placed in cold calcium and magnesium-free PBS.
mate of the FDR (q-value) was also calculated to take into account
The decidua was freed from the muscular uterine lining, and the
for multiple comparisons.
embryo was shelled out from the decidua using forceps under a dis-
secting microscope. Reichert’s membrane was removed [15], and
embryos were divided at the embryonic/extraembryonic junction
Immunohistochemistry
(circumferential groove) [16]. The embryonic portion, or epiblast,
Pregnant female mice were sacrificed at e12.5 or e18.5 using CO2
was placed in 50 μl extraction buffer (Arcturus PicoPure RNA Iso-
and cervical dislocation. The gravid uterus was dissected free and
lation Kit), incubated at 42◦ F for 30 min, and then snap frozen and
placed in cold PBS on ice. Individual implantation sites were sep-
stored at –140◦ F. Similarly, the extraembryonic portion which in-
arated, and placentas were dissected from the fetus, divided in
cludes the EPC and extra-embryonic ectoderm [17] was separately
half, placed in 10% formalin, and sent to the Children’s Hospi-
processed in the same manner. Total RNA was isolated from tissues
tal of Philadelphia Pathology core for staining with plasmalemma
using the Arcturus PicoPure RNA Isolation Kit (Thermo Fisher), in-
vesicle-associated protein (PLVAP) antibody. PLVAP antibody is a
cluding an on-column DNase digestion (Qiagen). RNA quality was
mouse-specific antibody that stains endothelial cells in embryonic
assessed using the Agilent Bioanalyzer RNA 6000 Pico Kit (Agilent)
and adult tissues and can therefore be used to quantify vascular-
ensuring all RNA Integrity Numbers were >8. Real-time quanti-
ity via the endothelial cells lining the vascular beds. Placentas were
tative reverse-transcription polymerase chain reaction (qPCR) was
embedded in paraffin, sectioned, and stained with PLVAP antibody
used on the RNA extracted from the corresponding epiblast to de-
(Biorad MCA2539T, RRID AB 1102821, Supplemental Table 1).
termine the sex of the embryo using TaqMan probes for inactive X
Staining was performed on a Bond Max automated staining system
specific transcripts (Xist) as a positive control (Mm01232884 m1)
(Leica Biosystems). The Bond Intense R staining kit (Leica Biosys-
and eukaryotic translation initiation factor 2, subunit 3, structural
tems DS9263) was used. Standard protocol was followed with the
gene Y-linked (Eif2s3y) (Mm01210630 m1) as a marker of male sex
exception of the primary antibody incubation, which was extended
(Applied Biosystems). Ribosomal and mitochondrial RNA were re-
to 1 h at room temperature. Avidin Biotin Blocking (Vector Labs
moved from total RNA with the RiboGone—Mammalian kit (Clon-
SP-2001) and a Peptide Blocking step were added (DAKO X0909).
tech). RNA-Seq libraries were prepared on individual sexed EPC (n
PLVAP antibody was used at a 1:100 dilution, and antigen retrieval
= 5 for female embryos and n = 4 for male embryos from each
was performed with E1 (Leica Biosystems) retrieval solution for 20
diet group) using the SMARTer Stranded RNA-Seq kit (Clontech).
min. Biotinylated Anti-Rat Secondary (Vector BA-4001, Supplemen-
Twenty-five million 75 base pair (bp) paired-end reads were ob-
tal Table 1) was used at a 1:200 dilution. Slides were rinsed, dehy-
tained for each biologic replicate on the NextSeq500 platform (Illu-
drated through a series of ascending concentrations of ethanol and
mina), aligned to the mm10 genome using STAR [18]. Genes were
xylene, then coverslipped. Slides were scanned on a Scanscope CS-O
counted using featureCounts [19] with Gencode M9, and differ-
slide scanner at 20× magnification and viewed with Aperio Im-
ential expression was done using EdgeR and corrected for multi-
ageScope software (Leica Biosystems). The Microvessel Analysis V1
ple testing using the Bonferroni correction [20]. qPCR was used to
algorithm (Leica Biosystems) was used to quantify vascularity. This
replicate RNA-Seq findings on a separate cohort of EPCs derived
algorithm detects and quantifies microvessels on slides stained with
from control and obese mice (n = 10 EPCs in each group, total 20
endothelial markers, such as PLVAP antibody. Microvessel density
mice). Select differentially expressed genes involved in early vascular
was calculated by the algorithm as the number of vessels per unit
development (core 1 synthase, glycoprotein-N-acetylgalactosamine
of analysis area in square microns. The analysis area included the
3-beta-galactosyltransferase, 1 (C1galt1), endothelial PAS domain
entire placenta. Placentas were sexed using genomic DNA isolated
protein 1 (Epas), platelet derived growth factor, alpha (Pdgfa), and
from the tails of corresponding fetuses via the QIAamp DNA Mini
transforming growth factor, beta receptor II (Tgfbr2)) identified in
kit (Qiagen), and qPCR was performed with Taqman probes for
RNA-Seq studies were examined with qPCR using TaqMan probes
Xist (Mm01232884 m1) and sex determining region of Chr Y (Sry)
(Mm00473987, Mm01236112, Mm01205760, Mm03024091).
(Mm00441712 s1). Results from the quantification of vascularity
were analyzed using two-way ANOVA (PRISM software version
Metabolomic analysis in maternal plasma 7.0a; GraphPad), and significance was defined as P value ≤ 0.05. At
Six days after detection of a vaginal plug (e6.5) female mice were e12.5, one slide each from seven control males, five control females,
briefly anesthetized with isoflurane, and blood was collected via tail seven HFD males, and seven HFD females were analyzed. At e18.5,
798 T. J. Stuart, 2018, Vol. 98, No. 6

Table 1. Weights of control and HFD dams, placentas, and fetuses. Data are presented as mean ± SEM.

Control Diet High-Fat Diet P-value

Dam weight after 12 weeks on diet 22.41 g ± 0.349 (n = 40) 29.58 g ± 0.696 (n = 38) <0.0001
E18.5 fetal weight 1.163 g ± 0.028 1.045 g ± 0.017 0.0003
E18.5 female fetus weight 1.178 g ± 0.033 (n = 22) 1.040 g ± 0.019 (n = 22) 0.0007
E18.5 male fetus weight 1.182 g ± 0.044 (n = 16) 1.051 g ± 0.028 (n = 21) 0.0127
E18.5 placenta weight 0.089 g ± 0.002 0.083 g ± 0.002 0.067
E18.5 female placenta weight 0.087 g ± 0.003 (n = 22) 0.082 g ± 0.004 (n = 22) 0.2947
E18.5 male placenta weight 0.095 g ± 0.004 (n = 16) 0.088 g ± 0.002 (n = 21) 0.0998

