ArticlePDF AvailableLiterature Review

Maternal DNA Methylation During Pregnancy: a Review

Authors:

Abstract and Figures

Multiple environmental, behavioral, and hereditary factors affect pregnancy. Recent studies suggest that epigenetic modifications, such as DNA methylation (DNAm), affect both maternal and fetal health during the period of gestation. Some of the pregnancy-related risk factors can influence maternal DNAm, thus predisposing both the mother and the neonate to clinical adversities with long-lasting consequences. DNAm alterations in the promoter and enhancer regions modulate gene expression changes which play vital physiological role. In this review, we have discussed the recent advances in our understanding of maternal DNA methylation changes during pregnancy and its associated complications such as gestational diabetes and anemia, adverse pregnancy outcomes like preterm birth, and preeclampsia. We have also highlighted some major gaps and limitations in the area which if addressed might improve our understanding of pregnancy and its associated adverse clinical conditions, ultimately leading to healthy pregnancies and reduction of public health burden.
Content may be subject to copyright.
REVIEW
Maternal DNA Methylation During Pregnancy: a Review
Jagyashila Das
1
&Arindam Maitra
1
Received: 29 June 2020 / Accepted: 29 December 2020
#Society for Reproductive Investigation 2021
Abstract
Multiple environmental, behavioral, and hereditary factors affect pregnancy. Recent studies suggest that epigenetic modifica-
tions, such as DNA methylation (DNAm), affect both maternal and fetal health during the period of gestation. Some of the
pregnancy-related risk factors can influence maternal DNAm, thus predisposing both the mother and the neonate to clinical
adversities with long-lasting consequences. DNAm alterations in the promoter and enhancer regions modulate gene expression
changes which play vital physiological role. In this review, we have discussed the recent advances in our understanding of
maternal DNA methylation changes during pregnancy and its associated complications such as gestational diabetes and anemia,
adverse pregnancy outcomes like preterm birth, and preeclampsia. We have also highlighted some major gaps and limitations in
the area which if addressed might improve our understanding of pregnancy and its associated adverse clinical conditions,
ultimately leading to healthy pregnancies and reduction of public health burden.
Keywords Maternal .DNA methylation .Pregnancy .Pregnancy-related anemia .Gestational diabetes .Preterm birth .
Preeclampsia
Introduction
Pregnancy involves complex dynamic cascades of physiolog-
ical processes in the mother and the fetus. Both genetic and
environmental factors modulate pregnancy [1]. According to
the Developmental Origins of Health and Disease (DOHaD)
theory, which was based on epidemiological observations,
early-life environment, i.e., of the fetus, determines the health
trajectories in later life [2]. Women, who are malnourished
and/or underweight during pregnancy, are more likely to have
inadequate gestational weight gain leading to adverse impact
on fetal development [35]. On the other hand, maternal obe-
sity, high weight gain during pregnancy and gestational dia-
betes leads to increased risk of cancer, type 2 diabetes and
obesity in the offspring during adulthood [6,7].
Adverse birth outcomes have heterogeneous underlying
etiologies. Most adverse outcomes have familial aggregation
and have genetic underpinnings. Although multiple genome-
wide association studies (GWAS) have been undertaken to
identify genetic variants in adverse pregnancy outcomes, most
of these studies do not explain a substantial proportion of the
risk [811]. Epidemiologic observations suggest a role for
other factors like maternal obesity, stress, socioeconomic sta-
tus (SES), environmental conditions [12]. Studies also suggest
impact of early-life stress during the developmental stages on
adverse pregnancy outcomes [13]. These factors modulate the
cellular functioning primarily via DNA methylation (DNAm),
non-coding RNA, and/or histone modifications, of which
DNAm is the most widely studied phenomenon [1416].
Studies have only recently been initiated on pregnancy, birth,
and child health to investigate the impact of such factors on
the epigenome during gestation [1720].
