ArticlePDF AvailableLiterature Review

Gene expression in the placenta: Maternal stress and epigenetic responses

Authors:

Abstract and Figures

Successful placental development is crucial for optimal growth, development, maturation and survival of the embryo/fetus into adulthood. Numerous epidemiologic and experimental studies have demonstrated the profound influence of intrauterine environment on life, and the diseases to which one is subject as an adult. For the most part, these invidious influences, whether maternal hypoxia, protein or caloric deficiency or excess, and others, represent types of maternal stress. In the present review, we examine certain aspects of gene expression in the placenta as a consequence of maternal stressors. To examine these issues in a controlled manner, and in a species in which the genome has been sequenced, most of these reported studies have been performed in the mouse. Although each individual maternal stress is characterized by up- or down-regulation of specific genes in the placenta, functional analysis reveals some patterns of gene expression common to the several forms of stress. Of critical importance, these genes include those involved in DNA methylation and histone modification, cell cycle regulation, and related global pathways of great relevance to epigenesis and the developmental origins of adult health and disease.
Content may be subject to copyright.
Gene Expression in the Placenta:
Maternal Stress and Epigenetic Responses
Ciprian P. Gheorghe, Ravi Goyal, Ashwani Mittal, and Lawrence D. Longo
Center for Perinatal Biology, Departments of Physiology and Obstetrics and Gynecology, Loma
Linda University School of Medicine, Loma Linda, CA 92350
Keywords
placenta; gene regulation; microarray; DNA methylation; epigenetics
Introduction
In the Western world, cardiovascular disease, along with diabetes mellitus and its
complications, are leading causes of death. A number of important risk factors have been
associated with the virtual pandemic of these killers. These include smoking, sedentary
lifestyle, high body mass index, hypertension, and so forth. Nonetheless, many individuals
who develop cardiovascular and/or metabolic disease do not have these risk factors. Thus, it
is clear that as yet unrecognized and underappreciated factors must be considered in the genesis
of these disorders. In his monumental volume Stress, Hans Selye (1907-1982) observed that
stress to the organism, in essentially any of its forms -- dietary, environmental, disease, and
others -- could result in cellular, hormonal, and related damage, with the body mounting a
response he termed the “General-Adaptation-Syndrome” (Selye, 1950). Writing years before
the nuances of biochemical and molecular mechanisms were established, Selye envisioned an
orchestrated biological defensive response to such challenges. This concept is of special
relevance to the developing fetus, as during the course of gestation a number of stresses to the
mother are known to affect placental, as well as embryonic/fetal development, many with life-
long consequences.
Along this line, a factor that has received increasing attention is the idea of “programming”
during fetal life, often as a consequence of maternal stress. Special features of antenatal
programming include: critical periods of vulnerability, failure or unsatisfactory completion of
specific developmental milestones, association with functional defects, the permanent nature
of such sequelae, and so forth (Barker, 1989a; 1989b; 1992; 1994; 1995a; 1995b; 1998;
2004; Barker et al, 1995; Nijland et al, 2008). The concept of the developmental origins of
adult health and disease, first articulated by David J.P. Barker in the mid-1980s, “... suggested
that poor nutrition in early life increases susceptibility to the effects of an affluent diet” (Barker
& Osmond, 1986a). In a related analysis, these authors noted the high correlation of
cerebrovascular accidents in the 1970s, to increased infant mortality six decades earlier during
the years 1911-1914 (Barker & Osmond, 1987). In addition, in men born from 1911 to 1930,
Barker and his group have shown an inverse correlation between the weights both at birth and
Send Correspondence to: Lawrence D. Longo, M.D., FACOG, FRCOG, Director, Center for Perinatal Biology, Loma Linda University,
School of Medicine, Loma Linda, CA 92350, E-mail: llongo@llu.edu, Tel: 909 558 4325.
Additional Authors: Ciprian P. Gheorghe: cgheorghe@llu.edu
Ravi Goyal: rgoyal@llu.edu
Ashwani Mittal: asmittal@llu.edu
NIH Public Access
Author Manuscript
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
Published in final edited form as:
Int J Dev Biol. 2010 ; 54(2-3): 507–523. doi:10.1387/ijdb.082770cg.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
at 1 year of age to coronary artery disease as adults (Barker et al, 1989a; 1989b). A subsequent
study disclosed a similar trend with birthweight among women (Osmond et al, 1993).
Since this concept first was proposed, supporting evidence has been provided by a series of
epidemiologic studies from a number of countries and cultures. These include studies
correlating adult mortality from acute myocardial infarction with high infant mortality rates in
a given population, follow-up studies correlating adult hypertension, coronary artery disease,
and type II diabetes with low birth weight, the relation of increased mortality from coronary
artery disease to low weight at 1 yr of age, and the relation of both newborn ponderal index
[weight (g) x 102/crown-heel length (cm)2] and placental-to-fetal weight ratio to hypertension
in the adult (see Barker, 2003, and Barker et al, 1995 for review). These associations are
independent of adult life style risk factors (Barker et al, 1993). Among a number of other
maternal stress-induced sequelae are those of immune dysfunction (Götz et al, 2007; Merlot
et al, 2008), cortisol secretion later in life (Reynolds et al, 2007; Tu et al, 2007), increased
incidence of schizophrenia (Hoek et al, 1996; Hulshoff et al, 2000; Susser & Lin, 1992), and
many others (Ham & Tronick, 2006). More recently, numerous studies in experimental animals
have demonstrated a relation between intrauterine fetal stress, particularly that of maternal food
deprivation and/or emotional stress, and adult disease (Gluckman et al, 2008; Green & Hanson,
2004; Hanson & Gluckman, 2005; Jansson & Powell, 2007). Among the major known
intrauterine stresses about which the effects on subsequent adult health are largely unknown,
are maternal hypoxia and dietary imbalance.
The placenta, a fetomaternal organ joining mother and offspring during pregnancy in mammals,
serves as an endocrine organ in the “maternal-placental-fetal” complex, in addition to its role
in the exchange of respiratory gases, a multitude of nutrients, an immunologic barrier, and
other functions. As has been recognized for many years, compromised placental function can
have both short- and long-term consequences for the developing conceptus. In the present
review, we examine the current state of knowledge of placental gene expression responses to
maternal stress such as hypoxia, protein deficiency, and caloric excess. For the most part these
studies are in rodents, however, when applicable, we also review those studies of placental
gene expression in the human. (We will not review thoroughly the field of antenatal origins of
adult health and disease, nor the role of environmental toxins in developmental disorders, as
these topics have been reviewed in extenso elsewhere). Importantly, beyond mere description,
we attempt to place these gene expression changes into a framework of the biochemical
pathways and molecular mechanisms, by which stresses to the maternal organism can result
in alterations of great biologic and epigenetic importance to the developing embryo and fetus.
Finally, we consider important issues for future investigation, i.e., questions that probe the
limits of our understanding.
Why Study Gene Expression in the Placenta?
Successful placental development is crucial for optimal growth, maturation, and survival of
the embryo/fetus. The placenta not only nurtures the fetus, but protects it from harmful waste
products by acting as an excretory route, and also presents an immunologic barrier between
the maternal and fetal circulatory beds. Although the nucleus of every cell in the body carries
a complete set of DNA, these cells differ in function with placental and embryological
development consisting of an elegantly orchestrated switching of genes on and off in the
transition from single fertilized cell to fully formed placenta and fetus. Deviation in the normal
gene expression pattern may lead to altered placental phenotype, as well as a modified
phenotype of the conceptus. This is evidenced by the numerous lethal embryonic null mutants
secondary to placental failure. The mouse has been employed as a useful model of placental
development. While the mouse placenta is not identical to its human counterpart, many studies
have shown that similar cell lineages are largely conserved, and similar genes direct placental
Gheorghe et al. Page 2
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
development in both species. Placental cell lineages derive from trophoectoderm precursors.
The mural trophoectoderm differentiates into primary trophoblast giant cells, while the polar
trophoectoderm gives rise to the extraembryonic ectoderm and the ectoplacental cone. In many
mammals, including the mouse, the extraembryonic ectoderm forms the chorion that fuses with
the allantois, an outgrowth of extraembryonic mesoderm, at around embryonic day 8 (E8) to
form the placental labyrinthine layer. The spongiotrophoblast layer of the murine placenta
derives from ectoplacental precursor cells and forms the middle layer of the placenta, also
known as the junctional zone. The outermost placental cells are the trophoblast giant cell layer.
In addition to the primary trophoblast cells derived from the mural trophoectoderm, secondary
trophoblast giant cells are derived from the spongiotrophoblast. Later in placental development,
around E12.5, glycogen-filled trophoblast cells appear in the spongiotrophoblast layer.
Although their function is unclear, these cells express several important gene products, and
migrate into the decidua later in pregnancy. Several reviews have detailed placental cell
lineages, and some of the genes involved in their differentiation (Cross, 2005; Simmons &
Cross, 2005).
Recent studies reveal some of the fundamental mechanisms underlying placental development
(Cross et al, 2003; Daoud et al, 2005; Gheorghe et al, 2006; Hemberger, 2007; Sood et al,
2006; Tanaka et al, 2000). Numerous genes are required for proper development of the
placenta, and their number has increased greatly, in part, due to the discovery of numerous
lethal embryonic null mutants secondary to placental failure (Adams et al, 2000; Schorpp-
Kistner et al, 1999; Schreiber et al, 2000; Yamamoto et al, 1998). For example, the disruption
of many genes, including growth factors, transcription factors, extracellular matrix proteins,
and proteins involved in cell signaling, leads to embryonic lethality secondary to placental
failure (Rossant & Cross, 2001). In human trophoblast in vitro, several gene classes are strongly
up- and down-regulated in the course of differentiation (Aronow et al, 2001). Another study
compared differentially expressed genes between the murine placenta and the embryo itself at
E12.5 (Tanaka et al, 2000). Microarray analysis has provided insights into aspects of the genetic
mechanisms of development, cell growth both normal and abnormal, responses to stress, and
numerous other processes (Chu et al, 1998; Gasa et al, 2004; Iyer et al, 1999).
To What Extent is Placental Gene Expression Altered During Gestation?
The molecular basis of placental development remains incompletely understood. Recent
studies have begun to shed some light on this process, and numerous genes have been
demonstrated to be essential for the proper differentiation of placental cell lineages and fetal
survival. Unfortunately, the details of the interactions and effects of these genes are unclear.
In an effort to understand this process at a more fundamental level, we examined gene
expression patterns in the developing murine placenta at days E10.5 E12.5, E15.5, and E17.5,
testing the hypothesis that from E10.5 until E17.5, numerous placental genes are up- or down-
regulated to a significant degree, and that specific functional groups of genes are regulated at
the different developmental ages (Gheorghe et al, 2006). To examine gene clustering and
functional analysis of pathways, we focused on those genes most highly regulated by
development. At E10.5, several functional categories were over-represented, including genes
involved in angiogenesis and blood vessel development, morphogenesis, and organogenesis,
and genes involved in lipid metabolism and transport. At E12.5, over-represented gene
categories were involved in cell cycle control and RNA binding proteins. At E15.5, notably
over-represented were genes involved in cellular transport and cell growth and maintenance.
At E17.5, we noted the up-regulation of an over-abundance of genes involved in the regulation
of transcription and numerous proteins that localize to the nucleus.
This study identified several subsets of genes highly regulated during placental development.
Clustering according to their expression patterns, suggests that at crucial times during placental
Gheorghe et al. Page 3
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
ontogeny particular subsets of diverse genes are induced or repressed in concert (Fig. 1). Genes
up-regulated early in placental development clearly underlie the rapid tissue growth, cell
proliferation, and vascular development occurring during this period. At E10.5, genes involved
in several key processes were strongly up-regulated. These include angiogenesis, lipid
metabolism, and cell cycle regulation. Genes such as ELK3, c-fos induced growth factor,
plasminogen (the precursor to angiostatin), serine (or cysteine) proteinase inhibitor clade F
member 1, all are involved in blood vessel morphogenesis, and were up-regulated at E10.5. At
both E10.5 and E12.5 cyclin, D1, cyclin E2, cyclin C, MAD 2, pleiotrophin, BRCA 2, which
are involved in cell cycle control also are up-regulated strongly. At E12.5, ribosomal genes
were notably up-regulated, as were several genes involved in lipid transport and metabolism
including: apolipoprotein B, apolipoprotein C-II, lysophospholipase, microsomal triglyceride
transfer protein and adiponectin receptor 1. As must be evident, lipid transport and metabolism
are important for the proper fetal development (Shekhawat et al, 2003) and disruption of lipid
transporters leads to embryonic lethality (Farese et al, 1996;Gimeno et al, 2003). Previous null
mutant experiments have identified several of these genes as embryonic lethal, further
confirming their importance in development. For instance, Cops2 mutants died soon after
implantation (Lykke-Anderson et al, 2003), Pten mutants died at E9.5 secondary to placental
failure (Yamamoto et al, 1998), and connexin 43 mutants died shortly after birth, due to cardiac
and vascular abnormalities (Reaume et al, 1995).
Genes such as growth hormone releasing hormone prolactin-like protein I, secretin, and
chorionic somatomammotropin hormone 2 also were upregulated during the course of placental
development. Recent studies in the sheep suggest that growth hormone releasing hormone
regulates the expression of both placental growth hormone and lactogen (Lacroix et al,
2002). Insulin-like growth factor II (IGF-II) and Insulin-like growth factor binding protein 2,
genes have been shown to be expressed in the placenta (Zollers et al, 2001), were up-regulated
from E10.5 to E12.5. Previous studies have shown that after E12.5 IGF-II is mainly produced
by trophoblast glycogen cells (Redline et al, 1993); however, in our study it was up-regulated
at E12.5 and later (Gheorghe et al, 2006). IGF-II also appears to have key functions in placental
transport and permeability (Sibley et al, 2004). A number of prolactin-like proteins have been
shown to be regulated with development, such as: prolactin-like protein C 1, prolactin-like
protein F, prolactin-like protein I, prolactin-like protein K. The prolactin gene family in the
mouse has at least 26 identified members (Wiemers et al, 2003), and several studies have shown
that in the placenta this gene family performs key reproductive and regulatory functions (Ain
et al, 2003; 2004). In the near-term human placenta, mRNA for a number of factors associated
with angiogenesis (vascular endothelial growth factor and annexin V) and homeostasis
(plasminogen activator factor, thrombomodulin, and others) are widely distributed (Chinni et
al, 2008). Circulating fetal fibrocytes, and perhaps other cells, also play a role in the
development of the placenta and the umbilical arteries and vein (Kim et al, 2008).
To What Extent is Placental Gene Expression Altered by Maternal Hypoxia?
As noted above, a number of stressors can lead to altered placental and fetal growth and
development. Of great importance in this regard, is the less than optimal supply of oxygen
(O2), e.g., hypoxia. Hypoxia has been identified as a major stressor in development, and is
believed to be a contributing cause to placental pathology such as that associated with
preeclampsia (Austgulen et al, 2004; Challier & Uzan, 2003). Hypoxia can lead to low birth
weight and intrauterine growth restriction (IUGR) and disease of the newborn such as persistent
pulmonary hypertension (Zamudio, 2003). Little is known, however, about the adaptive
mechanisms involved in the placental responses to suboptimal oxygen availability. Several
studies have attempted to harness the power of microarray and proteomic analysis to elucidate
responses to hypoxia in cultured human cytotrophoblasts (Hoang et al, 2001), and in rat
embryos and placentas (Huang et al, 2004). As with other cell types, oxygen is a critical
Gheorghe et al. Page 4
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
regulator of the normal trophoblast cell development, which undergo differentiation and/or
proliferation in response to varying O2 concentrations. Acting through aryl receptor nuclear
transporter and hypoxia-inducible factor 1α (HIF-1α), oxygen regulates placental cell
phenotypes and gene expression (Adelman et al, 2000). Hypoxic stress can lead to placental
cell death and dysregulation of vasculogenesis, which negatively affects the development of
the placental vascular bed (Kingdom et al, 2000). In response to exposure to high altitude,
long-term hypoxia, the ovine placentomes undergo significant structural changes (Penninga &
Longo, 1998), and the vasculature displays significant increases in capillary density, vessel
tortuosity, and a decrease in diffusion distance from maternal to fetal blood (Krebs et al,
1997). The human placenta also shows significant morphologic and morphometric changes in
response to high altitude hypoxia (Zhang et al, 2002). Nonetheless, essentially nothing is
known about the molecular basis of these changes.