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


one slide each from five control males, six control females, three disrupted at the earliest stage of placenta development, gene ex-
HFD males, and five HFD females were analyzed. pression profiling was performed on TE of e4.5 blastocysts from
control and HFD dams. Using an FDR of <0.05, 5285 genes were
differentially expressed between HFD and controls (1 gene down-
Uterine and umbilical artery doppler imaging
regulated and 5284 genes upregulated) (Supplemental Table 2). IPA
All doppler imaging was performed by the Small Animal Imag-
revealed disruption of over 400 canonical and biologic pathways
ing Facility at the University of Pennsylvania using the Vevo
including endothelin-1 (ET-1) signaling, leptin signaling in obesity,
2100 Imaging System (FUJIFILM Visual Sonics) as described by
pigment epithelium-derived factor (PEDF) signaling, corticotropin-
Hernandez-Andrade et al. [21]. Mice were anesthetized with 2%
releasing hormone (CRH) signaling, eNOS signaling, protein kinase
isoflurane, and abdominal hair was removed using a chemical hair
A signaling, p38 mitogen-activated protein kinase (p38 MAPK) sig-
remover (Nair). Mice were positioned on a heated pad and body tem-
naling, fatty acid α-oxidation, and IL-12 signaling and production
perature and respiratory rate were continuously monitored. Umbili-
in macrophages (Supplemental Table 3).
cal artery flow was measured at the level of umbilical cord insertion
The ET-1 signaling pathway was upregulated in HFD TE. A to-
at the abdomen of the fetus. Three measurements of the peak systolic
tal of 77 of 181 genes in this pathway were differentially expressed
velocity (PSV) and end diastolic velocity (EDV) were taken per cord,
(P = 2.76E-05). Genes such as endothelin 1 (Edn1), fibroblast
and the mean of each was used to calculate the resistive index (RI)
growth factor receptor 3 (Fgfr3), and mitogen-activated protein ki-
using the standard calculation of (PSV-EDV)/PSV. All images were
nase 3 (Mapk3) were all significantly upregulated compared to con-
obtained at <60o . At e12.5–13.5, umbilical cord measurements were
trols. ET-1 is a potent vasoconstrictor, and elevated levels in the
taken on 12 HFD fetuses and 16 control fetuses (from three litters
placenta are thought to play a role in the development of preeclamp-
per diet), and bilateral uterine artery measurements were taken on
sia that is associated with maternal obesity [24].
two control dams and two HFD dams. At e18.5, umbilical cord
A total of 42 of 109 genes in the p38 MAPK signaling pathway
measurements were taken on six HFD fetuses and four control fe-
were differentially expressed leading to predicted activation by an
tuses (from three litters per diet), and uterine artery measurements
HFD (P = 1.33E-02). This pathway is triggered by stress stimuli
were taken on two control dams and three HFD dams. Results were
such as death ligands, inflammatory cytokines, and oxidative stress
compared using an unpaired t-test (PRISM software version 7.0a;
and leads to transcription of genes responsible for apoptosis and
GraphPad).
production of proinflammatory cytokines, such as IL-6 [25].
Importantly, we found that the pathway for leptin signaling in
obesity was upregulated in TE of mice fed an HFD (34/84 genes al-
Results
tered, P = 1.08E-02). Genes such as thymoma viral proto-oncogene
Maternal, fetal, and placenta characteristics 1 (Akt1), growth factor receptor bound protein 2 (Grb2), and
At the time of mating, mice fed an HFD were significantly heav- phosphoinositide-3-kinase regulatory subunit 5 (Pik3r5) were all
ier, had elevated 2 h glucose tolerance tests (GTT), and increased significantly upregulated.
fasting leptin, as we have previously described [11]. Overall, there
were no differences in resorption rates at e6.5 (2.5% control vs
Maternal obesity alters upstream regulators and
4.5% HFD), litter size at delivery (6.3 control pups vs 7.6 HFD
pups), or the male offspring to female offspring ratio between the
regulatory networks
two groups. Similar to previous studies [11, 22], male and fe- IPA can identify the cascade of upstream transcriptional regu-
male HFD e18.5 fetuses were significantly growth restricted; how- lators that may explain changes in gene expression and illumi-
ever, placentas did not differ in weight (Table 1). We did not as- nate the biological activities occurring in the tissue or cell of in-
sess fertility or fetal malformations in this study; however, previ- terest. We identified differential expression of multiple upstream
ous studies demonstrate that female mice fed a 60% HFD have transcriptional regulators including catenin (cadherin associated pro-
decreased fertility and higher rates of fetal developmental defects tein), beta 1 (CTNNB1) (Wnt signaling), p38 MAPK, eph receptor
[8, 22, 23]. B2 (ERK) 1/2, IL4, toll-like receptor 2 (TLR 2), and CREB binding
protein (CREBBP), which were all activated. Important upstream
regulators that were inhibited included RE1-silencing transcription
Maternal obesity alters transcriptomic pathways factor (REST), protein kinase, AMP-activated, alpha 2 catalytic sub-
related to inflammation and vascular development in unit (PRKAA2), and additional sex combs like 1 (ASXL1). REST re-
the TE presses the expression of the cholinergic gene in non-neuronal cells.
To elucidate the underlying mechanisms linking maternal obesity Interestingly, the placenta contains large amounts of acetylcholine
to altered offspring development and identify pathways that were and choline acetyltransferase, which are thought to regulate blood
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 799

flow in the placenta [26, 27]. However, its biological functions in factor (GEF) 6 (Arhgef6), fibroblast growth factor receptor 1 (Fgfr1),
the TE are still unclear. PRKAA2 mediates differentiation of tro- integrin alpha 2 (Itga2), and platelet derived growth factor receptor,
phoblast stem cells [28]. Reduction in activity of PRKAA1/2 inhibits beta polypeptide (Pdgfrb).
the production of hormones that are necessary for differentiation ILK signaling was downregulated with 12 of 196 genes differ-
of trophoblast stem cells, thereby inhibiting further differentiation entially expressed including myosin, heavy polypeptide 3, skeletal
[29]. ASXL1, a polycomb protein, is obligate for normal embryonic muscle, embryonic (Myh3) and myelocytomatosis oncogene (Myc)
development. (P = 6.46E-03). ILK is a ubiquitously expressed protein that acts in
Individual genes of interest that were significantly altered in the critical phases of cell polarization, migration, proliferation, and
TE of HFD dams include LYR motif containing 4 (Lyrm4) (−27 survival [41]. It is highly expressed in villous cytotrophoblast cells
fold change, q < 0.15), OPA1, mitochondrial dynamin like GTPase in first trimester human chorionic floating villi, and its activity is

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


(Opa1) (–2.072 fold change, q < 0.15), phosphatidylinositol-4,5- increased during syncytialization [42].
bisphosphate 3-kinase catalytic subunit beta (Pik3cb) (–2.75 fold The PPAR signaling pathway was inhibited in HFD EPCs with
change, q < 0.15), and fibroblast growth factor receptor 2 (Fgfr2) a total of nine genes differentially expressed (P = 9.74E-04). These
(–3.428 fold change, q < 0.15). Lyrm4 encodes for an iron–sulfur genes include interleukin 1 receptor-like 2 (Il1rl2), Pdgfrb, tumor
cluster protein important in oxidative phosphorylation [30]. Opa1 necrosis factor receptor superfamily, member 1b (Tnfrsf1b), and nu-
encodes for a mitochondria-shaping protein that controls efficiency clear receptor co-repressor 2 (Ncor2). PPARs are known to modulate
of steroidogenesis in trophoblasts [31], and Pik3cb is involved in inflammatory responses and energy homeostasis and are critically
trophoblast differentiation [32]. The fibroblast growth factor (FGF) important to early placenta development [43, 44].
signaling pathway has been shown to be critical for trophoblast pro- Additionally, upstream analysis using IPA revealed activation of
liferation and differentiation [33]. Fgfr2 mutant mice display multi- transforming growth factor, beta 1 (TGF-b1), tumor necrosis factor
ple placental defects, including a failure of the labyrinthine layer to (TNF), interleukin 6 (IL-6), and SMAD family member 4 (SMAD
develop, and die shortly after implantation [33], therefore downreg- 4). Examples of individual genes that were significantly altered in
ulation of Fgfr2 in TE of HFD dams is of particular interest. EPC of HFD dams include chemokine (C-C motif) receptor-like 2
(Ccrl2), a chemoattractant receptor, which was downregulated 2.46
log2FC in HFD EPCs, and phospholipase A2, group IIF (Pla2g2f),
Maternal obesity alters multiple pathways in EPC which is involved in lipoprotein modification [45] and was downreg-
To determine the progression of changes that were identified early ulated 2.26 log2FC in HFD EPCs. fibroblast growth factor 4 (Fgf4),
in development in HFD TE, we generated strand-specific total RNA- which is vital to for the maintenance of undifferentiated trophoblast
Seq libraries after ribosomal RNA depletion of EPC from mouse stem cells, was upregulated 5.08 log2FC, and bone morphogenetic
embryos at e6.5. 32,193 unique transcripts were identified in the protein 7 (Bmp7), which can induce trophoblast differentiation, was
EPC. Using an FDR < 0.05, 350 genes were differentially expressed upregulated 1.99 log2FC.
between control and HFD EPCs (89 genes downregulated and 261
genes upregulated), and using an FDR < 0.1, 595 genes were
differentially expressed (188 downregulated and 407 upregulated) Sex-specific differences in the transcriptome of the EPC
(Supplemental Figure 1), (Supplemental Table 4). It is well established that the placenta is a sexually dimorphic or-
We again utilized IPA to highlight key alterations between all gan [46]. However, the timing at which this dimorphism first de-
(males and females combined) HFD mice and all control mice. Key velops is not known nor whether this process occurs prior to the
top canonical pathways disrupted included the signaling pathways production of sex hormones and is thus dependent only on sex chro-
of sphingosine-1-phosphate, sphingomyelin metabolism, p21 acti- mosomes. Therefore, we next stratified our RNA-Seq data by sex.
vated kinases (PAK), ceramide, integrin-linked kinases (ILK), and Using an FDR of <0.05, there were 14 genes that were differentially
peroxisome proliferator-activated receptor (PPAR) (Supplemental expressed between male control and male HFD EPCs, including 3-
Table 3). hydroxybutyrate dehydrogenase, type 1 (Bdh1) which is important
Sphingosine-1-phosphate (S1P) is a bioactive lipid derived from in fatty acid catabolism (–1.47 log2FC in HFD males) and coagula-
the metabolism of sphingomyelin [34], and it plays a crucial role tion factor II (thrombin) receptor (F2r), which is a thrombin receptor
in vascular development, immune cell trafficking, and neurogenesis. gene (1.42 log2FC in HFD males). By expanding the FDR to < 0.15,
The sphingosine-1-phosphate signaling pathway was significantly we identified 50 genes that were differentially expressed between
altered in EPC of obese dams (P = 1.9E-03). A total of 10 of 125 male control and male HFD EPCs, of which 17 genes were down-
genes comprising this pathway were differentially expressed. Many regulated and 33 were upregulated (Supplemental Table 5). Of major
of the genes in this pathway, including Pik3cb, N-acylsphingosine significance, there were no genes that were differentially expressed
amidohydrolase 1 (Asah1), sphingomyelin phosphodiesterase 1, acid between female control and female HFD EPCs, suggesting that males
lysosomal (Smpd1), sphingomyelin phosphodiesterase 2, neutral are much more susceptible to the effects of maternal obesity.
(Smpd2), and growth factor receptor bound protein 2 (Grb2) have Canonical pathways altered by an HFD in male EPCs as com-
been correlated with human obesity and insulin resistance [35–38]. pared to control male EPCs included pathways previously identified
Of note, S1P is elevated in the plasma of obese mice and humans, such as axonal guidance, vitamin D receptor/retinoid X receptor
and plasma S1P concentrations positively correlate with measures of (VDR/RXR) activation, PPAR signaling, and PAK signaling, as well
metabolic disease in humans [39]. as pathways found to be unique to the males such as sertoli cell-
PAK signaling was activated by maternal obesity (P = 3.63E-04). sertoli cell junction signaling, ketogenesis, and androgen biosynthe-
PAKs are protein kinases involved in a wide array of biologic func- sis (Supplemental Table 3). Upstream regulators that were activated
tions including gene transcription, apoptosis, and cellular morphol- included TGF-β1, parathyroid hormone (PTH), and endothelin-1
ogy [40]. A total of 10 of 101 genes in this pathway were differen- (EDN1). PPARG and Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2)
tially expressed, including Rac/Cdc42 guanine nucleotide exchange were repressed by an HFD in EPC of male embryos.
800 T. J. Stuart, 2018, Vol. 98, No. 6