The covalently bound methyl groups on 5cytosine sites
are not easily lost during routine DNA extraction, which ren-
ders DNAm as a reliable epigenomic marker which can be
utilized in large investigations [21]. Some DNAm marks are
inherited from parents, while others are acquired during the
lifetime of the individual [22,23]. The inherited marks can
persist for multiple generations, thus behave similar to DNA
mutations [24]. They may contribute to the missing heritabil-
ityof complex traits that are otherwise not explained by
DNA mutations alone [25]. On the other hand, dynamic
DNAm changes regulate the variable expression of the ge-
nome in diverse maternal tissues like the uterus, breasts, etc.
and in response to environmental factors from nulligravid to
postpartum state [26]. Orchestrated switching of DNAm
*Arindam Maitra
am1@nibmg.ac.in
1
National Institute of Biomedical Genomics, Kalyani, West
Bengal 741251, India
Reproductive Sciences
https://doi.org/10.1007/s43032-020-00456-4
marks across specific windows of gestation is key to healthy
pregnancy [2]. Recent evidences suggest that besides individ-
ual genome, maternal lifestyle shapes the trajectory of preg-
nancy with lasting impact onthe child [10]. One such example
is the Hunger Winter Families study, where it was found that
the adults who were prenatally exposed to famine, harbor
hypomethylation patterns in maternally imprinted insulin-
like growth factor II (IGF2) gene, a key factor in human
growth and development [2729]. Hence, studies on
epigenomics of pregnancy may reveal important information
on the interactions between the maternal as well as the fetal
genome and the environment, which may ultimately lead to
reduction in occurrences of adverse pregnancy outcomes [30].
DNAm plays a crucial role in gene expression regulation,
normal cellular function, and embryonic development [31].
Understanding DNAm changes during pregnancy would help
to explain the underlying biological mechanisms important for
physiological alterations and adaptations that are required for
fetal development, as well as to prepare the mother for child-
birth and postnatal period. In this review we have focused on
discussing the information available on DNAm in women
during gestation and its relationship to adverse clinical condi-
tions associated with pregnancy.
DNA Methylation in Normal Pregnancy
After conception, a pregnant woman undergoes substantial
physiological and anatomical changes throughout pregnancy
to nurture the developing fetus. This is followed by invasion
of the fetal trophoblast cells by epithelial to mesenchymal
transition (EMT) and migration within the endometrium, pro-
moting angiogenesis and establishing an exchange of nutri-
ents, gases and waste between fetus and mother [32,33].
Muller et al. compared methylation status of healthy controls
with breast cancer patients and pregnant women [34]. They
showed that methylation profiles of candidate genes (CDH1,
PTGS2,APC,RASSF1A) behaved similarly in both the preg-
nant group and the advanced cancer group. These genes show
similar DNAm pattern in trophoblasts, to that of invading
tumor, which is also a characteristic of progressive cancer
[35]. However, age is known to effect methylation status
and this study was limited by the lack of matching of age of
participants [36].
Natural killer cells and macrophages facilitate implantation
and placentation [37]. During this process and subsequently,
maternal leukocytes come into contact with the placental tro-
phoblast cells of paternal origin. Immune tolerance is induced
during pregnancy in order to avoid rejection of the
semiallogenic fetus [38,39]. Hence the first and early second
trimester of pregnancy is considered to be the inflammatory
phase [40,41]. Leukocytes play a pivotal role in proliferation,
invasion, inflammation, and immune tolerance, critical for
maternal adaptation for normal placental and fetal develop-
ment [42]. The modifications of the immune system altogether
make the uterus an immunity hotspot.Agenome-wide
DNAm study on maternal leukocytes found evidences of
global hypomethylation in pregnant state compared to two
non-pregnant states, i.e., before pregnancy and 6 months post-
partum. The study identified eight hypomethylated genes,
which are related to immunity (IL1R2,HPR,TREML1), ga-
metogenesis (SPAG4,CCIN), and a few housekeeping genes
(PC,NDFUS2)[43]. Involvement of interleukin-1 (IL1)fam-
ily of genes has been previously implicated in the establish-
ment and progression of pregnancy [44]. Hypomethylation of
haptoglobin-related protein (HPR) gene might lead to in-
creased HPR expression during normal pregnancy [45,46].
The study showed that transient methylation in maternal leu-
kocyte DNA during pregnancy might result in
immunotolerant adaptation, which is lost soon after delivery.
A study on maternal venous white blood cells (WBCs)
identified changes in methylation profiles of the LINE-1 ele-
ments (long interspersed nucleotide elements 1), which are
typically heavily methylated in normal conditions, but are
hypomethylated during cellular stress [47]. The study sug-
gested that hypermethylation of these elements in early preg-
nancy may lead to PTB [48]. Whether these alterations are
specific for pregnancy remains undetermined as data on
DNAm status of the LINE1 elements in women before and
after pregnancy is unreported.