To understand in greater detail the role of hypoxia in placental gene expression, we tested the
hypothesis that hypoxia-induced altered placental morphology is accompanied by significant
changes in expression profiles. We compared gene expression levels between the normal
murine placenta at E17.5, and that from dams exposed to 0.5 atmosphere hypoxia (10.5%
O2) for 48 hours from E15.5 to E17.5. In response to this stress, some of the most highly up-
regulated genes were those related to metabolism (alpha-keto reductase family-1 member 7,
mitochondrial solute carrying protein, acetyl-coenzyme A synthetase 2, and NADPH oxidase
4), oxygen transport (erythroid associated factor, hemoglobin Y beta-like embryonic chain,
erythrocyte protein band 4.2), proteolysis (cathepsin G, kallikrein 4, dipeptidase 1, serine
protease 32), cell death (Bcl-like 2, perforin 1, glutathione peroxidase 1), and metabolism of
reactive oxygen species (Gheorghe et al, 2007). Of particular note, several genes related to
DNA methylation and epigenetic control were up-regulated (DNA methyltransferase 3B,
methyl-CpG binding domain protein 1, RNA binding motif protein 3). Of the chromosomal
distribution of genes up-regulated by hypoxia, we noted an over-abundance of genes from
chromosome 14 (9.5% of the regulated genes as compared to 3.5% of genes on the array).
Many of these correspond to the granzyme family of proteases.
Several of these genes have been noted previously to be regulated by hypoxia and/or involved
in angiogenesis and metabolic responses. For instance, NAPDH oxidase 4 has been identified
as a potential O2 sensor and regulator of HIF-1α (Zhu et al, 2002). Ferrocheloelastase is
involved in heme metabolism, and has been shown to be up-regulated by hypoxia (Liu et al,
2004). Aminolevulinic acid synthase 2, glutathione peroxidase 1, and peroxiredoxin 2 are
involved in the metabolism of reactive oxygen species; their activity, and expression levels
have been identified as regulated by hypoxia (Abu-Farha et al, 2005; Mysore, 2005).
Glycophorin A is involved in erythroid differentiation, and is regulated by erythropoietin
(Gubin et al, 1999). Lactotransferrin is an antioxidant involved in iron metabolism and in
scavenging of free radicals under hypoxic conditions (Morris et al, 1995). As noted above,
hypoxia upregulated several members of the granzyme gene family. These are serine proteases
expressed by lymphocytes, and thought to be involved in T-cell-mediated cytotoxicity.
Granzymes are released along with perforin by natural killer (NK) cells and cytotoxic T
lymphocytes, and trigger apoptosis in target cells through several mechanisms. A number of
studies have demonstrated placental expression of granzymes, and they may play a broader
role in placental development (Allen et al, 1998; Hirst et al, 2001). We also observed an up-
regulation of perforin, which suggests that in response to hypoxia a greater number of NK cells,
which have been shown to mediate a number of important functions, invade the placenta
(Parham, 2004). Thus, the data suggest that granzymes not only play a role in normal placental
development, but also are involved in hypoxia-mediated responses.
Again, of particular note in our study was the significant up-regulation of genes involved in
DNA methylation and epigenetic control: DNA methyltransferase 3b and methyl CpG binding
Gheorghe et al. Page 5
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
domain protein 1 (Gheorghe et al, 2007). As noted below, epigenetic modification is an
important mechanism of gene expression regulation that does not involve modification of the
DNA sequence, but rather DNA methylation and histone modification. These changes in
expression patterns may be of importance in development, genomic imprinting, and the
development of cancer (Turek-Plewa & Jagodzinski, 2005). The generation of reactive oxygen
species (ROS) and the manipulation of glutathione metabolism have been shown to regulate a
number of cellular processes, including DNA methylation (Fratelli et al, 2005). As depicted
in Fig. 2, this suggests complex genetic regulation that commences with hypoxia and leads to
alternations that result in long-term programming. In turn, this activates the DNA methylation
machinery, and ultimately leads to long-term changes in the organism unrelated to modification
of the nucleotide sequence. A key to unraveling this mechanism will be the identification of
the targets for altered methylation and subsequent long-term down-regulation of transcription.
Among the down-regulated genes, most notable were several transcription factors
(transformation-related protein 63, doublesex- and mab-3-related transcription factor 1, glial
cells missing homolog 1, zinc finger, imprinted 1), cell cycle (cyclin M2), cell structure (keratin
complex 2 basic, gene 8, procollagen C-proteinase enhancer protein, fibromodulin). We have
presented a list of the genes up- or down-regulated and their known function, and verified the
regulation of several of these genes using real time PCR (Gheorghe et al, 2007). In response
to hypoxia, placental morphology also was altered significantly e.g. having a greater vascular
density and containing many more red blood cells.
Several other studies have examined gene expression changes in response to hypoxia at the
global level. One study catalogued the hypoxic-induced responses in the rat embryo to hypoxic
exposure for both 24 hours and 11 days. Glycolysis-related genes, calcium homeostasis-related
genes, and inflammatory genes (particularly as related to oxidative stress) were up-regulated,
while cell growth-related genes were down-regulated (Huang et al2004). Other studies also
have examined human trophoblast responses in-vitro to low oxygen tension. Observed were
up-regulation of antioxidants (superoxide dismutase) and glycolysis-related genes, and the
upregulation of glutathione-S-transferase (Nelson et al, 2003; Roh et al, 2005). In addition,
gene expression changes in placentas from pregnancies complicated by pre-eclampsia and
IUGR have been catalogued. In particular, H4 histone was down-regulated in women with
severe pre-eclampsia (Chen et al, 2006; Soleymanloo et al, 2005), and up-regulation of
glutathione-S-transferase was observed in human placentas from women at high altitude
(3,100m) (Chen et al, 2006; Roh et al, 2005). These studies have highlighted the diverse manner
in which placental cells respond to hypoxic stress.
In the placenta of patients that experienced chronic hypoxic ischemia, mRNA levels of both
leptin and insulin-like growth factor (IGF)-1 were upregulated significantly (Trollmann et al,
2007). In near-term placental explants cultured in 1% O2, expression of the tumor suppressor
protein p53, that promotes cell cycle arrest or apoptosis, was significantly elevated (Heazell
et al, 2008). Exposure of human placental villous explants to 3% O2 for 48 h, resulted in
significant increase in endoglin, a co-receptor for transforming growth factor (TGF-β3)
pathway (Yinon et al, 2008). In the placentae of patients with severe preeclampsia at 31±2
weeks gestation, mRNA of the antioxidant protein glutathione reductase was reduced
significantly, while that for thioredoxin peroxidase was increased (Vanderlelie et al, 2008).
This suggests that oxidative stress may play a key role in the pathophysiology of the placentae
in cases of preeclampsia.
In summary, the murine placenta appears to respond to hypoxia through several adaptive
mechanisms. These include up-regulation of genes associated with erythropoiesis, increases
in heme and iron metabolism, and in genes involved in proteolysis and peptidolysis. These
varied responses suggest that the placenta responds by increasing its oxygen carrying capacity,
Gheorghe et al. Page 6
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
increasing metabolic and antioxidant responses, and initiating tissue growth, turnover, and
remodeling. Studies in both the human (Roh et al, 2005) and sheep (Krebs et al, 1997; Penninga
& Longo, 1998) have demonstrated that the placenta undergoes multiple morphological and
genetic changes in response to prolonged hypoxia. Hypoxic insults secondary to preeclampsia,
maternal smoking, or exposure to high altitude can contribute to placental insufficiency and
may lead to intrauterine growth restriction (Jones & Fox, 1980; Levi & Nelson, 2000;
Reshetnikova et al, 1994; Spira et al, 1997). The exact mechanisms of these changes are not
understood. Extracellular matrix remodeling, the modulation of apoptosis, altered cellular
metabolism, and epigenetic changes all appear to be crucial steps in the physiological
adaptations of the placenta to hypoxia. It is hoped that these, and other studies, will provide
insights at the molecular level into these mechanisms and important clinical problems.
To What Extent is Placental Gene Expression Altered by Maternal Protein Restriction?
Several lines of evidence demonstrate that nutritional deprivation of the pregnant mother may
have deleterious consequences for the progeny. For instance, a shocking “experiment” in
humans was that during World War II of the “Hunger Winter” in Amsterdam and Western
Holland from November 1944 until the Allied victory in May 1945. This tragedy provides
useful lessons on the effects of caloric restriction/malnutrition on fetal development and disease
prevalence in adulthood. During this seven month period, the caloric ration fell from 2400
to 400-800 calories per day, less than 25% of the recommended intake for adults. Although
children, and to some extent pregnant and lactating women, received extra rations during the
early part of this disastrous famine, they too suffered severe dietary deficiency (Roseboom et
al, 2001a). In essence, upon reaching adulthood, the infants that were small at birth had
significantly greater prevalence of cardiovascular disease, type II diabetes (Kyle & Prichard,
2006; Painter et al, 2005a; Roseboom et al, 2001a; 2001b; Stein & Susser, 1975; Stein et al,
2004), and mood and personality disorders (Godfrey, 1998). Those fetuses exposed to maternal
caloric restriction in mid-gestation had a much greater incidence of pulmonary disease,
including bronchitis (Lopuhaa et al, 2000), and renal disease as evidenced by microalbuminuria
(Painter et al, 2005b). Females who were conceived during the famine also had a much higher
prevalence of obesity as adults (Ravelli et al, 1999), and both males and females showed
atherogenic lipid profiles (Roseboom et al, 2000). Concommently during WWII, the people
of St. Petersburg and surrounding area of Russia were subjected to severe dietary restrictions
due to interdiction of food supplies by the German army. The children born under these
conditions were not only small for gestational age, but also developed health problems later in
life (Neugebauer et al, 1999). However, records of the long-term sequelae of these individuals
are not as clear as those in Holland (Lind, 1984; Ravelli et al, 1976). Importantly, the
mechanisms of these in utero “programming” effects are unknown.
Epidemiologic data on the role of maternal nutrition in determining the long-term health of
offspring derives largely from the studies of Barker and colleagues (Barker, 1995b; 2003;
Barker & Clark, 1997; Barker & Osmond, 1986a; 1986b). Studies in several countries have
correlated maternal dietary deficiencies that result in the newborn infant being small for
gestational age, or growth restricted, with the prevalence of cardiovascular disease (Barker &
Osmond, 1986b; Barker et al, 1989a), type 2 diabetes (Hales et al, 1991; Ravelli et al, 1998),
and numerous other conditions in the adult. Maternal nutritional deprivation may influence
placental growth and morphology, alter the hormonal milieu of the developing fetus, and cause
subsequent cardiovascular, hormonal and behavioral consequences in the adult (Barker,
1992; 1994; 2003; Barker & Osmond, 1986a; 1986b; Barker et al, 1989; Gluckman et al,
2008).
The epidemiologic observations made in human subjects have been confirmed in animal
models, and have led to speculation regarding the cellular mechanisms of changes in the
Gheorghe et al. Page 7
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
placenta, and their effects on the developing fetus (Armitage et al, 2004; Hoet & Hanson,
1999). An important question is the extent to which these observed effects result from an overall
caloric restriction, as opposed to a qualitative component in the diet that triggers the responses.
Evidence from animal studies points to protein deprivation as a major factor in these defects.
For example in the rat, the growth reducing effects of a low calorie diet can be reversed only
by a dietary increase in protein levels, while vitamin supplements and caloric increases through
carbohydrates did not reverse the effects observed (Hsueh et al, 1967). Other studies have
revealed that dietary amino acid balance is a key mediator of some of the cardiovascular and
metabolic effects observed in response to protein deprivation (Boujendar, 2003). Overall, the
several studies indicate that nutritional deprivation, and protein restriction in particular, can
have immediate deleterious effects on the placenta and the fetus, and may result in long-term
sequelae that extend into adulthood.
In a recent study in the mouse, we tested the hypothesis that moderate maternal protein
deprivation would alter gene expression patterns in the placenta. We compared gene expression
levels between normal placentas at E17.5, and those from pregnancies in which the mothers
were exposed to seven days of protein deprivation (i.e., 10% protein by weight versus the 20%
of normal chow) from E10.5 to E17.5. Of particular note, a number of genes involved in the
p53 oncogene pathway were up-regulated. In addition to p53 itself, its positive regulators Zmis,
Jmy, and Hipk2, as well as genes activated by p53 (Inpp5d, Cebpa), were induced (Figure 3A).
These p53 pathway proteins are important regulators of cell growth and proliferation. This
pathway serves as a G1 checkpoint, and arrests growth and/or induces apoptosis in response
to cellular damage. Mutations in the p53 gene have been implicated in a number of cancers
and other pathological processes (Ryan et al, 2001). Hipk2, an upstream regulator of p53,
activates its transcriptional activity and pro-apoptotic activities through phosphorylation at Ser
46 (Hoffman et al, 2002). Cebpa, is a transcription factor induced by p53, and mediates some
of the downstream effects of p53 activation (Yoon & Smart, 2004). Among the gene ontology
classes most over-represented in the up-regulated group, we noted the mitogen-activated
protein kinase pathway, regulators of apoptosis (Bcl2-like 2, p53, endophilin, Fas-activated
serine/threonine kinase), negative regulators of cell growth (farnesyltransferase CAAX box
beta, cadherin 5, CCAAT/enhancer binding protein (C/EBP) alpha, inositol polyphosphate-5-
phosphatase D, p53), and negative regulators of cellular metabolism (nuclear receptor co-
repressor 2, histone deacetylase 7A, SPEN homolog, transcriptional regulator). Acting in
concert, activation of these genes could result in growth restriction during pregnancy (Fig. 3B).
Among down-regulated genes, particularly striking were those related to nucleotide
metabolism. For selected genes, we confirmed these results using qRT-PCR.
Another potentially important finding, is that protein deprivation altered the expression of
several genes involved in DNA methylation, histone acetylation, and epigenetic regulation of
gene expression. The expression levels of histone deacetylase 7A and methionine
adenosyltransferase II, alpha were elevated several fold. Histone acetylation triggers changes
in chromatin structure, and regulates transcriptional availability of genes. In turn, histone
deacetylation increases histone affinity for DNA, thereby repressing transcription (Bulger,
2005). Methionine adenosyltransferase II alpha synthesizes AdoMet the direct precursor used
for DNA methylation by methyltransferases (Mao et al, 1998). Histone 2 (h3c2) is down-
regulated, along with Mcm6 and telomeric repeat binding factor 1. These proteins contribute
to DNA replication, stability, and structure (O’Connor et al, 2004; Yu et al, 2004). In the
placenta of patients with preeclampsia, phosphorylation of extracellular signal-regulated
kinase1/2 was significantly less frequent in the invasive trophoblasts, as compared to control
(Moon et al, 2008). In another study, in placentas of preeclamptic patients, contrary to
expectations, polymorphisms of several enzymes associated with oxidative stress (copper/zinc
superoxide dismutase, manganese superoxide dismutase, glutathione-S-transferase, and
others) did not differ from controls (Zhang et al, 2008). In contrast, in the placentas of patients
Gheorghe et al. Page 8
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
with HELLP (Hemolysis, Elevated Liver Enzymes, Low Platelets) syndrome, genes encoding
vascular endothelial growth factor receptor, leptin, and several other proteins, were up-
regulated, as compared with the placenta of both normal control patients and those with
preeclampsia (Buimer et al, 2008).
Because the various tissues and organ systems undergo critical, often brief, periods of growth
and development during fetal life (Winick & Noble, 1966), “programming” as a consequence
of maternal stress should not be unexpected, with insults to the developing organism having
consequences later in life’s course. Studies in ruminants also have demonstrated that under-
nutrition can have profound consequences for the fetus. In sheep, restricted maternal nutrition
in early to mid-gestation was associated with an increase in placental weight, an increase in
crown-rump length, and lower fetal to placental weight ratios (Heasman et al, 1998). Maternal
under-nutrition also altered cardiovascular homeostatic regulation by the renin-angiotensin
system, and exposed the lambs to higher levels of glucocorticoids (Edwards et al, 1999), and
development of hypertension (Dodic et al, 2001). Protein restriction in bovines also resulted
in an increase in placental weight and altered placental morphology (Perry et al, 1999).