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


Figure 1. Venn diagram of some of the most significant canonical pathways found using IPA that were either unique to TE (e4.5) or EPC (e6.5) or overlapped.

When we compared all female EPCs to all male EPCs, we found ternal plasma at e6.5 to investigate whether changes in the ma-
68 genes differentially expressed with an FDR < 0.05. Of these ternal metabolic profile correlated with changes in the early pla-
68 genes, 12 are known to be sex-linked. When we opened the centa transcriptome. The results of global metabolic profiling of
FDR to 0.15 (326 genes, Supplemental Table 6) and performed IPA, maternal plasma demonstrated that an HFD has a profound effect
we found that several canonical pathways were downregulated in on metabolism, with greater than a third of metabolites measured
males including glutamate receptor signaling (Supplemental Table 3). demonstrating a significant difference between obese and control
Downregulation of this pathway was largely in part to decreased ex- dams. A total of 522 compounds of known identity were detected in
pression of glutamate receptor, metabotropic 1 (Grm1), which had a plasma samples. Levels of 192 biochemicals were significantly differ-
–6.8 log2FC (FDR 0.05) as compared to females. Inhibited upstream ent between HFD and control samples (P ≤ 0.05); 89 were present in
regulators included Sonic hedgehog (SHH) and brain-derived neu- higher levels and 103 were present in lower levels in plasma derived
rotrophic factor (BDNF). Activated upstream regulators included from obese dams compared to controls (Supplemental Table 7).
Growth factor receptor bound protein 2 (GRB2) and SRY-box 7 Principal components analysis revealed distinct separation be-
(SOX7). tween the plasma samples derived from the control and obese ani-
mals (Figure 2). The large degree of separation between these sample
Multiple pathways overlapped between HFD TE and types indicates that global metabolism changed in a significant man-
HFD EPC ner with diet.
There was considerable overlap in canonical pathways altered by
an HFD between TE at e4.5 and EPC at e6.5, including VDR/RXR Multiple lipid species in maternal plasma are increased
activation, ET-1 signaling, axonal guidance signaling, p38 MAPK by obesity
signaling, and PPARα/RXRα activation suggesting the importance
Exposure to an HFD resulted in significant changes in the lev-
of these pathways in the development of abnormal placenta function
els of biochemicals involved in fatty acid metabolism. There were
in an obese pregnancy (Figure 1). Additionally, there were changes
significant increases in the levels of several fatty acid species in plasma
in expression of genes at e4.5 that persisted at e6.5. For example,
(Table 2) in the HFD dams. Levels of long-chain fatty acids (LCFA)
several antiangiogenic genes, including angiopoietin 2 (Angpt2), ar-
myristate, myristoleate, palmitate, and stearate were significantly el-
restin, beta 1 (Arrb1), collagen, type IV, alpha 1 (Col4a1), collagen,
evated in the plasma of HFD dams. Similar to LCFA, several polyun-
type IV, alpha 2 (Col4a2), serine (or cysteine) peptidase inhibitor,
saturated fatty acids (PUFA) were also significantly increased by an
clade E, member 1 (Serpine1), and serine (or cysteine) peptidase in-
HFD. Specifically, in HFD maternal plasma, the levels of ω-6 fatty
hibitor, clade F, member 1 (Serpinf1) were significantly increased by
acids (dihomo-linolenate, adrenate, docosapenaenoate, and dihomo-
an HFD (P < 0.05 vs. control) in both TE and EPC.
linoleate) were elevated (>2 fold) compared to controls, while the
level of ω-3 fatty acids did not differ between the groups.
Obesity alters the metabolome in maternal plasma at When dietary fats are in abundance, animals can metabolize them
e6.5 in multiple ways. They can store them in adipose tissue, shunt them
Given that our transcriptome results implicated early changes in to other complex lipids (phospholipids, sphingolipids), or they can
lipid metabolism, we next performed metabolic profiling in ma- oxidize them for energy production via mitochondrial β-oxidation.
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 801

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


Figure 2. Principal component analysis of plasma samples from control (pink) and HFD (red) pregnant dams at e6.5. N = 10 per diet intervention.

Table 2. Fatty acids altered in maternal plasma at e6.5 as a result of diet-induced obesity as detected using liquid chromatography/mass
spectroscopy. N = 10 per diet intervention for plasma analyses. Red and green shaded cells indicate P ≤ 0.05 (red indicates that the mean
values are significantly higher; green values significantly lower). Light red and light green shaded cells indicate 0.05 < P < 0.10.

The increases in glycerophospholipids and sphingomyelin species Finally, the levels of metabolites involved in sphingolipid
together with decreases in carnitine and its metabolic precursor de- metabolism were dramatically altered in the plasma of obese dams.
oxycarnitine detected in the plasma of obese dams suggest significant Of the 24 metabolites in this pathway, >50% were significantly
alterations in β-oxidation. higher in HFD dams (Table 3).
802 T. J. Stuart, 2018, Vol. 98, No. 6

Table 3. Sphingolipid metabolites altered in maternal plasma at e6.5 as a result of diet-induced obesity as detected using liquid chromatog-
raphy/mass spectroscopy. N = 10 per diet intervention. Red and green shaded cells indicate P ≤ 0.05 (red indicates that the mean values
are significantly higher; green values significantly lower). Light red and light green shaded cells indicate 0.05 < P < 0.10.

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


Table 4. Lysine metabolites altered in maternal plasma at e6.5 as a result of diet-induced obesity as detected using liquid chromatogra-
phy/mass spectroscopy. N = 10 per diet intervention. Red and green shaded cells indicate P ≤ 0.05 (red indicates that the mean values are
significantly higher; green values significantly lower). Light red and light green shaded cells indicate 0.05 < P < 0.10.