In a recent longitudinal study, Gruzieva et al. compared
global epigenomic alterations between two non-pregnant (be-
fore conception and 24 days after delivery) states, and
through period of gestation (1014 weeks, 2628 weeks into
pregnancy) [49]. The per-pregnancy state was used as the
baseline for methylation levels, for comparison with later
stages of pregnancy. An overall decrease in methylation of
178 CpGs among the 196 significant sites during pregnancy
was observed. There was an overlap of about 47% of signif-
icantly associated probes among the discovery and replication
cohort, with most probes exhibiting the same direction of
change of DNAm level. Some of the pathways identified were
those which control various metabolic processes such as insu-
lin receptor signaling, mammary gland fat development, and
adipose tissue development in the body. However, this study
on peripheral blood from pregnant women did not account for
the contribution of circulating fetal cells in the detected meth-
ylation patterns, which are expected to affect the findings [50].
It was observed that SERPINB5 (maspin) promoter was
completely methylated in maternal blood, whereas it was
unmethylated in the placenta in case of normal pregnancy
outcomes [51]. In another study, RASSF1A gene was found
to be hypomethylated in maternal blood throughout gestation,
while it was hypermethylated in placenta [52]. In a study on
assessment of intergenerational impact of childhood maltreat-
ment (CM) on DNAm changes in peripheral blood
Reprod. Sci.
mononuclear cells (PBMCs) from mothers and neonates [53],
researchers identified adaptive immune cell specific DNAm
alteration in stress-response related genes like FKBP5,
CRHR1,andNR3C1 (involved in cortisol signaling), which
were however not transmitted from mothers with CM to neo-
nates directly after birth. These DNAm differences of the
same genes between the mother and the fetus might thereby
enable identification of fetal-specific markers for non-invasive
prenatal diagnosis.
Most methylation studies conducted so far are mostly
based on candidate genes in fetal (placental, cord blood) tis-
sues, emphasizing on disorders and co-morbidities of preg-
nancy, such as preeclampsia, in vitro fertilization, and gesta-
tional diabetes [5458]. However, identifying the genome-
wide differential methylation patterns in women during nor-
mal or healthy pregnancies might result in improved under-
standing of the genes and corresponding pathways that are
important for pregnancy. This might help us eventually to
improve our understanding of human pregnancies.
Pregnancy-Related Complications
and Maternal DNA Methylation
Gestational Diabetes
Altered secretion of hormones, such as cortisol, human pla-
cental lactogen insulin, estrogen and progesterone leads to
glucose-insulin imbalance during pregnancy, which results
in development of gestational diabetes (GD) [59,60]. In ad-
dition to these, a multitude of factors like ethnicity, polycystic
ovary syndrome, hypertension, etc. also contribute towards
GD in approximately 50% of the cases [61,62]. Most studies
of DNAm in GD have been undertaken in fetal tissues like the
cord blood and placenta [58,63]. Enquobahrie et al., in their
study on multigravidae women who have suffered from GD in
at least any one of the pregnancies, identified genes that were
hypermethylated (SEP11,ZAR1,DDR1) and those that were
hypomethylated (NDUFC,HAPLN3,HHLA3,RHOG)inma-
ternal peripheral blood mononuclear cells (PBMCs), as com-
pared to non-GD multigravidae women [64]. Interestingly,
one of the hypomethylated genes, NDUFC1, encodes for a
protein belonging to the complex I of the electron transport
chain located in the mitochondrial membrane [65]. It is known
that mitochondrial function in tissues like liver, muscles, and
pancreatic beta cells is critical for cellular metabolism and
modulation of oxidative activity and nutrient load [66,67].
However, roles of most of the other genes in pregnancy are
yet to be delineated.
Kang et al. compared global DNAm patterns of GD affect-
ed pregnant women and their children, with that of healthy
mother-offspring pairs, in blood samples drawn upon admis-
sion before delivery, and cord blood within short time interval
after delivery, in a cohort in Taiwan. The study identified 151
differentially methylated genes in GD mothers [68].
Association between GD and differentially methylated genes
belonging to carbohydrate and lipid metabolism pathways
have been previously reported [69]. This study also found
genes SLC22A4,ADRA1A,CACNB2,andSERPINE1 which
belong to the lipid metabolism pathways, to be differentially
methylated in GD mothers. Of these, ADRA1A has been re-
ported to be also involved in carbohydrate metabolism [70].