Studies in rodents have shown similar effects. In rats, maternal protein restriction triggers
hypertension in the pups in adulthood (Langley & Jackson, 1994), probably by augmentation
of the pups’ renin-angiotensin system. In the spontaneously hypertensive rat placenta, several
proteins, angiotensin receptor type I and inducible nitric oxide synthase (NOS), were up-
regulated, while angiotensin converting enzyme and peroxisome proliferator-activated
receptors alpha and gamma were downregulated (Raso et al, 2008). An alteration of placental
glucocorticoid (GC) metabolism also was observed in placentae of rats fed a protein restricted
diet, namely the activity of 11β-hydoxysteroid dehydrogenase that metabolizes
glucocorticoids. This placental enzyme, which normally protects the pups from maternal
glucocorticoid excess, was reduced in protein restricted rats (Langley-Evans et al, 1996), thus
exposing the fetus to abnormally high GC concentrations. Elevated circulating cortisol
concentrations, with modified responsiveness of the hypothalamic-pituitary-adrenal axis, and
elevated mean arterial blood pressure with increased left ventricular wall thickness and mass,
also were observed in guinea pigs in which the dam received only 70% of normal chow during
either the first or second half of gestation. Some of these changes persisted in the F2 generation
(Bertram et al, 2008). Another hormonal alteration in nutritionally deprived rat pups, was an
increase in somatostatin expression in the periventricular nucleus. This led to much lower levels
of growth hormone, and had deleterious effects on the growth of the pups post-partum
(Huizinga et al, 2000). Fetal undernourishment also led to neuronal sequelae. The facial motor
nucleus in pups was under-developed, resulting in decrease in the ability of pups to suckle and
chew (Perez-Torrero et al, 2001). These observations also may relate to the epidemiologic
findings, noted above, that abnormal antenatal nutrition may be associated with the
development of schizophrenia and other mental illness.
In several animal models, in addition to the potential deleterious effects referenced above, a
positive aspect of nutritional deprivation in the adult is that of prolonged lifespan and reduced
cancer rates. A proposed mechanism for these benefits is that nutritional restriction without
severe malnutrition inhibits cellular proliferation and induces apoptosis. This effect has been
shown in mice lacking p53, in which -/- and +/- mutants have lowered spontaneous cancer
rates when fed a calorically reduced, but otherwise complete, diet (Hursting et al, 2004). In
the adult and aging animal, nutritional restriction has been shown to have beneficial effects
that increased life span (Nikolich-Zugich & Messaoudi, 2005). A different picture has emerged
in the fetus, however. As discussed above, caloric and protein deprivation have been shown to
trigger fetal programming of adult disease, and lead to an increased prevalence of metabolic
disorders in adulthood (Barker, 1995b; 1998; Barker & Clark, 1997). In the developing fetus,
numerous animal studies have shown negative long-term effects of caloric and protein
Gheorghe et al. Page 9
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
deprivation on the cardiovascular, renal, nervous system and metabolism (for review see
McMillan & Robinson, 2005). A different form of nutritional compromise, that of placental
restriction in sheep by removal of the endometrial caruncles in the nonpregnant ewe prior to
mating, alters the expression of a number of genes associated with adipogenesis in adipose
tissue of the fetus (Duffield et al, 2008). These findings emphasize the interrelation of placental
development and its gene expression, to development of the fetus and its repertoire of gene
expression.
To What Extent is Placental Gene Expression Altered by Maternal Caloric Excess?
Because maternal obesity poses an increased risk to the fetus during pregnancy, and has long-
term consequences for the progeny, we tested the hypothesis that maternal caloric excess effects
growth-related gene expression changes in the placenta. We fed female C57BL/65 mice a
hypercaloric diet (20% fat, 38% sugar) or standard chow for six weeks prior to mating and
throughout pregnancy. Near-term (E18), the dams were euthanized. We measured gene
expression changes in the placenta, and performed pathway analysis on regulated genes.
Maternal overfeeding was associated with a two-fold increase in body fat mass, with several
genes related to obesity, diabetes, DNA methylation, and the transforming growth factor-beta
(TGF-β) pathway being differentially expressed (Poston et al, in preparation). The TGF-β
superfamily comprises 30 growth and differential factors, including several TGF-βs, activins,
inhibins, and other growth and cell cycle control factors (Goumans & Mummery, 2000; Kitisin
et al, 2007; Massague et al, 2000; Roberts & Mishra, 2005; Roberts & Wakefield, 2003). Thus,
our findings may have important implications for placental growth and epigenetic regulation.
In other studies in mice, the chow was supplemented with methyl supplements (Wolff et al,
1998) or folic acid, vitamin B-12, choline, and betaine to enhance metabolism of cellular methyl
donors (S-adenosylmethionine) (Waterland & Jirtle, 2003). These interventions resulted in
altered coat color phenotype with concomitant increase in DNA methylation at the Avy locus.
Conversely, in mice fed a methyl-donor-deficient diet that lacked folic acid, vitamin B-12, and
choline the imprinted Igf2 gene was down-regulated with altered DNA methylation (Waterland
et al, 2006a). Human studies also have demonstrated effects in the placenta on maternal dietary
supplementation (Rush et al, 1984).
What is the Role Epigenetics in Placental Gene Expression?
During the course of life and reproduction, cells store information that has been handed down
from their ancestors, and that will be transmitted to their descendents. For the most part, this
“memory” is encoded in the sequence of nucleic acids that comprise the DNA of the genome,
the genotype or entire compliment of genes that provides the stability and accurate heritability
from generation to generation. Much traditional research has explored the combined effects of
genetics and the environment in germline mutations of the coding and promoter regions of
genes. In addition, cells can inherit and transmit information that is not part of the genomic
sequence. This “epigenetic” [from Greek, upon, over, or beyond conventional genetic], cellular
memory involves the heritable transmission of gene expression patterns that persist through
cell division, but do not involve an alteration in DNA sequence. Epigenetic processes act in a
cell specific, temporally-regulated manner to direct development, differentiation,
organogensis, and related processes. Some have compared epigenetic mechanisms to the
software to orchestrate and/or modulate the DNA hardware. One major class of epigenetic
mechanisms termed “cytoplasmic”, is determined by cis-acting factors associated with DNA
methylation and/or histone modification by acetylation/methylation/phosphorylation. DNA
with accompanying histones are packaged in nucleosomes, the core of which contains an
octamere of histone proteins. Four basic forms of histones (H2A, H2B, H3, and H4, as well as
minor variants), are encircled by 146 base pairs of DNA (Finch et al, 1977); a fifth histone,
H1, serves as a linker protein (Bernstein et al, 2007). The histone modifications noted above,
Gheorghe et al. Page 10
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
and DNA methylation, confer a great increase in the regulatory capacity of each nucleosome,
allowing specific functions such as DNA repair and gene activation to be modulated in the
appropriate manner (Sarma & Reinberg, 2005). Enzymes critically associated with these
nucleosomal modifications include: DNA methyltransferases (DMT), histone
acetyltransferases (HAT), histone methyltransferase (HMT), histone deacetylases (HDAC),
histone demethylases (HDM), and others (Dodd et al, 2007; Klose et al, 2006). It is by these
nucleosomal modifications, with their influence on proximate genes, that genes may be
regulated to affect phenotype by activity, chromatin structure, dosage compensation, and
epigenetic memory, without changes in the nucleic acid code per se (Martin & Zhang, 2005;
Wolffe & Matzke, 1999).
Epigenetic changes play a key role in normal cellular function, as well as the development and
differentiation of various cell types (Drake & Walker, 2004; Monk, 1998; Rahnama et al,
2006; Reik, 2007). Examples include X-chromosome inactivation in female mammals, and
genomic imprinting in which one parental allele is altered resulting in parent-of-origin, or
random modification of gene transcription (Willard et al, 1993). The epigenetic state can be
disrupted by maternal environmental influences such as hypoxia, protein deprivation, caloric
excess, and so forth which alter DNA methylation or modify histones. Also importantly, a wide
variety of environmental toxins, including low dose radiation and psychological stress, have
been demonstrated to be important in epigenetic mechanisms (Dolinoy et al, 2007; Feinberg,
2007; Hertz-Piccioto et al, 2008; Jirtle & Skinner, 2007; Pryce et al, 2002; Szyf et al, 2007).
Increasingly, epigenetic changes are being recognized to be of importance in ageing, and the
development of cancer and other diseases. Despite the general understanding that DNA and/
or histone modifications constitute a major factor in the pathogenesis of epigenesis, little is
known of the molecular mechanisms whereby these chemical reactions/changes are regulated,
and/or how they are transmitted between generations (Bird, 2007).
From an historical context, epigenetics has several facets. For the pioneer Edinburgh geneticist
Conrad Hal Waddington (1905-1975), who coined the term, epigenetics was the study of how
phenotypes arise from genotypes during development (Waddington, 1939; 1940; 1942;
1957). Epigenetics later was defined as heritable changes in gene expression not due to any
alteration in DNA sequence (Holliday, 1987). In the mid-1970s, the concept of covalent
chemical DNA modifications, including methylation was proposed to account for this
phenomenon (Holliday & Pugh, 1975; Riggs, 1975). More recently, others have defined
epigenesis as the study of mitotically and/or meiotically heritable changes in gene function
without a change in DNA sequence (Dolinoy et al, 2007; Russo et al, 1996). As defined by
Adrian Bird, epigenetics is “the structural adaptation of chromosomal regions so as to register,
signal or perpetuate altered activity states” (Bird, 2007). The latter a definition focuses on
chromosomes and genes, including those aspects such as DNA repair, cell-cycle phases, and
those stable changes maintained from generation to generation.
In terms of perspective, epigenetics has a history that antedates understanding the genome and
its regulation. The French biologist/zoologist and comparative anatomist who contributed
greatly to classification of life forms, Jean-Baptiste Pierre Antoine de Mont, Chevalier de
Lamarck (1774-1829), noted that organisms may inherit traits acquired during their parent’s
lifetime. Although discredited by many, in part, because of his view that simple life forms arose
from dead matter by spontaneous generation to become more complex as they were
transformed into new species, Lamarck held that organisms inherit characteristics acquired
during their parent’s lifetimes, evolving in a constant process of striving toward greater
complexity and “perfection” (Lamarck, 1801; 1809; 1815-1822). Lamarck’s “First Law ...”
stated that a change in the environment alters the needs of organisms in that environment,
resulting in a change in behavior. Such behavioral alteration leads to greater or lesser use of a
given structure with resultant increase or decrease in the size of that structure or organ. His
Gheorghe et al. Page 11
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
“Second Law” stated that all such changes were heritable as a result (For instance, that a
giraffe’s neck elongated as it ate from the highest leaves on a tree, and this feature would be
seen in the next generation) (Haig, 2006; West-Eberhard, 2007). Of course, neo-Lamarckian
biologists soundly reject such an idea. In more contemporary times, the views of the Russian
biologist-agronomist, Trofim Denisovich Lysenko (1898-1976), gained considerable press for
the agricultural “revolution” he promoted, in concert with Soviet collectivization policies. In
essence, Lysenko held that acquired characteristics of a plant (or other organism) could be
inherited by succeeding generations. An ideological-political creation, Lysenkoism held the
study of classic genetics to be “bourgeois” or “fascist” pseudoscience. Lysenkoism invoked
by biological determinists, as with the eugenics and scientific racism adopted by social
constructivists, may be seen as the extremes to which political dogma can use science in
promoting its propaganda (Soyfer, 2001).
As noted, environmental influences may have profound effects on gene regulation. This is
clearly evident in the cells of multicellular organisms; although being genetically
homogeneous, they are structurally and functionally heterogenesis. Many of these differences
in gene expression arise during development and are retained through mitosis. Such stable,
epigenetic changes, although heritable in the short term, are not a consequence of DNA
mutation. Rather, as recent studies are demonstrating, to a great degree epigenesis appears to
be a consequence of DNA methylation and histone modification. The term “epigenomics” has
been applied to the study of altered chromatin structure, such as complex folding, altered
nucleosome configuration, and related phenomena (Murrell et al, 2005). Importantly, several
lines of evidence indicate that, in addition to maternal to fetal transfer, epigenetic modifications
may be inherited across generations (Anway et al, 2005; 2006; Crews et al, 2007; Lane et al,
2003; Morgan et al, 1999; Pembray et al, 2006; Rakyan et al, 2003).
What are the Roles of DNA Methylation and Histone Modification in Placental Gene
Expression?
As noted above, both DNA methylation and histone modification play important roles in
development. These changes also may be important aspects of the ageing process and the
development of cancer. In this review, we concentrate on the epigenetic influences of maternal
diet, hypoxia, and related stress as observed in the placenta and fetal tissues, and that may have
long-term consequences in the fetal origins of adult health and disease.
As with most phenomena of biology and life, the molecular mechanisms whereby genes are
repressed or active in a stable manner are exceedingly complex. The best studied of these
epigenetic modifications is that of DNA methylation, which first was suggested in 1975 by
two groups (Holliday & Pugh, 1975; Riggs, 1975). This post-replication, covalent methylation
occurs predominantly in repetitive genomic regions, on the 5-carbon of cysteine residues that
are followed by a guanine residue, i.e., “CpG methylation” which induces gene repression or
“a silent chromatin state”, (The “p” in CpG refers to the phosphodiester bond between cytosine
and guanine). Occurring at or around promoter regions, forming CpG islands, this can occur
directly by inhibiting the binding of specific transcription factors, and indirectly by recruiting
methyl-CPG binding proteins, with their associated repressive chromatin-remodeling activities
(Razin & Riggs, 1980). A seeming paradox in this scenario, is that methylation of some specific
DNA sequences may permit expression of neighboring genes. Both intrinsic factors and
environmental/nutritional factors can determine the activity of methyltransferases upon which
DNA methylation are dependent (Bestor, 2000). Because of the requirement for a high DNA
synthesis rate during both gametogenesis and early embryogenesis, considerable activity in
DNA methylation/demethylation patterning occurs during both this period of development, a
time during which the cells are vulnerable to abnormal environmental factors. Nonetheless,
nuances of molecular regulation of DNA methylation and histone modification during
Gheorghe et al. Page 12
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
embryogenesis are beyond the scope of this synopsis, and a number of reviews on this topic
are available (Bird, 2007c; Jaenisch, 1997; Jaenisch & Bird, 2003; Jones & Takai, 2001; Santos
et al, 2002).
During the course of mammalian development, a wave of DNA demethylation occurs during
cleavage, followed by genome-wide de novo methylation following implantation (Jaenish,
1997). Although the male genome is widely demethylated shortly after fertilization (Mayer et
al, 2000; Oswald, 2000), the maternal genome is only partly demethylated with subsequent
cleavage divisions (Li, 2002). In the gastrulating embryo, the extent of methylation is high,
decreasing in various tissues during the course of differentiation (Ehrlich et al, 1982). For the
developing embryo and fetus, the methylation/demethylation patterns while being of great
significance, are enormously complex.
Additionally, gene expression is determined by the biochemical organization of the histones
in the nucleosomes around which the DNA is wrapped. Several post-translational covalent
modifications occur on the amino acids that constitute the histone N-terminal tails that modify
their interaction with DNA and/or other nuclear proteins. Acetylation, methylation,
phosphorylation and/or ubiquitination alone, or in combination play a key role in the regulation
by repression or expression of contiguous genes (Jenuwein & Allis, 2001; Strahl & Allis,
2000; Turner, 2000). Again, the regulation of histone modification by acetylation and/or
methylation is highly complex, has been shown to be specific for essentially every cell type,
and may act with DNA methylation to constitute a system of cellular memory (Bird, 2007).
The combination of the several epigenetic modifications of genes as well as non-coding
sequences, the so-called “epigenome” or “epigenotype”, determine the extent to which a given
gene is maintained repressed or active, and influences the phenotype at birth.
What is the Role of MicroRNAs in Placental Gene Expression?
MicroRNAs (miRNA) have emerged as important players in DNA methylation and post-
transcriptional gene regulation (Lujambio et al, 2007; Saito et al, 2006). These are subtypes
of small, non-coding RNA, which are 21-25 nucleotides in length. These miRNAs are capable
of base pairing with mRNA, and fine-tuning gene expression during development and
differentiation, by suppressing their expression in sequence specific manner. Following the
discovery of first miRNA “lin4” in 1993, as a small temporal RNA (Lee et al, 1993), there has
been enormous growth in this family, and identification of their targets. Although miRNAs are
similar to small interfering RNA (siRNA) in their generation pathway and molecular
characteristics, unlike siRNA, miRNA this does not degrade the target mRNA. Rather, they
target the 3 untranslated regions of mRNAs with which they share partial sequence
complementarily, thereby silencing post-transcriptional gene translation. In this way, the
biological system increases or decreases miRNA production to up- or down-regulate gene
expression according to the developmental need, producing desired morphologic and
physiological changes. Moreover, placental miRNA (miR-141, miR-149, miR-229-5p, and
miR135b) are secreted in maternal plasma, and their concentration decreases significantly after
parturition (Chim et al, 2008). This suggests that placental miRNA, in addition to regulating
gene expression in placenta, may be playing an important role in maternal conditions with
obscure etiology, such as preeclampsia or related hypertensive disorders. Studies reveal
differential expression of miRNA (miR-210 and miR-182) in placenta from patients with
preeclampsia and with small for gestational age newborn infants (Pineles et al, 2007). As must
be evident, additional studies will be vital to examine and understand the complexity of
placental genetic regulation, and their contribution to fetal and maternal health and disease.
Gheorghe et al. Page 13
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
What are the Human Correlates?
In several human population studies, it has been reported that the nutritional state of individuals
may have phenotypic consequences for their grandchildren (Kaati et al, 2002; Lumley, 1992).