Multiple amino acid pathways are influenced by is present, the oxidation of amino acids becomes an alterna-
maternal obesity tive energy source by entering the citric acid cycle at different
Plasma of obese dams was enriched in several lysine catabolites points.
(e.g. 2-aminoadipate, 2-oxoadipate, glutarylcarnitine), suggesting
that maternal obesity alters lysine availability and/or utilization
(Table 4). Acetylated lysine metabolites were also significantly lower Maternal HFD is associated with altered microbial
in plasma of HFD dams. metabolites in plasma
Glycine and several glycine metabolites were significantly re- It is well established that the gut microbiota is altered by obe-
duced in plasma of HFD dams. Interestingly, recent studies have sity [49–51]. In our study, HFD dams exhibited alterations in
shown that low glycine levels are associated with insulin re- microbial metabolites derived from tyrosine and tryptophan (phe-
sistance and increased visceral adiposity [47, 48]. Because the nol sulfate, p-cresol sulfate, p-cresol-glucuronide, indolepropionate,
utilization of carbohydrates is impaired when insulin resistance and 3-indoxyl sulfate) in their plasma (Figure 3). Further evidence for
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 803

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


Figure 3. Microbial metabolites significantly altered (P < 0.05) using Welch’s two-sample t-test in maternal plasma at e6.5 as detected using liquid chromatogra-
phy/mass spectroscopy. N = 10 per diet intervention.

an altered microbiome was manifested in the bile acid profiles. Bile compared to the controls (0.8731 ± 0.017 vs 0.8552 ± 0.013, P =
acids play an important role in lipid metabolism and obesity. While 0.408). At e18.5, the RI of the umbilical artery in the control group
the levels of multiple primary (taurocholate, chenocdeoxycholate) was 0.9031 ± 0.0107 vs 0.917 ± 0.0113 in the HFD group (P =
and secondary bile acids (taurodeoxycholate ursodeoxycholate, tau- 0.421). The average RI of the uterine arteries in the control group at
roursodeoxycholate, hyodeoxycholate) were altered by exposure to e12.5 was 0.6097 ± 0.0532 vs 0.6713 ± 0.1254 in the HFD group
HFD in plasma, the changes in secondary bile acids were more pro- (P = 0.635), and at e18.5, the control group uterine artery average
nounced. Secondary bile acids are generated when microbiota in the RI was 0.5634 ± 0.0424 vs 0.5954 ± 0.0059 in the HFD group (P
gut deconjugate and dehydroxylate bile acids synthesized by the host. = 0.398). Therefore, the RI was slightly, but not significantly, ele-
Taken together, these findings strongly suggest that the microbiome vated in the uterine arteries of dams fed an HFD by 10.1% at e12.5
is altered by exposure to an HFD, and importantly, the early embryo and 5.7% at e18.5. Additionally, the RI of the umbilical arteries of
may be exposed to altered microbial metabolites generated by the HFD fetuses also trended upward slightly compared to controls by
maternal microbiome in the setting of maternal obesity. 2.1% at e12.5 and 1.5% at e18.5. However, these results were not
The results of the above transcriptomic and metabolomic studies statistically significant.
are summarized in Table 5.

Vascular development is impaired in placenta of HFD


dams Discussion
Given that maternal exposure to an HFD altered several path- In this study, we demonstrated that maternal obesity alters the ma-
ways and metabolites vital to vascular development such as ternal metabolome very early in pregnancy and is associated with
sphingosine-1-phosphate, the ET-1 signaling pathway, and sphin- marked changes in expression of genes involved in key pathways such
golipid metabolism, we sought to explore downstream phenotypic as sphingolipid metabolism, angiogenesis, and fatty acid metabolism
changes in placenta vasculature. Therefore, we next stained mid and in cells that will eventually give rise to the placenta. While multiple
late gestation placentas from control and HFD dams with PLVAP studies have demonstrated that obesity in pregnancy alters placenta
antibody to quantify vascularity. At e12.5, placentas of control males structure and function [11] [52–54], our data surprisingly show that
had the highest microvessel density, followed by control females and the abnormal metabolic milieu of maternal obesity sets the stage
HFD females. HFD males had the lowest microvessel density, which very early in pregnancy by altering the transcriptome of placenta
was 31.4% lower than control males (P < 0.05) (Figure 4). The progenitor cells in the TE and EPC, which in turn are associated
changes in microvessel density persisted throughout the remainder of with profound implications for placenta development and function.
gestation, and e18.5 male placentas from HFD dams had ∼30% less In the mouse, by embryonic day 4 (e4), the totipotent cells of the
microvessel density when compared to both control male and con- morula differentiate into two distinct lineages: the outer layer of
trol female placentas (P < 0.05). HFD female placentas had ∼20% cells, the TE, develops into the extraembryonic tissue, and the inner
decreased microvessel density when compared to controls (Figure 5). cell mass gives rise to the fetal structures [55]. The multipotent layer
Given that we found changes in microvessel density of HFD pla- of TE cells anchors to the uterus and divides to form the extraem-
centas, we sought to determine whether an HFD altered arterial flow bryonic ectoderm and EPC which then forms the spongiotrophoblast
and resistance in the placenta and uterus. Umbilical artery and uter- and trophoblast giant cells [56]. Therefore, the window between em-
ine artery dopplers were performed on HFD and control dams and bryonic days 4 and 6.5 provides an ideal time to investigate early
fetuses at e12.5 and e18.5. The average RI measured in the umbilical alterations of the placenta that are direct consequences of maternal
artery of fetuses at e12.5 was slightly increased in the HFD group as exposures.
Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020
T. J. Stuart, 2018, Vol. 98, No. 6

Table 5. Summary of major metabolomic and transcriptomic results.


804
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 805

e12.5 Placenta Microvessel Density Perhaps most striking was our finding of markedly altered sphin-
golipid metabolism in plasma of obese dams very early in pregnancy.
0.0008
* This directly correlated with changes in gene expression regulating
Vessels per unit area (um2) * pvalue <0.05 sphingosine-1-phosphate and ceramide signaling pathways in EPC,
0.0006 which persisted in placenta at e12.5 as shown in our previous studies
[11]. Upregulation of these pathways inhibit angiogenesis and result
0.0004 in endothelial dysfunction by abolishing vascular endothelial growth
factor (VEGF) or insulin-stimulated endothelial nitric oxide synthase
activation and subsequent nitric oxide production from endothelial
0.0002
cells [62]. Indeed, many of these key pathways were disrupted in TE

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


and EPC. Further, excess ceramides play a major role in lack of re-
0.0000 modeling of the maternal spiral arteries that occurs in preeclampsia,
which is associated with maternal obesity [63]. Thus, changes in ma-
e

e
al

al

al

al
M

m
ternal sphingolipid metabolism could be a mechanism through which
Fe

Fe
ol

FD
tr

obesity results not only in inflammation and insulin resistance in the


ol

FD
H
on

tr

H
C

on

mother but also activates a transcription program in placenta pro-


C

genitors resulting in abnormal vascularization in placenta of obese


Diet and Sex
dams.
Figure 4. Microvessel density was decreased in male HFD placenta at e12.5 Multiple phospholipids, including 1-stearoyl-2-arachidonoyl-
as measured via immunohistochemistry staining with PLVAP antibody (N = GPC, were elevated in maternal plasma of obese dams at e6.5. This
5–7 per group, ∗ P < 0.05). key phospholipid is an important regulator of p38 MAPK activity
under conditions of cell stress and lipotoxicity [64], and the p38
MAP kinase pathway was significantly elevated in EPC at e6.5. The
e18.5 Placenta Microvessel Density p38 group of MAP kinases serve as a nexus for signal transduction
* and play a vital role in numerous biological processes including in-
0.0006 * * p value < 0.05 flammation and placental angiogenesis [65], thus providing another
Vessels per unit area (um2)

link between changes in the maternal metabolome and changes in


gene expression in the EPC.
0.0004 HFD dams exhibited differences in several lysine metabolites
compared to controls. Acetylated lysine molecules are commonly
found in histone proteins; the acetylation status of lysine in histones
0.0002 is correlated with active gene expression as acetylation of lysine ef-
fectively removes the unmodified amino acid’s positive charge and
hence reduces the interaction between the histone tail and nucle-
0.0000 osome [66]. Additionally, high-throughput proteomic assays have
demonstrated that lysine acetylation of proteins involved in immune
e

e
al

al

al

al

modulation and metabolism occurs which can alter function [67].