Alteration in DNAm of genes belonging to the GD related
metabolic pathways such as the Janus kinase (JAK)/mitogen
activated protein kinase (MAPK) pathways were also identi-
fied, which have been traditionally associated with endo-
toxins, inflammatory cytokines, and environmental stress
[68].
GD has been associated with macrosomia, large for gesta-
tional age, perinatal mortality, preeclampsia etc. and hence
there is a growing need to address it in the early stages of
pregnancy [71]. In an attempt to find clinical biomarkers in
women with GD, Wu et al., identified genome-wide DNAm
alterations in maternal peripheral whole blood collected from
pregnant women 1216 weeks into pregnancy, prior to diag-
nosis of gestational diabetes mellitus (GDM). They identified
two differentially methylated genes (HOOK2,RDH1) which
were also reported in other studies conducted on placenta and
cord blood [72,73]. HOOK2 codes for a linker protein that
mediates binding to organelles and is responsible for morpho-
genesis of cilia and endocytosis [74]. RDH12 encodes a retinal
reductase, which plays a role in the metabolism of short-chain
aldehydes [75]. Five additional CpG loci in COPS8,PIK3R5,
HAAO,CCDC124,andC5orf34 genes were also identified,
although the CpG locations in the genomic context were not
mentioned [76]. These genes have the potential to be used as
biomarkers once validated in larger validation cohorts.
Pregnancy-Related Anemia
Globally, 41.8% of pregnant women are anemic [77]. The mid
trimester is marked by rapid feto-placental growth and devel-
opment, with increased blood volume but lowered viscosity
and reduced hemoglobin concentration in the mother, typical-
ly causing anemia [7880]. There is a greater requirement of
folate during pregnancy, which is likely to be related to the
fetal growth and development [81]. It is known that folate-
mediated 1-carbon metabolism is essential for the high-
fidelity synthesis of DNA, maintenance of DNAm and regu-
lation of chromatin structure [82]. Knight et al., in 2018, in-
vestigated the association between one carbon metabolism
and DNAm changes during pregnancy in maternal peripheral
blood collected at < 12 weeks of pregnancy and at delivery
[83]. Out of the 993 CpG sites whose methylation levels in
maternal peripheral blood were found to be altered during
pregnancy, one site was found to be negatively correlated with
Reprod. Sci.
dimethylglycine (DMG) level in blood [83]. DMG is one of
the metabolites of the one carbon metabolism. Two CpGs
in PSMB7 and B4GALT5 genes were found to be associat-
ed with DMG concentrations in cord blood. Most of the
other CpG sites did not belong to any gene. This study
identified genome-wide significant metabolite-
methylation associations. However, whether DNAm was
regulated by metabolites or metabolites regulated DNAm
warrants further evaluation.
Maternal DNA Methylation in Adverse
Pregnancy Outcomes
Preterm Birth
Preterm birth (PTB), i.e., birth before 37 complete weeks of
gestation, is globally the leading cause of neonatal morbidity
[84,85]. Studies suggest that nutritional deficiencies, stress,
other factors such as cigarette smoking, maternal infection
(bacterial vaginosis/intra-amniotic infection), and race in-
crease the risk of PTB. Since some of these factors are envi-
ronmental in nature, it is likely that epigenetic mechanisms
such as DNAm may be involved in modulation of this risk
[86]. Multiple studies have been undertaken on PTB, primar-
ily in Caucasians and African Americans [8789]. A study on
African-American women from the Boston Birth Cohort iden-
tified association of altered DNAm of two CpG sites with
spontaneous PTB (sPTB), in maternal WBCs collected 24
72 h after delivery [90]. These CpG sites, one each in the
promoter regions of CYTIP (cytohesin 1 interacting protein)
and LINC0114 (long non-coding RNA) genes, were
hypomethylated in mothers who delivered early sPTB (24
33.6/7 weeks) compared to those who delivered at term (39
42 weeks). CYTIP is involved in leukocyte trafficking, T cell
receptor mediated signaling and labor, events which are
known to play key roles in pregnancy [9194]. LINC0114 is
known to mediate cell differentiation and immune response,
which are important for pregnancy [9597]. These associa-
tions with sPTB were observed only in maternal blood but
not in cord blood.