An example of the role of diet in progeny DNA methylation status and phenotype is evident
in patients with hyper-homocysteinemia (Ingrosso et al, 2003). This disorder is characterized
by excess cellular adenosylhomocysteine, a potent inhibitor of S-adenosylmethionine-
dependent methyltransferases. This suggests the possibility of significantly altered DNA
methylation. In these patients, dietary supplementation with folate restored global methylation
levels, as well as that of the imprinted IGF2-H19 locus (Ingrosso et al, 2003). Several earlier
studies have indicated the developmental importance of folic acid as a dietary factory in
utero, and the manner in which it modulates disease risks later in life (Torrens et al, 2006). It
remains to be determined whether, as in the case of hyper-homocysteinaemia, these phenotypic
effects occur through altered DNA methylation (McKay et al, 2004).
An optimal uterine environment has been shown to be essential for establishment and
maintenance of embryonic epigenetic patterns (Vickaryous & Whitelaw, 2005). Because
embryo culture and manipulation are employed in contemporary assisted reproductive
technologies (ART), the question arises as to the extent to which ART or related procedures
alter DNA methylation patterns, thereby inducing epigenetic changes in the developing
organism (Brar et al, 2001; Feil, 2006; Khosla et al, 2001a; 2001b; Vickaryous & Whitelaw,
2005). Normally DNA methylation is confined to only one of the two parent alleles, thus
imprinted gene loci allow minor alterations to be detected. An issue of great importance is the
extent to which the chemical composition of culture medium, the duration of culture, or other
factors, play a role in effecting changes in DNA methylation or histone modification (Doherty
et al, 2000; Khosla et al, 2001a; 2001b; Mann et al, 2004; Young et al, 2001).
An additional consideration of importance, is the role of environmental toxins in producing
alterations in the nucleosome with epigenetic consequences. An obvious example from mid-
twentieth century is the ingestion of the estrogen-receptor agonist diethylstilbestrol (DES) by
women in an attempt to reduce the risk of spontaneous abortion. This was followed by vaginal
clear cell carcinoma (Swan, 2000), and altered limb development in the first generation, and
deafness in the second generation (Stoll et al, 2003). Anticancer drugs and other environmental
compounds may alter expression of specific genes, as well as the stress-related chaperone
protein heat shock protein (HSP)-90, which may play a role in histone modification (Feil,
2006; Rutherford & Lindquist, 1998). A host of environmental contaminants including
endocrine-disrupting chemicals are now known to demonstrate epigenetic effects on the germ
line, and promote disease across several generations (Crews et al, 2007).
In humans, a number of factors, genetic and epigenetic, can influence placental/fetal growth,
development, and long-term sequelae. Several hypotheses have been proposed to account for
these phenomena. The “thrifty genotype” hypothesis, proposes the existence of genes that
influence birthweight, and determining whether an infant will experience intrauterine growth
restriction (Ong & Dunger, 2000; Prentice et al, 2005; Stöger, 2008). The “thrifty phenotype”
hypothesis postulates that impairment of nutritional supply in early life results in permanent
changes in tissue/organ function to conserve glucose, and prioritize development of the brain,
heart, and other vital organs (Hales & Barker, 2001). A third hypothesis proposes that
epigenetic alterations in gene expression, in the absence of altered DNA sequence, can be
heritable, and may be reversible (Holness & Sugden, 2006). A challenge for our future is to
develop strategies to negate the long-term consequences of these molecular alterations. A
related issue of consequence is the epigenetic basis of dysregulation of gene expression in
cancer. Rather than isolated instances, this may be a major factor in the seemingly increasing
and intractable pandemic of this classes of diseases (for instance see Esteller, 2008; Gil-Yam
et al, 2008; Palii & Robertson, 2007). Recognizing the importance of these vital issues, a recent
Gheorghe et al. Page 14
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
National Institutes of Health initiative, as part of its “Roadmap” program, seeks applications
to study the “Epigenomics of Human Health and Disease” (RFA-RM-08-017).
What are the Overall Perspectives, and Critical Questions to be Explored?
For the long-term well-being of an individual, both placental and fetal growth are essential.
Thus, one would anticipate that profound inhibition of cellular growth at key time points during
development would have grave long-term consequences for the embryo/fetus. This suggests
that the timing of the treatment is a key determinant in the effect on the organism. Since their
development, cDNA and oligo microarrays have proven to be powerful tools in the elucidation
of gene expression patterns and discovery. In addition to examining cellular processes at the
global gene expression level, these instruments have allowed analysis of numerous facets of
normal growth and differentiation, as well as that occurring as a consequence of stress or
malignant transformation. Of particular value, such studies allow analysis of gene expression
by functional classes, as an aid in understanding pathways of cell metabolism, proliferation,
senescence, and death.
As with most tissues and organ systems, placental development and its response to stress
remains a poorly understood process. Placental malfunction or failure accounts for numerous
instances of fetal mortality (Cross et al, 2003), and may play an important role in the genesis
of intrauterine growth restriction (Kingdom et al, 2000), as well as some maternal disease
(Newstead et al, 2007). Numerous genes have been shown to be essential for placental function
as an organ of respiratory gas and nutrient exchange, hormonal synthesis, immune function,
and so forth. As is evident, maternal stresses, whether hypoxia, protein deprivation, caloric
excess, or other, can result in profound alterations in placental gene expression patterns, and
their consequences for growth, differentiation, and metabolism. Figure 4 presents in summary
fashion established and potential pathways by which stress to the mother, whether hypoxia,
protein deprivation, caloric excess, or others, can trigger changes in DNA methylation patterns
and/or histone modification to effect alterations in the patterns of gene expression in the
placenta and/or fetal organs.
Far from presenting a complete picture, the present review depicts but a fraction of what we
need to know to understand more completely the molecular regulation of placental growth and
development, and to lessen the ravages of placental dysfunction. A major challenge for the
future will be to identify those portions of the genome particularly vulnerable to epigenetic
modification which underlie states of health and diseases, and to understand the molecular
mechanisms by which these changes occur. A few of the most obvious questions follow. How
are the demonstrated gene expression profiles regulated? In terms of stress, what determines
the individual patterns of expression, as opposed to the up- or down-regulation of those genes
common to all stressors? What are the developmental stages/times of vulnerability to
environmental, nutritional, or other stress? What environmental factors alter the epigenome in
a deleterious manner, and what are their dose-response relations? What are the mechanisms
by which DNA methylation and/or histone acetylation/methylation are regulated? To what
extent do patterns of gene expression alterations in the placenta influence gene expression in
the several fetal tissues/organs? To what extent can we use the findings of gene expression
responses to stress, to gain an understanding of the phenomenon of epigenesis and its various
manifestations. What is the role of epigenesis in normal development, and in the etiology of
disease? How is it that epigenetic changes evident at the molecular level during embryonic/
fetal life, do not become manifest in the adult organism for many years or decades? What is
the relative importance of epigenetic, as opposed to genetic, changes for long-term sequelae?
To what extent can we develop systems using molecular signatures/adducts to detect invidious
interactions in early life? To what extent can an understanding of these issues provide us
Gheorghe et al. Page 15
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
effective means to contain or counteract their influence and consequences? Can epigenetic
biomarkers be identified that will allow disease detection at an early stage?
These are but a few of the vital questions that must be addressed in our pursuit to improve the
lives and well being of mothers and infants, and the latter’s life as an adult. As biomedical
scientists dedicated to betterment of the human condition, can we do less?
Summary
Successful placental development is crucial for optimal growth, development, maturation, and
survival of the embryo/fetus into adulthood. Numerous epidemiologic and experimental studies
demonstrate the profound influence of intrauterine environment on life, and the diseases to
which one is subject as an adult. For the most part, these invidious influences, whether maternal
hypoxia, protein or caloric deficiency or excess, and others, represent types of maternal stress.
In the present review, we examine certain aspects of gene expression in the placenta as a
consequence of maternal stressors. To examine these issues in a controlled manner, and in a
species in which the genome has been sequenced, most of these reported studies have been
performed in the mouse. Although each individual maternal stress is characterized by up- or
down-regulation of specific genes in the placenta, functional analysis reveals some patterns of
gene expression common to the several forms of stress. Of critical importance, these genes
include those involved in DNA methylation and histone modification, cell cycle regulation,
and related global pathways of great relevance to epigenesis and the developmental origins of
adult health and disease.
Acknowledgments
We thank Brenda Kreutzer and Jimin Suh for their assistance in the preparation of this manuscript. This work was
supported, in part, by USPHS grant HD-03807 to LDL.
REFERENCES
Abu-Farha M, Niles J, Willmore WG. Erythroid-specific 5-aminolevulinate synthase protein is stabilized
by low oxygen and proteasomal inhibition. Biochem Cell Biol 2005;83:620–630. [PubMed: 16234850]
Adams RH, Porras A, Alonso G, Jones M, Vintersten K, Panelli S, Valladares A, Perez L, Klein R,
Nebreda AR. Essential role of p38alpha MAP kinase in placental but not embryonic cardiovascular
development. Mol Cell 2000;6:109–116. [PubMed: 10949032]
Adelman DM, Gertsenstein M, Nagy A, Simon MC, Maltepe E. Placental cell fates are regulated in vivo
by HIF-mediated hypoxia responses. Genes Dev 2000;14:3191–3203. [PubMed: 11124810]
Ain R, Dai G, Dunmore JH, Godwin AR, Soares MJ. A prolactin family paralog regulates reproductive
adaptations to a physiological stressor. Proc Natl Acad Sci USA 2004;101:16543–16548. [PubMed:
15545614]
Ain R, Tash JS, Soares MJ. Prolactin-like protein-A is a functional modulator of natural killer cells at
the maternal-fetal interface. Mol Cell Endocrinol 2003;204:65–74. [PubMed: 12850282]
Allen MP, Nilsen-Hamilton M. Granzymes D, E, F, and G are regulated through pregnancy and by IL-2
and IL-15 in granulated metrial gland cells. J Immunol 1998;161:2772–2779. [PubMed: 9743335]
Anway MD, Cupp AS, Uzumcu M, Skinner MK. Epigenetic transgenerational actions of endocrine
disruptors and male fertility. Science 2005;308:1466–1469. [PubMed: 15933200]
Anway MD, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors. Endocrinology
2006;147:S43–S49. [PubMed: 16690803]
Armitage JA, Khan IY, Taylor PD, Nathanielsz PW, Poston L. Developmental programming of the
metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental
models in mammals? J Physiol (Lond.) 2004;561:355–377. [PubMed: 15459241]
Gheorghe et al. Page 16
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Aronow BJ, Richardson BD, Handwerger S. Microarray analysis of trophoblast differentiation: gene
expression reprogramming in key gene function categories. Physiol Genomics 2001;6:105–116.
[PubMed: 11459926]
Austgulen R, Isaksen CV, Chedwick L, Romundstad P, Vatten L, Craven C. Pre-eclampsia: associated
with increased syncytial apoptosis when the infant is small-for-gestational-age. J Reprod Immunol
2004;61:39–50. [PubMed: 14967220]
Barker, DJP. Fetal and Infant Origins of Adult Disease. BMJ Publishing; London: 1992.
Barker DJ. Fetal origins of coronary heart disease. BMJ 1995a;311:171–174. [PubMed: 7613432]
Barker DJ. Intrauterine programming of adult disease. Mol Med Today 1995b;1:418–423. [PubMed:
9415190]
Barker DJ. In utero programming of chronic disease. Clin Sci (Lond) 1998;95:115–128. [PubMed:
9680492]
Barker D. The midwife, the coincidence, and the hypothesis. Brit Med J 2003;327:1428–1430. [PubMed:
14684636]
Barker DJ. The developmental origins of chronic adult disease. Acta Paediatr Suppl 2004;93:26–33.
[PubMed: 15702667]
Barker, DJP. Fetal origins of adult disease. In: Polin, RA.; Fox, WW.; Abman, SH., editors. Fetal and
Neonatal Physiology. 3rd Ed.. Vol. 1. Saunders; Philadelphia, PA: 2004. p. 160-165.
Barker, DJP. Mothers, Babies, and Health in Later Life. Churchill Livingstone; Edinburgh: 1994.
Barker, DJP. Mothers, Babies, and Health in Later Life. 2nd Ed.. Churchill Livingstone; Edinburgh: 1998.
Barker, DJ.; Clark, PM. Rev Reprod. Vol. 2. 1997. Fetal undernutrition and disease in later life; p.
105-112.
Barker DJ, Gluckman PD, Godfrey KM, Harding JE, Owens JA, Robinson JS. Fetal nutrition and
cardiovascular disease in adult life. Lancet 1993;341:938–941. [PubMed: 8096277]
Barker DJ, Osmond C. Diet and coronary heart disease in England and Wales during and after the Second
World War. J Epidemiol Community Health 1986a;40:37–44. [PubMed: 3711768]
Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and
Wales. Lancet 1986b;1:1077–1081. [PubMed: 2871345]
Barker DJP, Osmond C. Death rates from stroke in England and Wales predicted from past maternal
mortality. BMJ 1987;295:83–86. [PubMed: 3113645]
Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME. Growth in utero, blood pressure in childhood
and adult life, and mortality from cardiovascular disease. BMJ 1989a;298:564–567. [PubMed:
2495113]
Barker DJ, Winter PD, Osmond C, Margetts B, Simmonds SJ. Weight in infancy and death from ischaemic
heart disease. Lancet 1989b;2:577–580. [PubMed: 2570282]
Barker, DJP.; Sultant, HY.; Hanson, MA.; Rodeck, CH.; Spencer, JAD., editors. Fetus and Neonate.
Physiology and Clinical Applications. Vol. III. Growth. Cambridge Univ. Press; Cambridge: 1995.
Fetal programming of human disease; p. 255-276.
Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell 2007;128:669–681. [PubMed:
17320505]
Bertram C, Khan O, Ohri S, Phillips DI, Matthews SG, Hanson MA. Transgenerational effects of prenatal
nutrient restriction on cardiovascular and hypothalamic-pituitary-adrenal function. J Physiol (Lond)
2008;586.8:2217–2229. [PubMed: 18292131]
Bestor TH. The DNA methyltransferases of mammals. Hum Mol Genet 2000;9:2395–2402. [PubMed:
11005794]
Bird A. Perceptions of epigenetics. Nature 2007;447:396–398. [PubMed: 17522671]
Boujendar S, Arany E, Hill D, Remacle C, Reusens B. Taurine supplementation of a low protein diet fed
to rat dams normalizes the vascularization of the fetal endocrine pancreas. J Nutr 2003;133:2820–
2825. [PubMed: 12949371]
Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, Levine SS, Wernig M, Tajonar A,
Ray MK, Bell GW, Otte AP, Vidal M, Gifford DK, Young RA, Jaenisch R. Polycomb complexes
repress developmental regulators in murine embryonic stem cells. Nature 2006;441:349–353.
[PubMed: 16625203]
Gheorghe et al. Page 17
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Brar AK, Handwerger S, Kessler CA, Aronow BJ. Gene induction and categorical reprogramming during
in vitro human endometrial fibroblast decidualization. Physiol Genomics 2001;7:135–148. [PubMed:
11773600]
Buimer M, Kijser R, Jebbink JM, Wehkamp D, van Kampen AHC, Boer K, van der Post JAM, Ris-
Stalpers C. Seven placental transcripts characterize HELLP-syndrome. Placenta 2008;29:444–453.
[PubMed: 18374411]
Bulger M. Hyperacetylated chromatin domains: lessons from heterochromatin. J Biol Chem
2005;280:21689–21692. [PubMed: 15840568]
Canli T, Qiu M, Omura K, Congdon E, Haas BW, Amin Z, Herrmann MJ, Constable RT, Lesch KP.
Neural correlates of epigenesis. Proc Natl Acad Sci USA 2006;103:16033–16038. [PubMed:
17032778]
Cetin I, Foidart JM, Miozzo M, Raun T, Jansson T, Tsatsaris V, Reik W, Cross J, Hauguel-de-Mouson
S, Illsley N, Kingdom J, Huppertz B. Fetal growth restriction: a workshop report. Placenta
2004;25:753–757. [PubMed: 15450396]
Challier JC, Uzan S. Le placenta humain et ses pathologies: l’oxygène en question. Med Sci (Paris)
2003;19:1111–1120. [PubMed: 14648482]
Chen CP, Wang KG, Chen CY, Yu C, Chuang HC, Chen H. Altered placental syncytin and its receptor
ASCT2 expression in placental development and pre-eclampsia. BJOG 2006;113:152–158.