M

m
Fe

Fe
ol

FD

Therefore, changes in acetylation of lysine could have profound


tr

ol

FD
H
on

tr

effects on development-related gene expression patterns and post-


C

on
C

translational effects by altering the function of proteins involved in


Diet and Sex
metabolic signaling.
Figure 5. Microvessel density remained decreased in male HFD placentas at
Another key change in the maternal metabolome of obese dams
e18.5 as measured via immunohistochemistry staining with PLVAP antibody that is likely to influence placenta and fetal development was a
(N = 3–6 per group, ∗ P < 0.05). marked decrease in myo and chiro-inositol. Inositols are a family
of simple carbohydrates, and these two stereoisomers play a key role
in the insulin pathway, acting synergistically as insulin-sensitizing
Not surprisingly, an HFD induced marked elevations in satu- agents through the enhancement of glucose peripheral tissue up-
rated fatty acids, including palmitate, myristoleate, myristate, and take and glycogen synthesis [68]. Of note, myo-inositol/D-chiro-
stearate, in plasma of obese dams in early pregnancy. Levels of these inositol supplementation in pregnant mice with a metabolic-like syn-
pro-inflammatory fatty acids are elevated in obese humans and are drome phenotype improves blood pressure, glucose tolerance, and
linked to the development of insulin resistance and other metabolic leptin levels and prevents increased maternal weight gain [69]. Myo-
disorders that accompany obesity [57]. Additionally, we observed a inositol also regulates endothelial function by decreasing oxidative
disrupted balance of ω-6:3 ratio in obese dams. ω-6 fatty acids are stress, enhancing endothelial nitric oxide synthase and nitric oxide
pro-inflammatory and enhance adipogenesis, whereas ω-3 fatty acids activity [70]. Thus, low levels of these important metabolites may
are associated with improved insulin action and decreased adipos- contribute to insulin resistance associated with maternal obesity.
ity [58–60]. We have previously shown that this pro-inflammatory Multiple studies have demonstrated that maternal obesity re-
metabolic milieu in the mother results in marked oxidative stress in sults in a pro-inflammatory state in the placenta [71, 72]. These
the preimplantation embryo [61], and prevention of oxidative stress studies have all been performed in term or near term placenta, yet
precludes the development of obesity in the offspring, thus demon- our transcriptome data showing activation of multiple inflammatory
strating the central role of oxidative stress and inflammation in the pathways and genes including IL-6 signaling, suggest that this pro-
programming of the offspring [61]. cess occurs very early in gestation. The inflammatory state in TE
806 T. J. Stuart, 2018, Vol. 98, No. 6

and EPC of HFD dams is likely due to the marked changes in lipid cies reduce cytotrophoblast proliferation and placental growth factor
species such as palmitate, omega-6 fatty acids, sphingolipids, and (PlGF) production and are thought to contribute to the development
their metabolites, like eicosanoids, that are significantly elevated in of preeclampsia [75].
the plasma of obese dams during early pregnancy. In addition to changes in placenta development and function,
There was significant overlap in the gene expression pathways obesity may impact fetal development by altering microbial metabo-
that were altered by an HFD in TE and EPC, implying that these lites and molecules to which the fetus is exposed. In our study,
particular pathways may be especially important to early placenta multiple microbial metabolites were altered in plasma of HFD dams.
development (Figure 1). These pathways include ET-1 signaling, p38 Several studies have shown that the composition of gut micro-
MAPK signaling, as well as PPARα/RXRα activation. ET-1 is an ex- biota is altered by obesity [49–51]; however, the functional im-
tremely potent vasoconstrictor that acts directly on vascular smooth plications of an altered microbiome on fetal growth and develop-

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


muscle cells and has been linked to diseases such as hypertension ment are not fully understood. Gomez de Aguero et al. showed
and atherosclerosis [73]. It is well known that obesity is associated that maternal microbiota-derived compounds are transferred from
with endothelial dysfunction, and ET-1-mediated vasoconstriction the mother to the offspring and shape the immune system of the
is increased in obese humans [74]. Additionally, elevated circulating offspring [83]. Therefore, gaining better understanding of the com-
plasma ET-1 levels have been positively correlated with preeclamp- pounds present in maternal plasma and, thus potentially transferred
sia [75] and the production of reactive oxygen species that promote to the offspring, is paramount. The obese maternal plasma pro-
apoptosis of trophoblasts [76]. This correlates with the finding that files showed altered levels of microbial-derived metabolites pro-
placentas of mice with diet-induced obesity display both oxidative duced from amino acids (tyrosine and tryptophan), which have
stress and decreased number of trophoblasts [77]. These changes been suggested to interact with host signaling pathways and affect
may be another mechanism by which an HFD leads to decreased host immunity [84]. Indole, a major tryptophan-derived microbial
microvessel density later in gestation. metabolite we found to be present in higher levels in plasma of
The PPARα/RXRα activation pathway was predicted to be in- HFD dams, has been shown to interact with inflammation-related
hibited by an HFD in both TE and EPC, and the more generalized processes in the human host [85]. Therefore, in addition to altered
PPAR signaling pathway was also predicted to be inhibited, though placenta function, our studies suggest that obesity alters the compo-
this was only statistically significant in EPC. This was an unexpected sition of biochemicals to which the embryo is exposed. This will not
finding given that the physiologic actions of fatty acids are medi- only affect fetal development, but also impact long-term offspring
ated by PPARs. However, it is possible that an overabundance of outcomes.
free fatty acids leads to a subsequent downregulation of these path- Male offspring are known to be more susceptible to the effects
ways. PPARs are highly expressed in the placenta and thought to be of intrauterine exposures such as maternal obesity and diabetes
involved in trophoblast differentiation and function [78], and defi- [86, 87], and it is possible that the placenta plays a key role in
ciencies of both PPARγ and PPARδ result in lethal placenta defects this finding. It is recognized that there are sex differences in gene
[44]. The placentas of PPARγ null mice display vascular anomalies expression in normal placenta, which have been attributed to sex
and poor differentiation of the trophoblast [78]. Additionally, in hu- hormones, and it is also known that maternal HFD can affect gene
man studies, gestational diabetes mellitus has been associated with a expression in a sex-specific manner [88]. However, given our pre-
downregulation in term placental PPARα, PPARγ , and RXRα [79] vious findings that a pregestational exposure to obesity also results
In addition to ET-1 and PPAR signaling, there were many other in sex-specific differences in long-term outcomes between males and
genes and pathways altered by an HFD in early placenta that are re- females [11], it is possible that sex-specific developmental program-
lated to angiogenesis and vascular development, including the PEDF ming is occurring even prior to the expression of sex hormones.
signaling pathway and the pathway for leptin signaling in obesity. When we compared all male EPCs to all female EPCs, we found
The PEDF signaling pathway was activated by an HFD in TE. PEDF alterations in several interesting pathways, many of which are related
is a widely expressed glycoprotein that acts as a potent antiangio- to placenta and vascular development. Glutamate receptor signaling
genic factor [80]. Huppertz et al. recently identified PEDF secretion was downregulated in males. Though glutamate signaling was pre-
as a mechanism by which human trophoblasts restrain angiogenesis viously thought to be limited to the central nervous system, it is
and vascularization later in pregnancy [81]. PEDF binds to VEGFR2 now known that functional glutamate receptors are found in many
and blocks the stimulatory effect of VEGF. These changes in key different organs, including skin and placenta [89]. Grm1 is upreg-
genes regulating vascular development in the TE and EPC were cor- ulated in some forms of cancer, and it has been found to enhance
related with changes we found in microvessel density in mid and late angiogenic signaling [90]. SHH and BDNF, both upstream regula-
gestation placenta, suggesting that the mechanism underlying abnor- tors, were inhibited in males as compared to females, and these two
mal placenta vascularization occurs prior to placentation in the very regulators also play important roles in placenta formation and an-
early embryo. giogenesis. Shh helps to control placenta labyrinth development, and
The canonical pathway for leptin signaling in obesity was also Shh null mice have pronounced defects in yolk sac vasculature [91].
activated by an HFD in TE. Leptin is an abundant circulating Additionally, studies have shown Shh to be indirectly angiogenic by
adipokine, and while it is widely known for its role in energy home- upregulating Vegf, angiopoietin 1 (Angpt1), and Angpt2 [92]. Of
ostasis and metabolism, it also participates in a myriad of other note, Vegfc was downregulated in males, which is consistent with
processes including immune function and inflammation [82]. Acti- deactivation of the SHH pathway.
vation of leptin receptors on endothelial cells leads to endothelial BDNF is neurotrophin thought to play key roles in placenta
cell dysfunction via increased oxidative stress responses as well as angiogenesis, cytotrophoblast proliferation, and protection of en-
increases in TNF-α and IL-6 [82], both of which were also predicted dothelial progenitor cells via increased expression of superoxide
to be activated by an HFD in EPC. In the more mature placenta, dismutase, and alterations have been implicated in a wide variety
leptin is produced by syncytiotrophoblast and controls growth of of diseases [93]. Lower levels of BDNF have been found in both pla-
the placenta. High levels of leptin in placentas from obese pregnan- centa and cord blood of women with preeclampsia [94], and BDNF
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 807

was recently shown to be sexually dimorphic in term human pla- sylvania Small Animal Imaging Facility, and the University of Pennsylvania
centas [95]. Baseline differences between male and female EPC may Epigenetics Program for their help with this research.
be one explanation for the increased susceptibility of males to the Conflict of Interest. The authors have declared that no conflict of interest exists.
effects of maternal obesity.
As mentioned above, we found that exposure to an HFD de-
creased microvessel density at both mid and late gestation. Notably, References
this decrease was most pronounced in the placentas of male fetuses. 1. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of Obesity in the
Additionally, we found increased uterine artery resistance in HFD United States, 2009–2010. NCHS Data Brief 2012;(82):1–8.
dams and a very slight trend toward increased umbilical artery re- 2. Marchi J, Berg M, Dencker A, Olander EK, Begley C. Risks associated
sistance in fetuses of dams fed an HFD. However, these results were