In another study on an US based African-American cohort
which analyzed genome-wide DNAm in maternal leukocytes,
two CpG sites were found to be significantly associated with
early PTB (24.134.0 weeks) [98]. Both of these sites
belonged to the regulatory-associated protein of MTOR
(RPTOR), component of a signaling pathway that regulates
cell growth in response to nutrient and insulin levels [99,
100]. This pathway has been previously implicated in
myometrial proliferation during pregnancy [101]. The study
also identified 5171 CpGs whose fetal methylation status
could be predicted from maternal signatures on those sites
(98.8% in the same direction). In an earlier study on a multi-
generational Caucasian cohort, about 75% of these sites were
found to be either single nucleotide polymorphisms (SNPs) or
methylation quantitative trait loci (meQTL) of multiple heri-
table traits, which suggested that most of these heritable CpG
sites might be under genetic influence [102].
Previously, genetic variants in matrix metalloprotease 1
gene (MMP1) were found to be associated with preterm pre-
mature rupture of the membranes (pPROM) [103]. In 2008, a
study reported association of promoter hypomethylation of a
SNP in MMP1 in fibroblast cells from the amnion of mothers
with pPROM, leading to sPTB [104].Thecombinedgenetic
and DNAm factors might determine MMP1 expression and
influence the association with pPROM. Such combinatorial
studies would help to identify birth outcome specific bio-
markers in pregnant women.
PTB is a complex phenotype of heterogeneous etiologies
[105]. In a recent study based on multi-omics approach in-
volving GWAS, RNA-Seq and Epigenome-wide association
(EWAS) using maternal blood collected after delivery,
Knijnenberg et al. compared full term birth (FTB, 37 to <
42 weeks) with very early PTB (VEPTB, < 28 weeks) and
early PTB (< 34 weeks) [106]. They found association of
RAB31 and RBPJ genes with PTB using all three platforms.
RAB31 is a member of the RAS oncogene family coding for a
small GTPase-binding protein, upregulated at full term com-
pared to midway through gestation which also has been im-
plicated in a GWAS on placental abruption [107,108]. RBPJ
is hypermethylated in VEPTB compared to FTB and is a
transcriptional regulator in the Notch signaling pathway
[109]. In addition to such studies, combining longitudinal epi-
genetic data with genetic data might help establish the causal
relationship of associated genes.
Investigation of temporal changes in DNAm coupled with
transcriptomic alterations in pregnant women, along with pre-
cise measurement of POG using ultrasound and metabolites,
might help to improve our understanding of PTB and hence
reduce health burdens associated with PTB outcomes.
Preeclampsia
Preeclampsia (PE) is a major cause of PTB and maternal and
infant mortality, characterized by hypertension and protein-
uria after 20 weeks of healthy pregnancy [110,111]. PE can
also lead to intrauterine growth restriction (IUGR) of fetus,
leading to PTB [112]. PE is associated with oxidative stress
and obesity, both of which are known to influence DNAm
[113116]. Substantial research has been conducted on the
epigenetics of PE, primarily on the placenta and cell-free fetal
(cff) DNA in maternal blood [51,117120]. Chim et al. ob-
served 5.7-fold higher abundance of unmethylated MASPIN
in maternal plasma from mothers with preeclampsia compared
to those with normal pregnancy outcome [51]. Tsui et al.
found elevated level of hypermethylated fraction of
Reprod. Sci.
RASSF1A gene in maternal plasma derived from cff DNA [120].
The abundance of cff DNA in maternal plasma of PE women are
most likely due to placental apoptosis, a characteristic of PE; or
due to reduced clearance of cff DNA from maternal system
[121123]. RASSF1A is primarily a tumor suppressor, mostly
found to be inactivated by promoter hypermethylation in many
tumor types [124]. It is also found to be hypermethylated in
placenta in complicated pregnancies [125]. In a study conducted
on maternal peripheral blood, placenta, and umbilical cord blood,
promoter hypermethylation of soluble-cytoplasmic COMT (s-
COMT) was detected, whereas, it was found hypomethylated
in PE-specific placenta [56]. Promoter hypomethylation in the
placenta may indicate facilitated interaction of transcription factor
GATA-binding protein 2 (GATA2) and E1A-binding protein
p300 (EP300) with the S-COMT promoter which play a role in
placental development and the homeostasis of placental oxygen
tension [126]. The above tissue-specific methylation changes
might serve as biomarkers of early prediction of PE in pregnant
women.