[PubMed: 16411991]
Chim SS, Shing TK, Hung EC, Leung TY, Lau TK, Chiu RW, Lo YM. Detection and characterization
of placental microRNAs in maternal plasma. Clin Chem 2008;54:482–490. [PubMed: 18218722]
Chinni E, Colaizzo D, Margaglione M, Rubini C, D’Ambrosio RL, Giuliani F, Di Vagno G, Grandone
E. Correlation between factors involved in the local haemostasis and angiogenesis in full term human
placenta. Thrombosis Res 2008;122:376–382.
Chu S, DeRisi J, Eisen M, Mulholland J, Botstein D, Brown PO, Herskowitz I. The transcriptional
program of sporulation in budding yeast. Science 1998;282:699–705. [PubMed: 9784122]
Collins FS, Barker AD. Mapping the cancer genome. Pinpointing the genes involved in cancer will help
chart a new course across the complex landscape of human malignancies. Sci Am 2007;296:50–57.
[PubMed: 17348159]
Cooney CA, Dave AA, Wolff GL. Maternal methyl supplements in mice affect epigenetic variation and
DNA methylation of offspring. J Nut 2002;132:2393S–2400S.
Crews D, Gore AC, Hsu TS, Dangleben NL, Spinetta M, Schallert T, Anway MD, Skinner MK.
Transgenerational epigenetic imprints on mate preference. Proc Natl Acad Sci USA 2007;104:5942–
5946. [PubMed: 17389367]
Cross JC. How to make a placenta: mechanisms of trophoblast cell differentiation in mice--a review.
Placenta 2005;26(Suppl A):S3–S9. [PubMed: 15837063]
Cross JC, Baczyk D, Dobric N, Hemberger M, Hughes M, Simmons DG, Yamamoto H, Kingdom JC.
Genes, development and evolution of the placenta. Placenta 2003;24:123–130. [PubMed: 12596737]
Daoud G, Amyot M, Rassart E, Masse A, Simoneau L, Lafond J. ERK1/2 and p38 regulate trophoblasts
differentiation in human term placenta. J Physiol (Lond) 2005;566.2:409–423. [PubMed: 15890698]
Dodd IB, Micheelsen MA, Sneppen K, Thon G. Theoretical analysis of epigenetic cell memory by
nucleosome modification. Cell 2007;129:813–822. [PubMed: 17512413]
Dodic M, Baird R, Hantzis V, Koukoulas I, Moritz K, Peers A, Wintour EM. Organs/systems potentially
involved in one model of programmed hypertension in sheep. Clin Exp Pharmacol Physiol
2001;28:952–956. [PubMed: 11703404]
Doherty AS, Mann MR, Tremblay KD, Bartolomei MS, Schultz RM. Differential effects of culture on
imprinted H19 expression in the pre-implantation embryo. Biol Reprod 2000;62:1526–1535.
[PubMed: 10819752]
Dolinoy DC, Weidman JR, Jirtle RL. Epigenetic gene regulation: Linking early developmental
environment to adult disease. Reprod Toxicol 2007;23:297–307. [PubMed: 17046196]
Drake AJ, Walker BR. The intergenerational effects of fetal programming: non-genomic mechanisms for
the inheritance of low birth weight and cardiovascular risk. J Endocrinol 2004;180:1–16. [PubMed:
14709139]
Gheorghe et al. Page 18
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Duffield JA, Vuocolo T, Tellam R, Yuen BS, Muhlhausler BS, McMillen IC. Placental restriction of fetal
growth decreases IGF1 and leptin mRNA expression in the perirenal adipose tissue of late gestation
fetal sheep. Am J Physiol Regul Integr Comp Physiol 2008;294:R1413–R1419. [PubMed: 18272661]
Edwards LJ, Simonetta G, Owens JA, Robinson JS, McMillen IC. Restriction of placental and fetal growth
in sheep alters fetal blood pressure responses to angiotensin II and captopril. J Physiol (Lond)
1999;515.3:897–904. [PubMed: 10066914]
Ehrlich M, Gama-Sosa MA, Huang LH, Midgett RM, Kuo KC, McCune RA, Gehrke C. Amount and
distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acids
Res 1982;24:2709–2721. [PubMed: 7079182]
Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet
2007;8:286–298. [PubMed: 17339880]
Esteller M. Molecular origins of cancer. Epigenetics in cancer. N Engl J Med 2008;358:1148–1159.
[PubMed: 18337604]
Farese RV Jr. Cases S, Ruland SL, Kayden HJ, Wong JS, Young SG, Hamilton RL. A novel function for
apolipoprotein B: lipoprotein synthesis in the yolk sac is critical for maternal-fetal lipid transport in
mice. J Lipid Res 1996;37:347–360. [PubMed: 9026532]
Feil R. Environmental and nutritional effects on the epigenetic regulation of genes. Mutation Res
2006;600:46–57. [PubMed: 16854438]
Feinberg AP. Phenotypic plasticity and the epigenetics of human disease. Nature 2007;447:433–440.
[PubMed: 17522677]
Finch JT, Lutter LC, Rhodes D, Brown RS, Rushton B, Levitt M, Klug A. Structure of nucleosome core
particles of chromatin. Nature 1977;269:29–36. [PubMed: 895884]
Fratelli M, Goodwin LO, Ørom UA, Lombardi S, Tonelli R, Mengozzi M, Ghezzi P. Gene expression
profiling reveals a signaling role of glutathione in redox regulation. Proc Natl Acad Sci USA
2005;102:13998–14003. [PubMed: 16172407]
Gal-Yam EN, Saito Y, Egger G, Jones PA. Cancer epigenetics: modifications, screening, and therapy.
Annu Rev Med 2008;59:267–280. [PubMed: 17937590]
Gasa R, Mrejen C, Leachman N, Otten M, Barnes M, Wang J, Chakrabarti S, Mirmira R, German M.
Proendocrine genes coordinate the pancreatic islet differentiation program in vitro. Proc Natl Acad
Sci USA 2004;101:13245–13250. [PubMed: 15340143]
Gheorghe CP, Mohan S, Oberg K, Longo LD. Gene expression patterns in the hypoxic murine placenta:
a role in epigenesis? Reprod Sci 2007;14:223–233. [PubMed: 17636235]
Gheorghe C, Mohan S, Longo LD. Gene expression patterns in the developing murine placenta. J Soc
Gynecol Invest 2006;13:256–262.
Gheorghe CP, Holweger J, Mohan S, Longo LD. Maternal protein deprivation and gene expression
patterns in the murine placenta. Physiol Genomics. Submitted.
Gimeno RE, Hirsch DJ, Punreddy S, Sun Y, Ortegon AM, Wu H, Daniels T, Stricker-Krongrad A, Lodish
HF, Stahl A. Targeted deletion of fatty acid transport protein-4 results in early embryonic lethality.
J Biol Chem 2003;278:49512–49516. [PubMed: 14512415]
Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of in utero and early-life conditions on
adult health and disease. N Engl J Med 2008;359:61–73. [PubMed: 18596274]
Godfrey KM. Maternal regulation of fetal development and health in adult life. Eur J Obstet Gynecol
Reprod Biol 1998;78:141–150. [PubMed: 9622311]
Götz AA, Wittlinger S, Stefanski V. Maternal social stress during pregnancy alters immune function and
immune cell numbers in adult male Long-Evans rat offspring during stressful life-events. J
Neuroimmunol 2007;185:95–102. [PubMed: 17350108]
Goumans MJ, Mummery C. Funcitonal analysis of the TGFβ receptor/Smad pathway through gene
ablation in mice. Int J Dev Biol 2000;44:253–265. [PubMed: 10853822]
Green, LR.; Hanson, MA. Programming of the fetal circulation. In: Polin, RA.; Fox, WW.; Abman, SH.,
editors. Fetal and Neonatal Physiology. 3rd Ed.. Vol. 1. Saunders; Philadelphia, PA: 2004. p. 727-732.
Gubin AN, Njoroge JM, Bouffard GG, Miller JL. Gene expression in proliferating human erythroid cells.
Genomics 1999;59:168–177. [PubMed: 10409428]
Gheorghe et al. Page 19
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Haig D. Weismann Rules! Epigenetics and the Lamarckian temptation. Biol Philosophy 2007;22:415–
428.
Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull 2001;60:5–20. [PubMed:
11809615]
Hales CN, Barker DJ, Clark PM, Cox LJ, Fall C, Osmond C, Winter PD. Fetal and infant growth and
impaired glucose tolerance at age 64. BMJ 1991;303:1019–1022. [PubMed: 1954451]
Ham J, Tronick E. Infant resilience to the stress of the still-face. Infant and maternal psychophysiology
are related. Ann NY Acad Sci 2006;1094:297–301. [PubMed: 17347365]
Hanson MA, Gluckman PD. Developmental processes and the induction of cardiovascular function:
conceptual aspects. J Physiol (Lond) 2005;565.1:27–34. [PubMed: 15731193]
Heasman L, Clarke L, Firth K, Stephenson T, Symonds ME. Influence of restricted maternal nutrition in
early to mid gestation on placental and fetal development at term in sheep. Pediatr Res 1998;44:546–
551. [PubMed: 9773844]
Heazel AEP, Lacey HA, Jones CJP, Huppertz B, Baker PN, Crocker IP. Effects of oxygen on cell turnover
and expression of regulators of apoptosis in human placental trophoblast. Placenta 2008;29:175–186.
[PubMed: 18155142]
Hemberger M. Epigenetic landscape required for placental development. Cell Mol Life Sci
2007;64:2422–2436. [PubMed: 17585370]
Hertz-Picciotto I, Park H-Y, Dostal M, Kocan A, Trnovec T, Sram R. Prenatal exposures to persistent
and non-persistent organic compounds and effects on immune system development. Basic Clin
Pharmacol Toxicol 2008;102:146–154. [PubMed: 18226068]
Hirst CE, Buzza MS, Sutton VR, Trapani JA, Loveland KL, Bird PI. Perforin-independent expression of
granzyme B and proteinase inhibitor 9 in human testis and placenta suggests a role for granzyme B-
mediated proteolysis in reproduction. Mol Hum Reprod 2001;7:1133–1142. [PubMed: 11719590]
Hoang VM, Foulk R, Clauser K, Burlingame A, Gibson BW, Fusher SJ. Functional proteomics:
examining the effects of hypoxia on the cytotrophoblast protein repertoire. Biochemistry
2001;40:4077–4086. [PubMed: 11300788]
Hoek HW, Susser E, Buck KA, Lumey LH, Lin SP, Gorman JM. Schizoid personality disorder after
prenatal exposure to famine. Am J Psychiatry 1996;153:1637–1639. [PubMed: 8942466]
Hoet JJ, Hanson MA. Intrauterine nutrition: its importance during critical periods for cardiovascular and
endocrine development. J Physiol (Lond.) 1999;514:617–627. [PubMed: 9882734]
Hofmann TG, Moller A, Sirma H, Zentgraf H, Taya Y, Droge W, Will H, Schmitz ML. Regulation of
p53 activity by its interaction with homeodomain-interacting protein kinase-2. Nature Cell Biol
2002;4:1–10. [PubMed: 11740489]
Holliday R. The inheritance of epigenetic defects. Science 1987;238:163–170. [PubMed: 3310230]
Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science
1975;187:226–232. [PubMed: 1111098]
Holness MJ, Sugden MC. Epigenetic regulation of metabolism in children born small for gestational age.
Curr Opin Clin Nutr Metab Care 2006;9:482–488. [PubMed: 16778581]
Hsueh AM, Agustin CE, Chow BF. Growth of young rats after differential manipulation of maternal diet.
J Nutr 1967;91:195–200. [PubMed: 6021221]
Huang ST, Vo KC, Lyell DJ, Faessen GH, Tulac S, Tibshirani R, Giaccia AJ, Giudice LC. Developmental
response to hypoxia. FASEB J 2004;18:1348–1365. [PubMed: 15333578]
Huizinga CT, Oudejans CB, Steiner RA, Clifton DK, Delemarre-van de Waal HA. Effects of intrauterine
and early postnatal growth restriction on hypothalamic somatostatin gene expression in the rat.
Pediatr Res 2000;48:815–820. [PubMed: 11102552]
Hulshoff HE, Hoek HW, Susser E, Brown AS, Dingemans A, Schnack HG, van Haren NEM, Ramos
LMP, Gispen-de Wied CC, Kahn RS. Prenatal exposure to famine and brain morphology in
schizophrenia. Am J Psychiatry 2000;157:1170–1172. [PubMed: 10873931]
Hursting SD, Lavigne JA, Berrigan D, Donehower LA, Davis BJ, Phang JM, Barrett JC, Perkins SN.
Diet-gene interactions in p53-deficient mice: insulin-like growth factor-1 as a mechanistic target. J
Nutr 2004;134:2482S–2486S. [PubMed: 15333746]
Gheorghe et al. Page 20
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Ingrosso D, Cimmino A, Perna AF, Masella L, De Santo NG, De Bonis ML, Vacca M, D’Esposito M,
D’Urso M, Galletti P, Zappia V. Folate treatment and unbalanced methylation and changes of allelic
expression by hyper-homocysteinaemia in patients with uraemia. Lancet 2003;17:1693–1699.
[PubMed: 12767735]
Iyer VR, Eisen MB, Ross DT, Schuler G, Moore T, Lee JC, Trent JM, Staudt LM, Hudson J Jr, Boguski
MS, Lashkari D, Shalon D, Botstein D, Brown PO. The transcriptional program in the response of
human fibroblasts to serum. Science 1999;283:83–87. [PubMed: 9872747]
Jablonka, E.; Lamb, MJ. Epigenetic Inheritance and Evolution: The Lamarkian Dimension. Oxford Univ.
Press; Oxford: 1995.
Jablonka E, Lamb MJ. The changing concept of epigenetics. Ann NY Acad Sci 2002;981:82–96.
[PubMed: 12547675]
Jaenisch R. DNA methylation and imprinting: Why bother? Trends Genet 1997;13:323–329. [PubMed:
9260519]
Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and
environmental signals. Nature Genetics (Suppl) 2003;33:245–254.
Jansson T, Powell TL. Role of the placenta in fetal programming: underlying mechanisms and potential
interventional approaches. Clin Sci 2007;113:1–13. [PubMed: 17536998]
Jenuwein T, Allis CD. Translating the histone code. Science 2001;293:1074–1079. [PubMed: 11498575]
Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet
2007;8:253–262. [PubMed: 17363974]
Jones CJ, Fox H. An ultrastructural and ultrahistochemical study of the human placenta in maternal pre-
eclampsia. Placenta 1980;1:61–76. [PubMed: 7443636]
Jones PA, Martienssen R. A blueprint for a Human Epigenome Project: the AACR Human Epigenome
Workshop. Cancer Res 2005;65:11241–11246. [PubMed: 16357125]
Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science 2001;293:1068–
1070. [PubMed: 11498573]
Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during
parents’ and grandparents’ slow growth period. Eur J Hum Genet 2002;10:682–688. [PubMed:
12404098]
Khosla S, Dean W, Reik W, Feil R. Culture of pre-implantation embryos and its long-term effects on
gene expression and phenotype. Hum Reprod Update 2001a;7:419–427. [PubMed: 11476355]
Khosla S, Dean W, Brown D, Reik R, Feil R. Culture of pre-implantation mouse embryos affects fetal
development and the expression of imprinted genes. Biol Reprod 2001b;64:918–926. [PubMed:
11207209]
Kim JS, Romero R, Tarca A, LaJeunesse C, Han YM, Kim MJ, Suh YL, Draghici S, Mittal P, Gotsch F,
Kusanovic JP, Hassan S, Kim CJ. Gene expression profiling demonstrates a novel role for fetal
fibrocytes and the umbilical vessels in human fetoplacental development. J Cell Mol Med. doi:
10.1111/j.1582-4934.2008.00284.x.
Kingdom J, Huppertz B, Seaward G, Kaufmann P. Development of the placental villous tree and its
consequences for fetal growth. Eur J Obstet Gynecol Reprod Biol 2000;92:35–43. [PubMed:
10986432]
Kitisin, K.; Saha, T.; Blake, T.; Golestaneh, N.; Deng, M.; Kim, C.; Tang, Y.; Shetty, K.; Mishra, B.;
Mishra, L. TGFβ signaling in development. 2007. p. 1-5.stke.org/cgi/content/full/2007/399/cm1
Klose RJ, Kallin EM, Zhang Y. JmjC-domain-containing proteins and histone demethylation. Nature
Rev Genetics 2006;7:715–727. [PubMed: 16983801]
Krebs C, Longo LD, Leiser R. Term ovine placental vasculature: comparison of sea level and high altitude
conditions by corrosion cast and histomorphometry. Placenta 1997;18:43–51. [PubMed: 9032809]
Kyle UG, Pichard C. The dutch Famine of 1944-1945 : A pathophsiological model of long-term
consequences of wasting disease. Curr Opin Clin Nutrition Metabolic Care 2006;9:388–394.