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


with obesity in pregnancy, for the mother and baby: a systematic review
not statistically significant. Given that the histological changes we of reviews. Obes Rev 2015; 16(8):621–638.
noted in microvessel density were more significant in males, and we 3. Deshmukh VL, Jadhav M, Yelikar K. Impact of HIGH BMI on pregnancy:
were not able to determine the sex of the fetuses undergoing doppler maternal and foetal outcome. J Obstet Gynecol India 2016; 66(S1):192–
197.
imaging, we speculate that this may be why the increase in RI in the
4. Yao R, Park BY, Caughey AB. The effects of maternal obesity on perinatal
uterine and umbilical arteries was not statistically significant.
outcomes among those born small for gestational age. J Matern Fetal
In conclusion, our data demonstrate that diet-induced obe- Neonatal Med 2017; 30(12):1417–1422.
sity in the female mouse profoundly affects the maternal plasma 5. O’Reilly JR, Reynolds RM. The risk of maternal obesity to the long-term
metabolome and alters gene expression pathways in the TE and EPC health of the offspring. Clin Endocrinol 2013; 78(1):9–16.
that are vital to placenta development and necessary to support the 6. Dimasuay KG, Boeuf P, Powell TL, Jansson T. Placental responses to
developing embryo. This is the first study to demonstrate that mater- changes in the maternal environment determine fetal growth. Front Physiol
nal obesity alters the transcriptome at the earliest stages of murine 2016; 7:12.
placenta development. IPA highlighted significant clustering of genes 7. Wang CY, Liao JK. A mouse model of diet-induced obesity and insulin
involved in sphingolipid metabolism, vascular development, and in- resistance. Methods Mol Biol 2012; 821:421–433.
8. Skaznik-Wikiel ME, Swindle DC, Allshouse AA, Polotsky AJ,
flammation, and we speculate that these early changes are directly
McManaman JL. High-fat diet causes subfertility and compromised
associated with the decreased placenta microvessel density we ob-
ovarian function independent of obesity in mice1. Biol Reprod 2016;
served later in gestation. These alterations may represent some of 94(5):108.
the many complex ways in which maternal obesity exerts its effects 9. Bibi S, Kang Y, Du M, Zhu MJ. Maternal high-fat diet consumption
on the offspring via the placenta. enhances offspring susceptibility to DSS-induced colitis in mice. Obesity
2017; 25(5):901–908.
10. Boudoures AL, Chi M, Thompson A, Zhang W, Moley KH. The effects
Supplementary data of voluntary exercise on oocyte quality in a diet-induced obese murine
Supplemental Table 1. Antibodies used for immunohistochemistry model. Reproduction 2016; 151(3):261–270.
11. Sasson I, Vitins AP, Mainigi MA, Moley KH, Simmons RA. Pre-gestational
staining.
vs gestational exposure to maternal obesity differentially programs the
Supplemental Table 2. Gene expression profiling of trophectoderm
offspring in mice. Diabetologia 2015; 58(3):615–624.
from e4.5 blastocysts from control and HFD dams. Using an FDR of 12. Howell KR, Powell TL. Effects of maternal obesity on placen-
≤0.05, 5285 genes were differentially expressed between HFD and tal function and fetal development. Reproduction 2017; 153(3):R97–
controls (1 gene downregulated and 5284 genes upregulated). R108.
Supplemental Table 3. Canonical pathways with P ≤ 0.05 for all 13. Hellmuth C, Lindsay KL, Uhl O, Buss C, Wadhwa PD, Koletzko B,
transcriptomic analyses. Entringer S. Association of maternal prepregnancy BMI with metabolomic
Supplemental Figure 1. Volcano plot of all genes sequenced, compar- profile across gestation. Int J Obes 2017; 41(1):159–169.
ing all control EPC samples to all HFD EPC samples at e6.5. Noted 14. Benton SJ, Ly C, Vukovic S, Bainbridge SA. Andree Gruslin award lecture:
in red are genes with an FDR ≤0.15. Metabolomics as an important modality to better understand preeclamp-
sia. Placenta 2017; 60(suppl 1):S32–S40.
Supplemental Table 4. RNA-Seq of e6.5 EPC, comparing all control
15. Shea K, Geijsen N. Dissection of 6.5 dpc mouse embryos. J Vis Exp 2007;
EPCs to all HFD EPCs. Using an FDR of ≤0.1, 595 genes were
(2):160.
differentially expressed (188 downregulated and 407 upregulated). 16. Downs KM, Davies T. Staging of gastrulating mouse embryos by mor-
Supplemental Table 5. RNA-seq of e6.5 EPCs, comparing male con- phological landmarks in the dissecting microscope. Development 1993;
trol EPCs to male HFD EPCs. Using an FDR of ≤0.15, there were 118(4):1255–1266.
50 genes that were differentially expressed (17 downregulated and 17. Tam PP, Rossant J. Mouse embryonic chimeras: tools for studying mam-
33 upregulated). malian development. Development 2003; 130(25):6155–6163.
Supplemental Table 6. RNA-seq of e6.5 EPCs, comparing all female 18. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut
EPCs to allmale EPCs. Using an FDR of ≤0.15, there were 326 genes P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner.
differentially expressed (277 downregulated and 49 upregulated). Bioinformatics 2013; 29(1):15–21.
19. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose
Supplemental Table 7. Global metabolic profiling of e6.5 mater-
program for assigning sequence reads to genomic features. Bioinformatics
nal plasma. Levels of 192 biochemicals were significantly different
2014; 30(7):923–930.
between HFD and control samples (P ≤ 0.05); 89 were present in 20. Datta S, Xia X-Q, Bhattacharjee S, Jia Z. Advances in statistical medicine.
higher levels, and 103 were present in lower levels. Comput Math Methods Med 2014; 2014:316153.
21. Hernandez-Andrade E, Ahn H, Szalai G, Korzeniewski SJ, Wang B, King
M, Chaiworapongsa T, Than NG, Romero1 R. Evaluation of utero-
Acknowledgments placental and fetal hemodynamic parameters throughout gestation in preg-
The authors would like to acknowledge John Tobias, Teri Ord, Metabolon Inc, nant mice using high-frequency ultrasound. Ultrasound Med Biol 2014;
the Children’s Hospital of Philadelphia Pathology core, University of Penn- 40(2):351–360.
808 T. J. Stuart, 2018, Vol. 98, No. 6

22. Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE, Moley 43. Stienstra R, Duval C, Müller M, Kersten S. PPARs, obesity, and inflam-
KH. Diet-induced obesity model: abnormal oocytes and persistent growth mation. PPAR Res 2007; 2007:1–10.
abnormalities in the offspring. Endocrinology 2010; 151(8):4039–4046. 44. Barak Y, Sadovsky Y, Shalom-Barak T. PPAR signaling in placental de-
23. Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Nor- velopment and function. PPAR Res 2008; 2008:1–11.
man RJ, Robker RL. High-fat diet causes lipotoxicity responses in 45. Sato H, Kato R, Isogai Y, Saka G-I, Ohtsuki M, Taketomi Y, Yamamoto
cumulus–oocyte complexes and decreased fertilization rates. Endocrinol- K, Tsutsumi K, Yamada J, Masuda S, Ishikawa Y, Ishii T et al. Anal-
ogy 2010; 151(11):5438–5445. yses of group III secreted phospholipase A2 transgenic mice reveal po-
24. Spradley FT, Palei AC, Granger JP. Immune mechanisms linking obesity tential participation of this enzyme in plasma lipoprotein modification,
and preeclampsia. Biomolecules 2015; 5(4):3142–3176. macrophage foam cell formation, and atherosclerosis. J Biol Chem 2008;
25. Han HJ, Lee YJ. Insulin stimulates Ca2+ uptake via PKC, cAMP, and p38 283(48):33483–33497.