In 2012, Mousa et al. reported hypomethylation of throm-
boxane synthase gene (TBXAS1) in systemic omental blood
vessels of mothers with PE [127]. TBXAS1 codes for an en-
zyme that catalyzes isomerization of prostaglandin H2 into
thromboxane [128]. Elevated thromboxane and decreased
prostacyclin acts as markers of PE [129,130].
Hypomethylation of TBXAS1 may contribute to the pathogen-
esis of PE and may increase maternal risk of cardiovascular
diseases later in life.
Anderson et al. 2013, identified 133 hypermethylated CpG
sites corresponding to 71 genes, out of 207 differentially meth-
ylated CpGs from peripheral WBCs during first trimester of
preeclamptic mothers, compared to normotensive (normal
blood pressure) mothers [131]. Seven of these belonged to the
pleckstrin homology domain, which although small in size, but
are present in a large variety of signaling proteins, serving as
lipid binding domains, and protease inhibitors. The remaining
74 CpGs from 38 genes were hypomethylated, and associated
with cellular metabolism, endothelin signaling, T cell activa-
tion, insulin signaling, progesterone-related oocyte maturation,
immune injury, phospholipase C-epsilon, G protein signaling,
encephalin release, and metabotropic glutamate receptor group
1 pathway. The same study identified thirteen differentially
methylated CpG sites as novel putative biomarkers of early
detection of PE (in the first trimester) in mothers [131]. These
genes play important roles in pregnancy, miscarriage, implan-
tation, and immune tolerance. KH homology domain and
stathmin family of genes play important roles in pregnancy
and implantation [132]. E2 class of ubiquitin conjugating en-
zyme has previously been implicated in early miscarriage,
CD80 in maternal immune tolerance to the fetus, RAP1A pro-
tein in GDM [133135].
Longitudinal characterization of the PE phenotype (early-
onset PE, or late-onset PE) and its epigenomic underpinnings
is an area yet to be investigated in depth. Studies on identifi-
cation of epigenetic predisposition for PE in the maternal sys-
tem might enable early medical intervention during pregnancy
to reduce incidence of PE.
Limitations and Future Directions
We have summarized the studies in Table 1. The genes and
associated CpG sites along with their location (if discussed in
the study) are provided in Supplementary Table 1.Moststud-
ies on pregnancy were aimed to capture differences between
normal and extreme phenotypes. However, causal association
of genes with the reported outcomes are not always conclu-
sive. There is a paucity of understanding of the DNAm chang-
es that occur during the sequential events in pregnancy.
Although intrauterine growth restriction (IUGR) is an impor-
tant adverse condition of pregnancy, there has been no study
conducted on the maternal DNA methylation changes in
IUGR till date. However, multiple studies have implicated
maternal factors like hypertension, maternal substance abuse,
age, GDM, and anemia that can lead to this outcome [112,
136].
Multiple epidemiological and genetic risk factors are associ-
ated with pregnancy and its outcomes. Environmental, behav-
ioral, nutritional, and psycho-social conditions also modulate
pregnancy [137]. Hence, there is a burgeoning need to study
the genetic and epigenetic factors like DNAm from minimally
invasive maternal tissue like whole blood, which could help
monitor feto-maternal conditions during pregnancy and subse-
quently the health of the neonate. Blood provides a unique
potential window into the health and phenotype of the individ-
ual and hence longitudinal epigenetic profiling of peripheral
blood DNA from pregnant women during pregnancy and after
delivery might provide improved insights into the physiological
changes taking place. This might serve to identify potential
predictive biomarkers of pregnancy outcomes [138].
Studies have improved our understanding of early-life
stressors on women, especially during pregnancy.
Epigenome-wide studies across mother-offspring dyads are
required in the future to find the impact of maternal early-
life stress induced specific DNAm marks on pregnancy and
neonatal health. This approach would illuminate the impact of
early-lifepsycho-social factors on pregnancy and how it might
prospectively modulate the potential transmission of such
stressors on neonates.
There is a general limitation of most studies based on
whole blood DNAm. Whole blood is composed of multiple
cell types, and each cell type contributes to CpG locus specific
DNA methylation signal. Research on DNAm in whole
blood conducted until date has not yet fully addressed the
issue of cellular heterogeneity. Although various methods ex-
ist for adjusting for cellular heterogeneity, they commonly use
Reprod. Sci.