Lacroix MC, Bolifraud P, Durieux D, Pauloin A, Vidaud M, Kann G. Placental growth hormone and
lactogen production by perifused ovine placental explants: regulation by growth hormone-releasing
hormone and glucose. Biol Reprod 2002;66:555–561. [PubMed: 11870057]
Lamarck, JBPA.; de Monet. Système des aminaux sans vertèbris. Chez l’anteur; Paris: p. 1801GM 215.5
Gheorghe et al. Page 21
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Lamarck, JBPA.; de Monet. Philosophie zoologicque. Vol. 2. J.B. Baillère; Paris: p. 1809GM 216
Lamarck, JBPA.; de Monet. Historoire naturelle des animaux sans vertèbris. Vol. 7. Verdière; Paris: p.
1815-1822.GM 316
Lane N, Dean W, Erhardt S, Hajkova P, Surani A, Walter J, Reik W. Resistance of IAPs to methylation
reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis
2003;35:88–93. [PubMed: 12533790]
Langley SC, Jackson AA. Increased systolic blood pressure in adult rats induced by fetal exposure to
maternal low protein diets. Clin Sci (Lond) 86:217–222. discussion 121, 1994. [PubMed: 8143432]
Langley-Evans SC, Phillips GJ, Benediktsson R, Gardner DS, Edwards CR, Jackson AA, Seckl JR.
Protein intake in pregnancy, placental glucocorticoid metabolism and the programming of
hypertension in the rat. Placenta 1996;17:169–172. [PubMed: 8730887]
Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with
antisense complementarity to lin-14. Cell 1993;75:843–854. [PubMed: 8252621]
Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE, Cole
MF, Isono K, Koseki H, Fuchikami T, Abe K, Murray HL, Zucker JP, Yuan B, Bell GW,
Herbolsheimer E, Hannett NM, Sun K, Odom DT, Otte AP, Volkert TL, Bartel DP, Melton DA,
Gifford DK, Jaenisch R, Young RA. Control of developmental regulators by Polycomb in human
embryonic stem cells. Cell 2006;125:301–313. [PubMed: 16630818]
Levy R, Nelson DM. To be, or not to be, that is the question. Apoptosis in human trophoblast. Placenta
2000;21:1–13. [PubMed: 10692245]
Li M, Liu RH, Jiao LH, Wang WH. Synergetic effects of epidermal growth factor and estradiol on
cytoplasmic maturation of porcine oocytes. Zygote 2002;10:349–354. [PubMed: 12463531]
Lillycrop KA, Phillips ES, Torrens C, Hanson MA, Jackson AA, Burdge GC. Feeding pregnant rats a
protein-restricted diet persistently alters the methylation of specific cytosines in the hepatic
PPARalpha promoter of the offspring. Bri J Nutr 2008;11:1–5.
Lind T. Would more calories per day keep low birthweight at bay? Lancet 1984;1:501–502. [PubMed:
6142217]
Liu YL, Ang SO, Weigent DA, Prchal JT, Bloomer JR. Regulation of ferrochelatase gene expression by
hypoxia. Life Sci 2004;75:2035–2043. [PubMed: 15312748]
Longo LD, Pearce WJ. Invited Review: Fetal cerebrovascular acclimatization responses to high altitude,
long-term hypoxia: A model for prenatal programming of adult disease? Am J Physiol Regul
Integrative Comp Physiol 2005;288:R16–R24.
Lopuhaa CE, Roseboom TJ, Osmond C, Barker DJ, Ravelli AC, Bleker OP, van der Zee JS, van der
Meulen JH. Atopy, lung function, and obstructive airways disease after prenatal exposure to famine.
Thorax 2000;55:555–561. [PubMed: 10856314]
Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C, Setién F, Casado S, Suarez-Gauthier A,
Sanchez-Cespedes M, Git A, Spiteri I, Das PP, Caldas C, Miska E, Esteller M. Genetic unmasking
of an epigenetically silenced microRNA in human cancer cells. Cancer Res 2007;67:1424–1429.
[PubMed: 17308079]
Lumey LH. Decreased birthweights in infants after maternal in utero exposure to the Dutch famine of
1944-1945. Paediatr Pernat Epidemiol 1992;6:240–253.
Lykke-Andersen K, Schaefer L, Menon S, Deng XW, Miller JB, Wei N. Disruption of the COP9
signalosome Csn2 subunit in mice causes deficient cell proliferation, accumulation of p53 and cyclin
E, and early embryonic death. Mol Cell Biol 2003;23:6790–6797. [PubMed: 12972599]
Mann MR, Lee SS, Doherty AS, Verona RI, Nolen LD, Schultz RM, Bartolomei MS. Selective loss of
imprinting in the placenta following pre-implantation development in culture. Development
2004;131:3727–3735. [PubMed: 15240554]
Mao Z, Liu S, Cai J, Huang ZZ, Lu SC. Cloning and functional characterization of the 5-flanking region
of human methionine adenosyltransferase 2A gene. Biochem Biophys Res Commun 1998;248:479–
484. [PubMed: 9703951]
Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol
2005;6:838–849. [PubMed: 16261189]
Massague J, lain SW, Lo RS. TGFβ signaling in growth control, cancer, and heritable disorders. Cell
2000;103:295–309. [PubMed: 11057902]
Gheorghe et al. Page 22
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Mayer W, Niveleau A, Walter J, Fundele R, Haaf T. Demethylation of the zygotic paternal genome.
Nature 2000;403:501–502. [PubMed: 10676950]
McKay JA, Williams EA, Mathers JC. Folate and DNA methylation during in utero development and
aging. Biochem Soc Trans 2004;32:1006–1007. [PubMed: 15506948]
McMillen IC, Robinson JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and
programming. Physiol Rev 2005;85:571–633. [PubMed: 15788706]
Merlot E, Couret D, Otten W. Prenatal stress, fetal imprinting and immunity. Brain Behav Immun
2008;22:42–51. [PubMed: 17716859]
Monk M. Genomic imprinting. Genes Dev 1988;2:921–925. [PubMed: 3049238]
Moon KC, Park JS, Norwitz ER, Kim DI, Oh KJ, Park C-W, Jun JK, Syn HC. Expression of extracellular
signal-regulated kinase1/2 and p38 mitogen-activated protein kinase in the invasive trophoblasts at
the human placental bed. Placenta 2008;29:391–395. [PubMed: 18358530]
Morgan HD, Sutherland HGE, Martin DIK, Whitelaw E. Epigenetic inheritance at the agouti locus in the
mouse. Nat Genet 1999;23:314–318. [PubMed: 10545949]
Morris CJ, Earl JR, Trenam CW, Blake DR. Reactive oxygen species and iron--a dangerous partnership
in inflammation. Int J Biochem Cell Biol 1995;27:109–122. [PubMed: 7767779]
Murrell A, Rakyan VK, Beck S. From genome to epigenome. Hum Mole Genet 2005;14(Suppl 1):R3–
R10.
Mysore TB, Shinkel TA, Collins J, Salvaris EJ, Fisicaro N, Murray-Segal LJ, Johnson LE, Lepore DA,
Walters SN, Stokes R, Chandra AP, O’Connell PJ, d’Apice AJ, Cowan PJ. Overexpression of
glutathione peroxidase with two isoforms of superoxide dismutase protects mouse islets from
oxidative injury and improves islet graft function. Diabetes 2005;54:2109–2116. [PubMed:
15983212]
Nanney DL. Epigenetic control systems. Proc Natl Acad Sci USA 1958;44:712–717. [PubMed:
16590265]
Nelson DM, Smith SD, Furesz TC, et al. Hypoxia reduces expression and function of system A amino
acid transporters in cultured term human trophoblasts. Am J Physiol Cell Physiol 2003;284:C310–
C315. [PubMed: 12388062]
Neugebauer R, Hoek HW, Susser E. Prenatal exposure to wartime famine and development of antisocial
personality disorder in early adulthood. JAMA 1999;282:455–462. [PubMed: 10442661]
Newstead J, von Dadelszen P, MaGee LA. Preeclampsia and future cardiovascular risk. Expert Rev
Cardiovasc Ther 2007;5:283–294. [PubMed: 17338672]
Nijland MJ, Ford SP, Nathanielsz PW. Prenatal origins of adult disease. Curr Opin Obstet Gynecol
2008;20:132–138. [PubMed: 18388812]
Nikolich-Zugich J, Messaoudi I. Mice and flies and monkeys too: caloric restriction rejuvenates the aging
immune system of non-human primates. Exp Gerontol 2005;40:884–893. [PubMed: 16087306]
O’Connor MS, Safari A, Liu D, Qin J, Songyang Z. The human Rap1 protein complex and modulation
of telomere length. J Biol Chem 2004;279:28585–28591. [PubMed: 15100233]
Ong KK, Dunger DB. Thrifty genotypes and phenotypes in the pathogenesis of type 2 diabetes mellitus.
J Pediatr Endocrinol Metab 2000;13(Suppl 6):1419–1424. [PubMed: 11202218]
Osmond C, Barker DJ, Winter PD, Fall CH, Simmonds SJ. Early growth and death from cardiovascular
disease in women. Brit Med J 1993;307:1519–1524. [PubMed: 8274920]
Oswald J, Engemann S, Lane N, Mayer W, Olek A, Fundele R, Dean W, Reik W, Walter J. Active
demethylation of the paternal genome in the mouse zygote. Curr Biol 2000;10:475–478. [PubMed:
10801417]
Painter RC, Roseboom TJ, Bleker OP. Prenatal exposure to the Dutch famine and disease in later life:
An overview. Reprod Toxicol 2005a;20:345–352. [PubMed: 15893910]
Painter RC, Roseboom TJ, van Montfrans GA, Bossuyt PM, Krediet RT, Osmond C, Barker DJ, Bleker
OP. Microalbuminuria in adults after prenatal exposure to the Dutch famine. J Am Soc Nephrol
2005b;16:189–194. [PubMed: 15548563]
Palii SS, Robertson KD. Epigenetic control of tumor suppression. Crit Rev Eukaryot Gene Expr
2007;17:295–316. [PubMed: 17725495]
Gheorghe et al. Page 23
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Parham P. NK cells and trophoblasts: partners in pregnancy. J Exp Med 2004;200:951–955. [PubMed:
15492121]
Parodi A, Neasham D, Vineis P. Environment, population, and biology. A short history of modern
epidemiology. Perspect Biol Med 2006;49:357–368. [PubMed: 16960306]
Pembrey ME. Time to take epigenetic inheritance seriously. Eur J Hum Genet 2002;10:669–671.
[PubMed: 12404095]
Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjöström M, Golding J, ALSPAC Study
Team. Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet
2006;14:159–166. [PubMed: 16391557]
Penninga L, Longo LD. Ovine placentome morphology: effect of high altitude, long-term hypoxia.
Placenta 1998;19:187–193. [PubMed: 9548186]
Perez-Torrero E, Torrero C, Salas M. Effects of perinatal undernourishment on neuronal development of
the facial motor nucleus in the rat. Brain Res 2001;905:54–62. [PubMed: 11423079]
Perry VE, Norman ST, Owen JA, Daniel RC, Phillips N. Low dietary protein during early pregnancy
alters bovine placental development. Anim Reprod Sci 1999;55:13–21. [PubMed: 10099675]
Pineles BL, Romero R, Montenegro D, Tarca AL, Han YM, Kim YM, Draghici S, Espinoza J, Kusanovic
JP, Mittal P, Hassan SS, Kim CJ. Distinct subsets of microRNAs are expressed differentially in the
human placentas of patients with preeclampsia. Am J Obstet Gynecol 2007;196:261.e1–e6.
[PubMed: 17346547]
Poston, L.; Taylor, P.; Gheorghe, CP.; Longo, LD. Maternal caloric excess: placental TGFβ and DNA
methylation pathway gene expression changes and epigenesis. In preparation
Prentice AM, Rayco-Solon P, Moore SE. Insights from the developing world: Thrifty genotypes and
thrifty phenotypes. Proc Nutr Soc 2005;64:153–161. [PubMed: 15960860]
Pryce CR, Ruedi-Bettschen D, Dettling AC, Feldon J. Early life stress: Long-term physiological impact
in rodents and primates. News Physiol Sci 2002;17:150–155. [PubMed: 12136043]
Rahnama F, Shafiei F, Gluckman PD, Mitchell MD, Lobie PE. Epigenetic regulation of human
trophoblastic cell migration and invasion. Endocrinology 2006;147:5275–5283. [PubMed:
16887905]
Rakyan VK, Chong S, Champ ME, Cuthbert PC, Morgan HD, Luu KV, Whitelaw E. Transgenerational
inheritance of epigenetic states at the murine AxinFu allele occurs after maternal and paternal
transmission. Proc Natl Acad Sci USA 2003;100:2538–2543. [PubMed: 12601169]
Raso GM, Bianco G, Esposito E, Iacono A, Meli R, Autore G. Evaluation of placental protein
modifications in normotensive and spontaneously hypertensive rats. Placenta 2008;29:429–435.
[PubMed: 18358529]
Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine exposure in utero and early infancy.
N Engl J Med 1976;295:349–353. [PubMed: 934222]
Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, Bleker OP. Glucose
tolerance in adults after prenatal exposure to famine. Lancet 1998;351:173–177. [PubMed:
9449872]
Ravelli ACJ, van der Meulen JHP, Osmond C, Barker DJP, Bleker OP. Obesity at the age of 50 y in men
and women exposed to famine prenatally. Am J Clin Nutr 1999;70:811–816. [PubMed: 10539740]
Razin A, Riggs AD. DNA methylation and gene function. Science 1980;210:604–610. [PubMed:
6254144]
Reaume AG, de Sousa PA, Kulkarni S, Langille BL, Zhu D, Davies TC, Juneja SC, Kidder GM, Rossant
J. Cardiac malformation in neonatal mice lacking connexin 43. Science 1995;267:1831–1834.
[PubMed: 7892609]
Redline RW, Chernicky CL, Tan HQ, Ilan J, Ilan J. Differential expression of insulin-like growth factor-
II in specific regions of the late (post day 9.5) murine placenta. Mol Reprod Dev 1993;36:121–129.
[PubMed: 8257562]
Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature
2007;447:425–432. [PubMed: 17522676]
Reshetnikova OS, Burton GJ, Milovanov AP. Effects of hypobaric hypoxia on the fetoplacental unit: the
morphometric diffusing capacity of the villous membrane at high altitude. Am J Obstet Gynecol
1994;171:1560–1565. [PubMed: 7802068]
Gheorghe et al. Page 24
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Reynolds RM, Godfrey KM, Barker M, Osmond C, Phillips DIW. Stress responsiveness in adult life:
influence of mother’s diet in late pregnancy. J Clin Endocrin Metab 2007;92:2208–2210.
Riggs AD. X inactivation, differentiation, and DNA methylation. Cytogenet Cell Genet 1975;14:9–25.
[PubMed: 1093816]
Roberts A, Mishra L. Role of TGF-β in stem cells and cancer. Oncogene 2005;24:5667.
Roberts AB, Wakefield LM. The two faces of transforming growth factor β in carcinogenesis. Proc Natl
Acad Sci USA 2003;100:8621–8623. [PubMed: 12861075]
Roh CR, Budhraja V, Kim HS, Nelson DM, Sadovsky Y. Microarray-based identification of differentially
expressed genes in hypoxic term human trophoblasts and in placental villi of pregnancies with
growth restricted fetuses. Placenta 2005;26:319–328. [PubMed: 15823618]
Roseboom TJ, van der Meulen JH, Ravelli AC, Osmond C, Barker DJ, Bleker OP. Effects of prenatal
exposure to the Dutch famine on adult disease in later life: an overview. Mol Cell Endocrinol 2001a;
185:93–98. [PubMed: 11738798]
Roseboom TJ, van der Meulen JH, Ravelli AC, Osmond C, Barker DJ, Bleker OP. Effects of prenatal
exposure to the Dutch famine on adult disease in later life: an overview. Twin Res 2001b;4:293–
298. [PubMed: 11869479]
Roseboom TJ, van der Meulen JH, Osmond C, Barker DJ, Ravelli AC, Bleker OP. Plasma lipid profiles
in adults after prenatal exposure to the Dutch famine. Am J Clin Nutr 2000;72:1101–1106.
[PubMed: 11063435]
Rossant J, Cross JC. Placental development: lessons from mouse mutants. Nat Rev Genet 2001;2:538–
548. [PubMed: 11433360]
Rush D, Kristal A, Navarro C, Chauhan P, Blanc W, Naeye R, Susser MW. The effects of dietary
supplementation during pregnancy on placental morphology, pathology, and histomorphometry.
Am J Clin Nutr 1984;39:863–871. [PubMed: 6372428]
Russo, VEA.; Martienssen, RA.; Riggs, AD., editors. Epigenetic Mechanisms of Gene Regulation. Cold
Spring Harbor Laboratory Press; Woodbury, NY: 1996.