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


MAPK in mouse embryonic stem cells. Life Sci 2005; 76(25):2903–2919. 46. Myatt L, Maloyan A. Obesity and placental function. Semin Reprod Med
26. Wessler I, Kilbinger H, Bittinger F, Unger R, Kirkpatrick CJ. The non- 2016; 34(01):042–049.
neuronal cholinergic system in humans: expression, function and patho- 47. Lustgarten MS, Price LL, Phillips EM, Fielding RA. Serum glycine is asso-
physiology. Life Sci 2003; 72(18–19):2055–2061. ciated with regional body fat and insulin resistance in functionally-limited
27. Sastry BV. Human placental cholinergic system. Biochem Pharmacol older adults. PLoS One 2013; 8(12):e84034.
1997; 53(11):1577–1586. 48. Cheng Y, Meng Q, Wang C, Li H, Huang Z, Chen S, Xiao F, Guo F.
28. Carey EA, Albers RE, Doliboa SR, Hughes M, Wyatt CN, Natale DR, Leucine deprivation decreases fat mass by stimulation of lipolysis in white
Brown TL. AMPK knockdown in placental trophoblast cells results in adipose tissue and upregulation of uncoupling protein 1 (UCP1) in brown
altered morphology and function. Stem Cells Dev 2014; 23(23):2921– adipose tissue. Diabetes 2010; 59(1):17–25.
2930. 49. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon
29. Zhong W, Xie Y, Abdallah M, Awonuga AO, Slater JA, Sipahi L, Puscheck JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA 2005;
EE, Rappolee DA. Cellular stress causes reversible, PRKAA1/2-, and 102(31):11070–11075.
proteasome-dependent ID2 protein loss in trophoblast stem cells. Repro- 50. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI.
duction 2010; 140(6):921–930. An obesity-associated gut microbiome with increased capacity for energy
30. Lim SC, Friemel M, Marum JE, Tucker EJ, Bruno DL, Riley LG, harvest. Nature 2006; 444(7122):1027–1131.
Christodoulou J, Kirk EP, Boneh A, DeGennaro CM, Springer M, Mootha 51. Murphy EF, Cotter PD, Healy S, Marques TM, O’Sullivan O, Fouhy F,
VK et al. Mutations in LYRM4, encoding iron–sulfur cluster biogenesis Clarke SF, O’Toole PW, Quigley EM, Stanton C, Ross PR, O’Doherty
factor ISD11, cause deficiency of multiple respiratory chain complexes. RM et al. Composition and energy harvesting capacity of the gut micro-
Hum Mol Genet 2013; 22(22):4460–4473. biota: relationship to diet, obesity and time in mouse models. Gut 2010;
31. Wasilewski M, Semenzato M, Rafelski SM, Robbins J, Bakardjiev AI, 59(12):1635–1642.
Scorrano L. Optic atrophy 1-dependent mitochondrial remodeling con- 52. Ramsay JE, Ferrell WR, Crawford L, Wallace AM, Greer IA, Sattar N.
trols steroidogenesis in trophoblasts. Curr Bio 2012; 22(13):1228–1234. Maternal obesity is associated with dysregulation of metabolic, vascular,
32. Kent LN, Konno T, Soares MJ Phosphatidylinositol 3 kinase modulation and inflammatory pathways. J Clin Endocrinol Metab 2002; 87(9):4231–
of trophoblast cell differentiation. BMC Dev Biol 2010; 10(1):97. 4237.
33. Kunath T, Yamanaka Y, Detmar J, MacPhee D, Caniggia I, Rossant J, 53. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima
Jurisicova A. Developmental differences in the expression of FGF receptors Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased
between human and mouse embryos. Placenta 2014; 35(12):1079–1088. oxidative stress in obesity and its impact on metabolic syndrome. J Clin
34. Mendelson K, Evans T, Hla T, Sphingosine 1-phosphate signalling. De- Invest 2004; 114(12):1752–1761.
velopment 2014; 141(1):5–9. 54. Rajasingam D, Seed PT, Briley AL, Shennan AH, Poston L. A prospec-
35. Wang K, Li WD, Zhang CK, Wang Z, Glessner JT, Grant SF, Zhao H, tive study of pregnancy outcome and biomarkers of oxidative stress in
Hakonarson H, Price RA. A genome-wide association study on obesity nulliparous obese women. Am J Obstet Gynecol 2009; 200(4):395.e1–
and obesity-related traits. PLoS One 2011; 6(4):e18939. 395.e9.
36. Le Stunff C, Dechartres A, Mariot V, Lotton C, Trainor C, Miraglia Del 55. Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse
Giudice E, Meyre D, Bieche I, Laurendeau I, Froguel P, Zelenika D, Fallin models. Nat Rev Genet 2006; 7(3):185–199.
D et al. Association analysis indicates that a variant GATA-binding site 56. Cross JC. Genes regulating embryonic and fetal survival. Theriogenology
in the PIK3CB promoter is a Cis-acting expression quantitative trait locus 2001; 55(1):193–207.
for this gene and attenuates insulin resistance in obese children. Diabetes 57. Bush NC, Basu R, Rizza RA, Nair KS, Khosla S, Jensen MD. Insulin-
2008; 57(2):494–502. mediated FFA suppression is associated with triglyceridemia and in-
37. Kolak M, Gertow J, Westerbacka J, Summers SA, Liska J, Franco- sulin sensitivity independent of adiposity. J Clin Endocrinol Metab 2012;
Cereceda A, Orešič M, Yki-Järvinen H, Eriksson P, Fisher RM. Expression 97(11):4130–4138.
of ceramide-metabolising enzymes in subcutaneous and intra-abdominal 58. Pisani DF, Ghandour RA, Beranger GE, Le Faouder P, Chambard JC,
human adipose tissue. Lipids Health Dis 2012; 11(1):115. Giroud M, Vegiopoulos A, Djedaini M, Bertrand-Michel J, Tauc M,
38. Desbuquois B, Carre N, Burnol AF. Regulation of insulin and type 1 Herzig S, Langin D et al. The ω6-fatty acid, arachidonic acid, regulates
insulin-like growth factor signaling and action by the Grb10/14 and the conversion of white to brite adipocyte through a prostaglandin/calcium
SH2B1/B2 adaptor proteins. FEBS J 2013; 280(3):794–816. mediated pathway. Mol Metab 2014; 3(9):834–847.
39. Kowalski GM, Carey AL, Selathurai A, Kingwell BA, Bruce CR. 59. Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ra-
Plasma sphingosine-1-phosphate is elevated in obesity. PLoS One 2013; tio in cardiovascular disease and other chronic diseases. Exp Biol Med
8(9):e72449. (Maywood) 2008; 233(6):674–688.
40. Jaffer ZM, Chernoff J. p21-activated kinases: three more join the Pak. Int 60. Blasbalg TL, Hibbeln JR, Ramsden CE, Majchrzak SF, Rawlings RR.
J Biochem Cell Biol 2002; 34(7):713–717. Changes in consumption of omega-3 and omega-6 fatty acids in the United
41. Widmaier M, Rognoni E, Radovanac K, Azimifar SB, Fässler R. Integrin- States during the 20th century. Am J Clin Nutr 2011; 93(5):950–962.
linked kinase at a glance. J Cell Sci 2012; 125(8):1839–1843. 61. Sen S, Simmons RA. Maternal antioxidant supplementation prevents
42. Butler TM, Elustondo PA, Hannigan GE, MacPhee DJ. Integrin-linked adiposity in the offspring of Western diet-fed rats. Diabetes 2010;
kinase can facilitate syncytialization and hormonal differentiation of the 59(12):3058–3065.
human trophoblast-derived BeWo cell line. Reprod Biol Endocrinol 2009; 62. Yazama H, Kitatani K, Fujiwara K, Kato M, Hashimoto-Nishimura M,
7(1):51. Kawamoto K, Hasegawa K, Kitano H, Bielawska A, Bielawski J, Okazaki
Obesity alters early placenta gene expression, 2018, Vol. 98, No. 6 809