Rutherford SL, Lindquist S. Hsp90 as a capacitor for morphological evolution. Nature 1998;396:336–
342. [PubMed: 9845070]
Ryan KM, Phillips AC, Vousden KH. Regulation and function of the p53 tumor suppressor protein. Curr
Opin Cell Biol 2001;13:332–337. [PubMed: 11343904]
Sadovsky Y, Wyatt SM, Collins L, Elchalal U, Kraus FT, Nelson DM. The use of needle biopsy for
assessment of placental gene expression. Am J Obstet Gynecol 2006;194:1137–1144. [PubMed:
16580313]
Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, Jones PA. Specific activation of
microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs
in human cancer cells. Cancer Cell 2006;9:435–443. [PubMed: 16766263]
Santos F, Hendrich B, Reik W, Dean W. Dynamic reprogramming of DNA methylation in the early
mouse embryo. Dev Biol 2002;241:172–182. [PubMed: 11784103]
Sarma K, Reinberg D. Histone variants meet their match. Nat Rev Mol Cell Biol 2005;6:139–149.
[PubMed: 15688000]
Schmidt I. Metabolic diseases: the environment determines the odds, even for genes. News Physiol Sci
2002;17:115–121. [PubMed: 12021382]
Schorpp-Kistner M, Wang ZQ, Angel P, Wagner EF. JunB is essential for mammalian placentation.
EMBO J 1999;18:934–948. [PubMed: 10022836]
Schreiber J, Riethmacher-Sonnenberg E, Riethmacher D, Tuerk EE, Enderich J, Bosl MR, Wegner M.
Placental failure in mice lacking the mammalian homolog of glial cells missing, GCMa. Mol Cell
Biol 2000;20:2466–2474. [PubMed: 10713170]
Selye, H. A treatise based on the concepts of the GENERAL-ADAPTATION-SYNDROME and the
DISEASES OF ADAPTATION. Acta Inc.; Montreal: 1950. The physiology and patheology of
exposure to STRESS.
Shekhawat P, Bennett MJ, Sadovsky Y, Nelson DM, Rakheja D, Strauss AW. Human placenta
metabolizes fatty acids: implications for fetal fatty acid oxidation disorders and maternal liver
diseases. Am J Physiol Endocrinol Metab 2003;284:E1098–1105. [PubMed: 12582009]
Gheorghe et al. Page 25
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Sibley CP, Coan PM, Ferguson-Smith AC, Dean W, Hughes J, Smith P, Reik W, Burton GJ, Fowden
AL, Constancia M. Placental-specific insulin-like growth factor 2 (Igf2) regulates the diffusional
exchange characteristics of the mouse placenta. Proc Natl Acad Sci USA 2004;101:8204–8208.
[PubMed: 15150410]
Simmons DG, Cross JC. Determinants of trophoblast lineage and cell subtype specification in the mouse
placenta. Dev Biol 2005;284:12–24. [PubMed: 15963972]
Sitras V, Paulssen RH, Grønaas H, Vårtun Å, Acharya G. Gene expression profile in labouring and non-
labouring human placenta near term. MHR-Basic Science of Reprod Med 2008;14:61–65.
Soleymanlou N, Jurisica I, Nevo O, Ietta F, Zhang X, Zamudio S, Post M, Caniggia I. Molecular evidence
of placental hypoxia in preeclampsia. J Clin Endocrinol Metab 2005;90:4299–4308. [PubMed:
15840747]
Sood R, Zehnder JL, Druzin ML, Brown PO. Gene expression patterns in human placenta. PNAS
2006;103:5478–5483. [PubMed: 16567644]
Soyfer VN. The consequences of political dictatorhsip for Russian science. Nature Rev Genetics
2001;2:723–729. [PubMed: 11533721]
Spira A, Philippe E, Spira N, Dreyfus J, Schwartz D. Smoking during pregnancy and placental pathology.
Biomedicine 1977;27:266–270. [PubMed: 588667]
Stein AD, Zybert PA, van de Bor M, Lumey LH. Intrauterine famine exposure and body proportions at
birth: the Dutch Hunger Winter. Int J Epidemiol 2004;33:831–836. [PubMed: 15166208]
Stein Z, Susser M. The Dutch famine, 1944-1945, and the reproductive process. II. Interrelations of caloric
rations and six indices at birth. Pediatr Res 1975;9:76–83. [PubMed: 1118194]
Stöger R. The thrifty epigenotype: An acquired and heritable predisposition for obesity and diabetes?
Bioessays 2008;30:156–166. [PubMed: 18197594]
Stoll C, Alembik Y, Dott B. Limb reduction defects in the first generation and deafness in the second
generation of intrauterine exposed fetuses to diethylstilbe strol. Annales de genetique 2003;46:459–
465. [PubMed: 14659782]
Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000;403:41–45. [PubMed:
10638745]
Susser ES, Lin SP. Schizophrenia after prenatal exposure to the Dutch Hunger Winter of 1944-1945.
Arch Gen Psychiatry 1992;49:983–988. [PubMed: 1449385]
Swan SH. Intrauterine exposure to diethylstilbestrol: Long-term effects in humans. APMIS
2000;108:793–804. [PubMed: 11252812]
Szyf M, Weaver I, Meaney M. Maternal care, the epigenome and phenotypic differences in behavior.
Reprod Toxicol 2007;24:9–19. [PubMed: 17561370]
Tanaka TS, Jaradat SA, Lim MK, Kargul GJ, Wang X, Grahovac MJ, Pantano S, Sano Y, Piao Y, Nagaraja
R, Doi H, Wood WH 3rd, Becker KG, Ko MS. Genome-wide expression profiling of mid-gestation
placenta and embryo using a 15,000 mouse developmental cDNA microarray. Proc Natl Acad Sci
USA 2000;97:9127–9132. [PubMed: 10922068]
Torrens C, Brawley L, Anthony FW, Dance CS, Dunn R, Jackson AA, Poston L, Hanson MA. Folate
supplementation during pregnancy improves offspring cardiovascular dysfunction induced by
protein restriction. Hypertension 2006;47:982–987. [PubMed: 16585422]
Trollman R, Klingmüller K, Schild RL, Rascher W, Dötsch J. Differential gene expression of
somatotrophic and growth factors in response to in vivo hypoxia in human placenta. Am J Obstet
Gynecol 2007;197:601.e1–e6. [PubMed: 18060947]
Tu MT, Grunau RE, Petrie-Thomas J, Haley DW, Weinberg J, Whitfield MF. Maternal stress and
behavior modulate relationships between neonatal stress, attention, and basal cortisol at 8 months
in preterm infants. Dev Psychobiol 2007;49:150–164. [PubMed: 17299787]
Turek-Plewa J, Jagodzinski PP. The role of mammalian DNA methyltransferases in the regulation of
gene expression. Cell Mol Biol Lett 2005;10:631–647. [PubMed: 16341272]
Turner BM. Histone acetylation and an epigenetic code. Bioessays 2000;22:836–845. [PubMed:
10944586]
Vadlamudi S, Kalhan SC, Patel MS. Persistence of metabolic consequences in the progeny of rats fed a
HC formula in their early postnatal life. Am J Physiol 1995;269:E731–E738. [PubMed: 7485488]
Gheorghe et al. Page 26
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Vanderlelie J, Gude N, Perkins AV. Antioxidant gene expression in preeclamptic placentae: A
preliminary investigation. Placenta 2008;29:519–522. [PubMed: 18395794]
Vickaryous N, Whitelaw E. The role of early embryonic environment on epigenotype and phenotype.
Reprod Fertil Dev 2005;17:335–340. [PubMed: 15745641]
Waddington CH. Preliminary notes on the development of the wings in normal and mutant strains of
drosophila. Proc Natl Acad Sci USA 1939;25:299–307. [PubMed: 16577903]
Waddington, CH. Organisers and Genes. Cambridge University Press; Cambridge: 1940.
Waddington CH. The epigenotype. Endeavour 1942;1:18–20.
Waddington, CH. The Strategy of the Genes. Allen & Unwin; London: 1957.
Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic gene
regulation. Mol Cell Biol 2003;23:5293–5300. [PubMed: 12861015]
Waterland RA, Dolinoy DC, Lin J-R, Smith CA, Shi X, Tahiliani K. Maternal methyl supplements
increase offspring DNA methylation at Axin (fused). Genesis 2006a;44:401–406. [PubMed:
16868943]
Waterland RA, Lin J-R, Smith CA, Jirtle RL. Post-weaning diet affects genomic imprinting at the insulin-
like growth factor 2 (Igf2) locus. Hum Mol Genet 2006b;15:705–716. [PubMed: 16421170]
Watkins AJ, Wilkins A, Cunningham C, Perry VH, Seet MJ, Osmond C, Eckert JJ, Torrens C, Cagampang
FRA, Cleal J, Gray WP, Hanson MA, Fleming TP. Low protein diet fed exclusively during mouse
oocyte maturation leads to behavioural and cardiovascular abnormalities in offspring. J Physiol
(Lond) 2008;586.8:2231–2244. [PubMed: 18308825]
Weaver ICG, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, Szyf M. Reversal of maternal
programming of stress responses in adult offspring through methyl supplementation: Altering
epigenetic marking later in life. J Neurosci 2005;25:11045–11054. [PubMed: 16306417]
Wehby GL, Murray JC. The effects of prenatal use of folic acid and other dietary supplements on early
child development. Matern Child Health J 2008;12:180–187. [PubMed: 17554612]
West-Eberhard MJ. Dancing with DNA and flirting with the ghost of Lamarck. Biol Philosophy
2007;22:439–451.
Wiemers DO, Shao LJ, Ain R, Dai G, Soares MJ. The mouse prolactin gene family locus. Endocrinology
2003;44:313–325. [PubMed: 12488360]
Willard HF, Brown CJ, Carrel L, Hendrich B, Miller AP. Epigenetic and chromosomal control of gene
expression: Molecular and genetic analysis of X chromosome inactivation. Cold Spring Harb Symp
Quant Biol 1993;58:315–322. [PubMed: 7956044]
Winick M, Noble A. Quantitative changes in ribonucleic acids and protein during normal growth of rat
placenta. Nature 1966;212:34–35. [PubMed: 5965566]
Wolffe GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl supplements affect
agouti gene expression in Avy/a mice. FASEB J 1998;12:949–957. [PubMed: 9707167]
Wolffe AP, Matzke MA. Epigenetics: regulation through repression. Science 1999;286:481–486.
[PubMed: 10521337]
Yamamoto H, Flannery ML, Kupriyanov S, Pearce J, McKercher SR, Henkel GW, Maki RA, Werb Z,
Oshima RG. Defective trophoblast function in mice with a targeted mutation of Ets2. Genes Dev
1998;12:1315–1326. [PubMed: 9573048]
Yinon Y, Nevo O, Xu J, Many A, Rolfo A, Todros T, Post M, Caniggia I. Severe intrauterine growth
restriction pregnancies have increased placental endoglin levels. Hypoxic regulation via
transforming growth factor-β3. Am J Pathol 2008;172:77–85. [PubMed: 18156205]
Yoon K, Smart RC. C/EBPalpha is a DNA damage-inducible p53-regulated mediator of the G1
checkpoint in keratinocytes. Mol Cell Biol 2004;24:10650–10660. [PubMed: 15572670]
Young LE, Fernandes K, McEvoy TG, Butterwith SC, Gutierrez CG, Carolan C, Broadbent PJ, Robinson
JJ, Wilmut I, Sinclair KD. Epigenetic changes in IGF2R is associated with fetal overgrowth after
sheep embryo culture. Nature Genet 2001;27:153–154. [PubMed: 11175780]
Yu Z, Feng D, Liang C. Pairwise interactions of the six human MCM protein subunits. J Mol Biol
2004;340:1197–1206. [PubMed: 15236977]
Zamudio S. The placenta at high altitude. High Alt Med Biol 2003;4:171–191. [PubMed: 12855050]
Gheorghe et al. Page 27
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Zhang EG, Burton GJ, Smith SK, Charnock-Jones DS. Placental vessel adaptation during gestation and
to high altitude: changes in diameter and perivascular cell coverage. Placenta 2002;23:751–762.
[PubMed: 12398815]
Zhang J, Masiocchi M, Lewis D, Sun W, Liu A, Wang Y. Placental anti-oxidant gene polymorphisms,
enzyme activity, and oxidative stress in preeclampsia. Placenta 2008;29:439–443. [PubMed:
18387669]
Zhu H, Jackson T, Bunn HF. Detecting and responding to hypoxia. Nephrol Dial Transplant 2002;17
(Suppl 1):3–7. [PubMed: 11812905]
Zollers WG Jr, Babischkin JS, Pepe GJ, Albrecht ED. Developmental regulation of placental insulin-like
growth factor (IGF)-II and IGF-binding protein-1 and -2 messenger RNA expression during primate
pregnancy. Biol Reprod 2001;65:1208–1214. [PubMed: 11566745]
Gheorghe et al. Page 28
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1.
Over-represented gene functional classes throughout mouse placental development. Listed are
the major groups of upregulated placental genes expressed from Embryonic day 10.5 to 17.5.
Gheorghe et al. Page 29
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2.
Proposed mechanism of hypoxia-mediated epigenetic changes with long-term programming.
Gheorghe et al. Page 30
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3.
A: p53 related genes up-regulated by protein restriction in the mouse placenta. + stands for
induced
B: Proposed mechanisms of protein restriction induced long-term changes in gene expression.
Gheorghe et al. Page 31
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4.
Proposed model of stress-induced long-term epigenetic changes in gene expression, adaptation,
and phenotype.
Gheorghe et al. Page 32
Int J Dev Biol. Author manuscript; available in PMC 2011 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... There was also evidence of impaired glucose tolerance in the PD group as the blood glucose concentrations remained higher and a failure of blood glucose concentration to return to baseline after the 2-hour test in all developmental stages. Glucocorticoids have been shown to inhibit pancreatic-β cells from secreting insulin directly, impair insulin-mediated glucose uptake, and interfere in the insulin signalling cascade in peripheral tissues such as skeletal muscle, thus inducing diabetogenic effects and insulin resistance [188,189]. However, studies show that non-diabetic individuals have the ability to counteract the insulin resistance caused by glucocorticoids by either enhancing pancreatic β-cell activity or increasing insulin secretion and sensitivity [190,191]. ...
Article
Full-text available
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic–pituitary–adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
... Maternal viral infection, such as infection of the enveloped ribonucleic acid (RNA) single-stranded Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), during pregnancy is a risk factor for pregnancy complications, neonatal and fetal complications, and long-term health issues [4,5]. Placental gene expression profiling (placental transcriptomics) studies have revealed evidence of differential expression at different stages of a placenta's development, physiology, and pathology [6][7][8][9][10]. Typically, genes are categorized into gene families or gene groups based on a variety of characteristics, including shared protein sequences and function [11]. ...
Article
Full-text available
The human placenta is a multifunctional, disc-shaped temporary fetal organ that develops in the uterus during pregnancy, connecting the mother and the fetus. The availability of large-scale datasets on the gene expression of placental cell types and scholarly articles documenting adverse pregnancy outcomes from maternal infection warrants the use of computational resources to aid in knowledge generation from disparate data sources. Using maternal Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection as a case study in microbial infection, we constructed integrated datasets and implemented visual analytics resources to facilitate robust investigations of placental gene expression data in the dimensions of flow, curation, and analytics. The visual analytics resources and associated datasets can support a greater understanding of SARS-CoV-2-induced changes to the human placental expression levels of 18,882 protein-coding genes and at least 1233 human gene groups/families. We focus this report on the human aquaporin gene family that encodes small integral membrane proteins initially studied for their roles in water transport across cell membranes. Aquaporin-9 (AQP9) was the only aquaporin downregulated in term placental villi from SARS-CoV-2-positive mothers. Previous studies have found that (1) oxygen signaling modulates placental development; (2) oxygen tension could modulate AQP9 expression in the human placenta; and (3) SARS-CoV-2 can disrupt the formation of oxygen-carrying red blood cells in the placenta. Thus, future research could be performed on microbial infection-induced changes to (1) the placental hematopoietic stem and progenitor cells; and (2) placental expression of human aquaporin genes, especially AQP9.
... Pre-eclampsia, the use of corticosteroids (and other medications, such as antibiotics) and stress have been associated with epigenetic changes affecting gene expression, placental immune function, growth retardation and congenital disabilities [67]. Maternal stress is critical for gene expression in the placenta, as adrenal glucocorticoid production modulates inflammatory gene expression via the hypothalamic-pituitary-adrenal axis, with recognized effects on glutathione metabolism and DNA methylation [68]. Inflammatory diseases during pregnancy can also alter the immune function of the placenta through the sex-specific production of cortisol [69]. ...
Article
Full-text available
Allergic diseases are one of the most common chronic conditions and their prevalence is on the rise. Environmental exposure, primarily prenatal and early life influences, affect the risk for the development and specific phenotypes of allergic diseases via epigenetic mechanisms. Exposure to pollutants, microorganisms and parasites, tobacco smoke and certain aspects of diet are known to drive epigenetic changes that are essential for immune regulation (e.g., the shift toward T helper 2-Th2 cell polarization and decrease in regulatory T-cell (Treg) differentiation). DNA methylation and histone modifications can modify immune programming related to either pro-allergic interleukin 4 (IL-4), interleukin 13 (IL-13) or counter-regulatory interferon γ (IFN-γ) production. Differential expression of small non-coding RNAs has also been linked to the risk for allergic diseases and associated with air pollution. Certain exposures and associated epigenetic mechanisms play a role in the susceptibility to allergic conditions and specific clinical manifestations of the disease, while others are thought to have a protective role against the development of allergic diseases, such as maternal and early postnatal microbial diversity, maternal helminth infections and dietary supplementation with polyunsaturated fatty acids and vitamin D. Epigenetic mechanisms are also known to be involved in mediating the response to common treatment in allergic diseases, for example, changes in histone acetylation of proinflammatory genes and in the expression of certain microRNAs are associated with the response to inhaled corticosteroids in asthma. Gaining better insight into the epigenetic regulation of allergic diseases may ultimately lead to significant improvements in the management of these conditions, earlier and more precise diagnostics, optimization of current treatment regimes, and the implementation of novel therapeutic options and prevention strategies in the near future.
... For instance, placental gene expression profiles have been linked to changes in brain morphology, altered stress reactivity, and behavioral outcomes in rodents and humans [74][75][76][77]. Recent studies have shown that placental miRNA expression patterns are altered by maternal stress exposure and can lead to long-term effects on offspring neurodevelopment [27,78,79]. Therefore, elucidating the specific molecular and cellular alterations in the placenta associated with prenatal stress and SERT genotype may provide additional insights into the pathogenesis of ASD and inform the development of novel preventative or therapeutic strategies. ...
Article
Full-text available
Autism Spectrum Disorder (ASD) has been associated with a complex interplay between genetic and environmental factors. Prenatal stress exposure has been identified as a possible risk factor, although most stress-exposed pregnancies do not result in ASD. The serotonin transporter (SERT) gene has been linked to stress reactivity, and the presence of the SERT short (S)-allele has been shown to mediate the association between maternal stress exposure and ASD. In a mouse model, we investigated the effects of prenatal stress exposure and maternal SERT genotype on offspring behavior and explored its association with maternal microRNA (miRNA) expression during pregnancy. Pregnant female mice were divided into four groups based on genotype (wildtype or SERT heterozygous knockout (Sert-het)) and the presence or absence of chronic variable stress (CVS) during pregnancy. Offspring behavior was assessed at 60 days old (PD60) using the three-chamber test, open field test, elevated plus-maze test, and marble-burying test. We found that the social preference index (SPI) of SERT-het/stress offspring was significantly lower than that of wildtype control offspring, indicating a reduced preference for social interaction on social approach, specifically for males. SERT-het/stress offspring also showed significantly more frequent grooming behavior compared to wildtype controls, specifically for males, suggesting elevated repetitive behavior. We profiled miRNA expression in maternal blood samples collected at embryonic day 21 (E21) and identified three miRNAs (mmu-miR-7684-3p, mmu-miR-5622-3p, mmu-miR-6900-3p) that were differentially expressed in the SERT-het/stress group compared to all other groups. These findings suggest that maternal SERT genotype and prenatal stress exposure interact to influence offspring behavior, and that maternal miRNA expression late in pregnancy may serve as a potential marker of a particular subtype of ASD pathogenesis.
... (The figure was made using vectorized images designed by Freepik.es) modifications, which regulate gene expression nutrient the availability depending manner (Gheorghe et al. 2010). The methylation of DNA and the histone-binding proteins control transcription, replication, recombination, and DNA repair, which interact to trigger a cascade of transduction signals responsible for generating gene-silencing networks. ...
Book
This edited volume aims to describe physiological and pathophysiological mechanisms that underlie human maternal-fetal interactions. The book emphasizes the structure and development of the fetoplacental unit, the endocrine and nutritional regulation of fetal development, nitric oxide signalling, solute carriers function and ion channels regulation in healthy pregnancies and diseases, like preeclampsia, gestational diabetes, and maternal obesity, among others. Also, we highlight novel mechanisms associated with language impairment in children, the use of serotonin inhibitors or cannabis during pregnancy, and maternal conditions' potential impact on cerebrovascular development in newborns and infants. The cellular and molecular understanding of maternal-fetal physiology and pathophysiology will allow the readers to understand the impact of diseases or conditions that are highly prevalent in pregnant women.
... (The figure was made using vectorized images designed by Freepik.es) modifications, which regulate gene expression nutrient the availability depending manner (Gheorghe et al. 2010). The methylation of DNA and the histone-binding proteins control transcription, replication, recombination, and DNA repair, which interact to trigger a cascade of transduction signals responsible for generating gene-silencing networks. ...
Chapter
Currently, more than 100,000 papers had been published studying the placenta in both physiological and pathological contexts. However, relevant health conditions affecting placental function, mostly found in low-income countries, should be evaluated deeper. This review will raise some - of what we think necessary - points of discussion regarding challenging topics not fully understood, including the paternal versus maternal contribution on placental genes imprinting, placenta-brain communication, and some environmental conditions affecting the placenta. The discussions are parts of an international effort to fulfil some gaps observed in this area, and Latin-American research groups currently evaluate that.
Article
Full-text available
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo–pituitary–adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Article
Asthma susceptibility is influenced by environmental, genetic and epigenetic factors. DNA methylation is one form of epigenetic modification that regulates gene expression and is both inherited and modified by environmental exposures throughout life. Prenatal development is a particularly vulnerable time period during which exposure to maternal asthma increases asthma risk in offspring. How maternal asthma affects DNA methylation in offspring, and what the consequences of differential methylation are in subsequent generations, is not fully known. In this study, we tested the effects of grandmaternal house dust mite (HDM) allergen sensitization during pregnancy on airway physiology and inflammation in HDM-sensitized and challenged second-generation mice. We also tested effects of grandmaternal HDM sensitization on tissue-specific DNA methylation in allergen-naïve and -sensitized second-generation mice. Descendants of both allergen- and vehicle-exposed grandmaternal founders exhibited airway hyperreactivity after HDM sensitization. However, grandmaternal allergen sensitization significantly potentiated airway hyperreactivity and altered the epigenomic trajectory in second-generation offspring after HDM sensitization compared to HDM-sensitized offspring from vehicle-exposed founders. As a result, biological processes and signaling pathways associated with epigenetic modifications were distinct between lineages. A targeted analysis of pathway-associated gene expression found that Smad3 was significantly dysregulated as a result of grandmaternal allergen sensitization. These data show that grandmaternal allergen exposure during pregnancy establishes a unique epigenetic trajectory that reprograms allergen responses in second-generation offspring and may contribute to asthma risk.
Article
Full-text available
The placenta is a temporary organ that is essential for the survival of the fetus, with a lifelong effect on the health of both the offspring and the dam. The functions of the placenta are controlled by its dynamic gene expression during gestation. In this study, we aimed to investigate the equine placental DNA methylome as one of the fundamental mechanisms that controls the gene expression dynamic. Chorioallantois samples from four (4M), six (6M), and ten (10M) months of gestation were used to map the methylation pattern of the placenta. Globally, methylation levels increased toward the end of gestation. We identified 921 differentially methylated regions (DMRs) between 4M and 6M, 1225 DMRs between 4M and 10M, and 1026 DMRs between 6M and 10M. A total of 817 genes carried DMRs comparing 4M and 6M, 978 comparing 4M and 10M, and 804 comparing 6M and 10M. We compared the transcriptomes between the samples and found 1381 differentially expressed genes (DEGs) when comparing 4M and 6M, 1428 DEGs between 4M and 10M, and 741 DEGs between 6M and 10M. Finally, we overlapped the DEGs and genes carrying DMRs (DMRs-DEGs). Genes exhibiting (a) higher expression, low methylation and (b) low expression, high methylation at different time points were identified. The majority of these DMRs-DEGs were located in introns (48.4%), promoters (25.8%), and exons (17.7%) and were involved in changes in the extracellular matrix; regulation of epithelial cell migration; vascularization; and regulation of minerals, glucose, and metabolites, among other factors. Overall, this is the first report highlighting the dynamics in the equine placenta methylome during normal pregnancy. The findings presented serve as a foundation for future studies on the impact of abnormal methylation on the outcomes of equine pregnancies.
Article
Aim: The placenta–brain axis reflects a developmental linkage where disrupted placental function is associated with impaired neurodevelopment later in life. Placental gene expression and the expression of epigenetic modifiers such as miRNAs may be tied to these impairments and are understudied. Materials & methods: The expression levels of mRNAs (n = 37,268) and their targeting miRNAs (n = 2083) were assessed within placentas collected from the ELGAN study cohort (n = 386). The ELGAN adolescents were assessed for neurocognitive function at age 10 and the association with placental mRNA/miRNAs was determined. Results: Placental mRNAs related to inflammatory and apoptotic processes are under miRNA control and associated with cognitive impairment at age 10. Conclusion: Findings highlight key placenta epigenome–brain relationships that support the developmental origins of health and disease hypothesis.
Article
Full-text available
Background: Small body size at birth has been reported to be associated with an atherogenic lipid profile in humans, and animal experiments have shown that undernutrition during pregnancy permanently alters cholesterol metabolism in the offspring. There is no direct evidence in humans that maternal malnutrition during pregnancy affects the lipid profiles of the offspring. Objectives: We assessed the effects of maternal malnutrition during specific periods of gestation on plasma lipid profiles in persons aged ≈50 y. Design: This was a follow-up study of men and women born at term as singletons in a university hospital in Amsterdam between 1 November 1943 and 28 February 1947 around the time of a severe famine. Results: Persons exposed to famine in early gestation had a more atherogenic lipid profile than did those who were not exposed to famine in utero. Their LDL-HDL cholesterol ratios were significantly higher (by 13.9%; 95% CI: 2.6–26.4%). Additionally, their plasma HDL-cholesterol and apolipoprotein A concentrations tended to be lower, and their plasma total cholesterol, LDL-cholesterol, and apolipoprotein B concentrations tended to be higher, although these differences were not statistically significant. The effect of famine was independent of size at birth and adult obesity. Conclusions: An atherogenic lipid profile might be linked to a transition from poor maternal nutrition in early gestation to adequate nutrition later on. This suggests that maternal malnutrition during early gestation may program lipid metabolism without affecting size at birth.
Article
Full-text available
Transgenerational effects of environmental toxins require either a chromosomal or epigenetic alteration in the germ line. Transient exposure of a gestating female rat during the period of gonadal sex determination to the endocrine disruptors vinclozolin (an antiandrogenic compound) or methoxychlor (an estrogenic compound) induced an adult phenotype in the F1 generation of decreased spermatogenic capacity (cell number and viability) and increased incidence of male infertility. These effects were transferred through the male germ line to nearly all males of all subsequent generations examined (that is, F1 to F4). The effects on reproduction correlate with altered DNA methylation patterns in the germ line. The ability of an environmental factor (for example, endocrine disruptor) to reprogram the germ line and to promote a transgenerational disease state has significant implications for evolutionary biology and disease etiology.
Article
Low birthweight is now known to be associated with increased rates of coronary heart disease and the related disorders stroke, hypertension and non-insulin-dependent diabetes. These associations have been extensively replicated in studies in different countries and are not the result of confounding variables. They extend across the normal range of birthweight and depend on lower birthweights in relation to the duration of gestation rather than the effects of premature birth. The associations are thought to be consequences of developmental plasticity, the phenomenon by which one genotype can give rise to a range of different physiological or morphological states in response to different environmental conditions during development. Recent observations have shown that impaired growth in infancy and rapid childhood weight gain exacerbate the effects of impaired prenatal growth. Coronary heart disease and the disorders related to it arise through a series of interactions between environmental influences and the pathways of development that preceded them. These diseases are the product of branching pathways of development in which the branchings are triggered by the environment before and after birth.
Chapter
In 1975, two papers suggested a role for DNA methylation in X chromosome inactivation. In one paper (Riggs, 1975), I argued that: 1) DNA methylation should affect protein-DNA interactions; 2) methylation patterns and a maintenance methylase should exist; and 3) DNA methylation should be involved in mammalian cellular differentiative processes. Holliday and Pugh (1975) argued similarly, although less weight was given to X inactivation and more weight was given to the possibility that 5-methylcytosine (5-meCyt) might be deaminated to thymidine; thus a specific mutational change would be generated, as suggested by Scarano (1971). Recently, several studies of X chromosome inactivation have contributed to the emerging body of evidence supporting a role for DNA methylation in mammalian gene regulation; it is these studies that will be reviewed in this chapter. More comprehensive reviews of X chromosome inactivation have been published recently (Gartler and Riggs, 1983; Graves, 1983).
Article
DES is the most carefully scrutinized EDC and its history provides valuable insights into the current evaluation of less well-studied EDCs. This review summarizes the health effects of prenatal exposure to diethylstilbestrol (DES) and emphasizes the role of DES as the first endocrine disrupting chemical (EDC). Vaginal clear cell adenocarcinoma (CCAC), the most severe consequence of prenatal exposure to DES, affected only 0.1% of exposed females, while the far more prevalent teratogenic and reproductive effects of DES were only discovered when DES daughter were screened for CCAC. Initial studies, conducted before most DES daughters had tried to conceive, examined vaginal cancer and vaginal, cervical and uterine abnormalities. Subsequently, several controlled studies demonstrated the increased risk of adverse reproductive outcomes in DES daughters. While most DES daughters can eventually experience a live birth, this is less likely in women with genital tract abnormalities, in whom there is a two-thirds chance that each pregnancy will be unsuccessful. In DES sons, who have been far less studied, results suggest male reproductive toxicity, but are less consistent. The importance of dose and gestational age at initial exposure are discussed, and the implications of DES findings for the evaluation of risks from current EDCs emphasized.
Article
• We tested the hypothesis that first-trimester exposure to acute food deprivation is a risk factor for schizophrenia. A sharp and time-limited decline in the food intake of the Dutch population following a Nazi blockade in 1944 to 1945 created a unique if tragic natural experiment to test this hypothesis in three regions of Holland (west, north, and south). In the west, or famine region, birth cohorts exposed to severe food deprivation (an average daily ration under 4200 kJ) during the first trimester showed a substantial increase in hospitalized schizophrenia for women but not for men. Relative risks for women were 2.17 for "broad" and 2.56 for "restricted" schizophrenia. Moderate food deprivation during the first trimester (average daily ration under 6300 kJ) was not associated with increased risk of schizophrenia in the famine region. In the north and south regions, numbers were smaller and there was no exposure to severe famine. Birth cohorts exposed to moderate food deprivation during the first trimester showed a trend toward increased risk of schizophrenia for women. These findings give plausibility to the proposition that early prenatal nutrition can have a gender-specific effect on the risk of schizophrenia.
Article
Adaptation to hypoxia is a topic of considerable clinical relevance, as it influences the pathophysiology of anaemia, polycythaemia, tissue ischaemia and cancer. A growing number of physiologically relevant genes are regulated in response to changes in intracellular oxygen tension. These include genes encoding erythropoietin, vascular endothelial growth factor and tyrosine hydroxylase. Studies on the regulation of the erythropoietin gene have provided insights into the common mechanism of oxygen sensing and signal transduction, leading to activation of the hypoxia‐inducible transcription factor 1 (HIF‐1). Activation of HIF‐1 by hypoxia depends on rescue of its α‐subunit from oxygen‐dependent degradation in the proteasome, allowing it to form a heterodimer with HIF‐1β. This then translocates to the nucleus. There, HIF‐1 assembles with a highly conserved orphan nuclear receptor, HNF‐4, and a critical transcriptional adaptor, p300. This complex binds to a 3′ enhancer on the erythropoietin gene, enabling transcription of erythropoietin. HIF‐1 also activates other genes, the cis‐acting elements of which contain cognate hypoxia response elements. There is growing evidence that the oxygen sensor is a flavohaem protein and that the signal transduction pathway involves changes in the level of intracellular reactive oxygen intermediates. We have recently cloned a novel fusion protein called cytochrome b5/b5 reductase, which is a cyanide‐insensitive NADPH oxidase and, therefore, a candidate to be the oxygen sensor. This flavohaem protein is widely expressed in cell lines and tissues, with localization in the perinuclear space. In the presence of oxygen and iron, it may induce oxidative modifications that target HIF‐1α for ubiquitination and degradation.