T. Dietary glucosylceramides suppress tumor growth in a mouse xenograft altered in pathologies of the human placenta: gestational diabetes mellitus,
model of head and neck squamous cell carcinoma by the inhibition of intrauterine growth restriction and preeclampsia. Placenta 2010; 31(3):
angiogenesis through an increase in ceramide. Int J Clin Oncol 2015; 222–229.
20(3):438–446. 80. Ho TC, Chen SL, Yang YC, Liao CL, Cheng HC, Tsao YP. PEDF in-
63. Melland-Smith M, Ermini L, Chauvin S, Craig-Barnes H, Tagliaferro A, duces p53-mediated apoptosis through PPAR gamma signaling in human
Todros T, Post M, Caniggia I. Disruption of sphingolipid metabolism umbilical vein endothelial cells. Cardiovasc Res 2007; 76(2):213–223.
augments ceramide-induced autophagy in preeclampsia. Autophagy 2015; 81. Loegl J, Nussbaumer E, Hiden U, Majali-Martinez A, Ghaffari-Tabrizi-
11(4):653–669. Wizy N, Cvitic S, Lang I, Desoye G, Huppertz B. Pigment epithelium-
64. Koeberle A, Pergola C, Shindou H, Koeberle SC, Shimizu T, Laufer derived factor (PEDF): a novel trophoblast-derived factor limiting feto-
SA, Werz O. Role of p38 mitogen-activated protein kinase in linking placental angiogenesis in late pregnancy. Angiogenesis 2016; 19(3):373–

Downloaded from https://academic.oup.com/biolreprod/article-abstract/98/6/795/4816726 by Universidad ICESI user on 05 March 2020


stearoyl-CoA desaturase-1 activity with endoplasmic reticulum homeosta- 388.
sis. FASEB J 2015; 29(6):2439–2449. 82. Adya R, Tan BK, Randeva HS, Differential effects of leptin and
65. Zarubin T, Han J. Activation and signaling of the p38 MAP kinase path- adiponectin in endothelial angiogenesis. J Diab Res 2015; 2015:1–12.
way. Cell Res 2005; 15(1):11–18. 83. Gomez de Aguero M, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura
66. Spange S, Wagner T, Heinzel T, Krämer OH. Acetylation of non-histone Y, Li H, Steinert A, Heikenwalder M, Hapfelmeier S, Sauer U, McCoy KD,
proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Macpherson AJ. The maternal microbiota drives early postnatal innate
Biol 2009; 41(1):185–198. immune development. Science 2016; 351(6279):1296–1302.
67. Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, 84. Boulange CL, Neves AL, Chilloux J, Nicholson JK, Dumas ME. Impact
Olsen JV, Mann M. Lysine acetylation targets protein complexes and of the gut microbiota on inflammation, obesity, and metabolic disease.
co-regulates major cellular functions. Science 2009; 325(5942):834–840. Genome Med 2016; 8(1):42.
68. Croze ML, Soulage CO. Potential role and therapeutic interests of myo- 85. Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu
inositol in metabolic diseases. Biochimie 2013; 95(10):1811–1827. Z, Maher L, Redinbo MR, Phillips RS, Fleet JC, Kortagere S et al. Sym-
69. Ferrari F, Facchinetti F, Ontiveros AE, Roberts RP, Saade MM, Blackwell biotic bacterial metabolites regulate gastrointestinal barrier function via
SC, Sibai BM, Refuerzo JS, Longo M. The effect of combined inositol the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 2014;
supplementation on maternal metabolic profile in pregnancies compli- 41(2):296–310.
cated by metabolic syndrome and obesity. Am J Obstet Gynecol 2016; 86. Flanagan DE, Moore VM, Godsland IF, Cockington RA, Robinson JS,
215(4):503.e1–503.e8. Phillips DI. Fetal growth and the physiological control of glucose tolerance
70. Nascimento NRF, Lessa LM, Kerntopf MR, Sousa CM, Alves RS, Queiroz in adults: a minimal model analysis. Am J Physiol Endocrinol Metab 2000;
MG, Price J, Heimark DB, Larner J, Du X, Brownlee M, Gow A et al. 278(4):E700–E706.
Inositols prevent and reverse endothelial dysfunction in diabetic rat and 87. De Blasio MJ, Dodic M, Jefferies AJ, Moritz KM, Wintour EM, Owens
rabbit vasculature metabolically and by scavenging superoxide. Proc Natl JA. Maternal exposure to dexamethasone or cortisol in early pregnancy
Acad Sci USA 2006; 103(1):218–223. differentially alters insulin secretion and glucose homeostasis in adult male
71. Challier JC, Basu S, Bintein T, Minium J, Hotmire K, Catalano PM, sheep offspring. Am J Physiol Endocrinol Metab 2007; 293(1):E75–E82.
Hauguel-de Mouzon S. Obesity in pregnancy stimulates macrophage ac- 88. Gabory A, Ferry L, Fajardy I, Jouneau L, Gothié JD, Vigé A, Fleur C,
cumulation and inflammation in the placenta. Placenta 2008; 29(3):274– Mayeur S, Gallou-Kabani C, Gross MS, Attig L, Vambergue A et al.
281. Maternal diets trigger sex-specific divergent trajectories of gene expression
72. Saben J, Lindsey F, Zhong Y, Thakali K, Badger TM, Andres A, Gomez- and epigenetic systems in mouse placenta. PLoS One 2012; 7(11):e47986.
Acevedo H, Shankar K. Maternal obesity is associated with a lipotoxic 89. Prickett TD, Samuels Y. Molecular pathways: dysregulated glutamatergic
placental environment. Placenta 2014; 35(3):171–177. signaling pathways in cancer. Clin Cancer Res 2012; 18(16):4240–4246.
73. Rocha NG, Templeton DL, Greiner JJ, Stauffer BL, DeSouza CA. 90. Wen Y, Li J, Koo J, Shin SS, Lin Y, Jeong BS, Mehnert JM, Chen S,
Metabolic syndrome and endothelin-1 mediated vasoconstrictor tone in Cohen-Sola KA, Goydos JS. Activation of the glutamate receptor GRM1
overweight/obese adults. Metabolism 2014; 63(7):951–956. enhances angiogenic signaling to drive melanoma progression. Cancer Res
74. Tesauro M, Schinzari F, Rovella V, Di Daniele N, Lauro D, Mores N, 2014; 74(9):2499–2509.
Veneziani A, Cardillo C. Ghrelin restores the endothelin 1/nitric oxide 91. Pan YB, Gong Y, Ruan HF, Pan LY, Wu XK, Tang C, Wang CJ, Zhu
balance in patients with obesity-related metabolic syndrome. Hyperten- HB, Zhang ZM, Tang LF, Zou CC, Wang HB et al. Sonic hedgehog
sion 2009; 54(5):995–1000. through Gli2 and Gli3 is required for the proper development of placental
75. Spradley FT, Palei AC, Granger JP. Increased risk for the develop- labyrinth. Cell Death Dis 2015; 6(2):e1653.
ment of preeclampsia in obese pregnancies: weighing in on the mech- 92. Pola R, Ling LE, Silver M, Corbley MJ, Kearney M, Blake Pepinsky R,
anisms. Am J Physiol Regul Integr Comp Physiol 2015; 309(11): Shapiro R, Taylor FR, Baker DP, Asahara T, Isner JM. The morphogen
R1326–R1343. Sonic hedgehog is an indirect angiogenic agent upregulating two families
76. Fiore G, Capasso A. Effects of vitamin E and C on placental oxidative of angiogenic growth factors. Nat Med 2001; 7(6):706–711.
stress: an in vitro evidence for the potential therapeutic or prophylactic 93. Dhobale M. Neurotrophins: Role in adverse pregnancy outcome. Int J
treatment of preeclampsia. Med Chem 2008; 4(6):526–530. Dev Neurosci 2014; 37:8–14.
77. Liang C, DeCourcy K, Prater MR. High–saturated-fat diet induces gesta- 94. D’Souza V, Patil V, Pisal H, Randhir K, Joshi A, Mehendale S, Wagh
tional diabetes and placental vasculopathy in C57BL/6 mice. Metabolism G, Gupte S, Joshi S. Levels of brain derived neurotrophic factors across
2010; 59(7):943–950. gestation in women with preeclampsia. Int J Dev Neurosci 2014; 37:36–
78. Fournier T, Tsatsaris V, Handschuh K, Evain-Brion D. PPARs and the 40.
placenta. Placenta 2007; 28(2-3):65–76. 95. Prince CS, Maloyan A, Myatt L. Maternal obesity alters brain derived neu-
79. Holdsworth-Carson SJ, Lim R, Mitton A, Whitehead C, Rice GE, Per- rotrophic factor (BDNF) signaling in the placenta in a sexually dimorphic
mezel M, Lappas M. Peroxisome proliferator-activated receptors are manner. Placenta 2017; 49:55–63.

You might also like