Table 1 Summary of the maternal DNA methylation studies discussed in the review
Study design Approach Phenotype (sample size) Time points Tissue Cell types Main findings References
Case-control Candidate
gene
Healthy pregnancy (32),
eclampsia, PE, HELLP
syndrome (17), healthy
controls (10), primary
breast cancer (26),
metastasized advance
breast cancer (10)
1015 weeks Maternal sera - CDH1,PTGS2,APC,
RASSF1A behave
similar in pregnant
and cancer groups
Muller, 2004 (32)
Healthy pregnancy (20),
PE (10)
At delivery Maternal peripheral
blood, placenta
- Hypermethylated RASSF1A
were 4.3-fold higher in
maternal plasma of PE
subjects than in controls
Tsui, 2007 (116)
PPROM (284), healthy
pregnancy (361)
At delivery Amnion Fibroblast promoter hypomethylation
of MMP1 in amnion of
pPROM patients lead
to sPTB
Wang, 2008 (100)
Childhood maltreatment
(CM+) mother (58):
infant (55), CM mother
(59): infant(58)
After delivery Maternal peripheral
blood, umbilical
blood mononuclear
cells
Peripheral blood
mononuclear
cells (PBMC)
FKBP5,CRHR1:CM+
mothers show less
methylation than CM
mothers, NR3C1:CM+
mothers show more
methylation than CM
mothers
Ramo-Fernández (53)
Genome
wide
Healthy pregnancy (6),
gestational diabetes (6)
~16 weeks Maternal peripheral
blood
Peripheral blood
mononuclear
cells (PBMC)
Hypomethylated (NDUFC1,
HAPLN3,HHLA3,RHOG),
hypermethylated (SEPT11,
ZAR1,DDR)
Enquobahrie,
2015 (61)
Healthy pregnancy (8),
PE (8)
1st trimester,
3rd trimester
Maternal peripheral blood
Maternal plasma, placenta
- methylated MASPIN in
maternal plasma
Chim, 2005 (49)
Healthy pregnancy (14),
nulligravid (14)
715 weeks, 68
months postpartum
Maternal peripheral blood Leukocytes IL1R2,HPR,TREML1,
SPAG4,CCIN,PC,
NDUFS2,AGAP4/CTGLF1
hypomethylated in
pregnant state
White, 2012 (41)
Healthy pregnant women
(5), PE cases (7)
At delivery Maternal omental arteries - 65 genes were identified,
which showed reduced
methylation in preeclampsia
Mousa, 2012 (123)
Nulliparous (55), PE (6) 1st trimester, At
delivery
Maternal peripheral
blood, placental
chorionic tissue
White blood
cells
12 genes with significant
differences in mean
methylation in maternal
white blood cells,
putative biomarkers
Anderson, 2014 (127
GDM (11), healthy
pregnancy (11)
1216 weeks Maternal peripheral
blood
-COPS8,PIK3R5,HAAO,
CCDC124,C5orf34 as
potential clinical
biomarkers
Wu, 2018 (73)
Reprod. Sci.
Table 1 (continued)
Study design Approach Phenotype (sample size) Time points Tissue Cell types Main findings References
Preterm delivery (16),
term delivery (24)
Time of admission
for labor
Maternal peripheral b
lood
Leukocytes 5171 CpGs maternal
methylation associated
with fetal methylation,
98.8% of which occurred
in the same direction
Parets, 2015 (94)
Discovery set: early sPTB
delivery (150), term
delivery (150); validation
set: (mother-child dyads)
early sPTBs (7 pairs), late
sPTBs (34 pairs), medically
indicated PTBs (14 pairs),
term birth (55 pairs)
2472 h after delivery Maternal peripheral
blood, cord blood
White blood
cells
CYTIP,LINC00114
methylation lower in
mothers with early
sPTBs than TB
Hong, 2018 (86)
Healthy mother-child (8 pairs),
GDM mother-child (8 pairs)
Before delivery,
minutes after delivery
Maternal peripheral
blood, cord blood
- Differential methylation
SLC22A4,ADRA1A,
CACNB2,SERPINE1
Kang, 2017 (67)
Healthy pregnancy (21),
PE (16)
At delivery Maternal plasma,
placenta
- Placenta-specific s-COMT
promoter, a potential
marker for early
prediction of PE in
maternal plasma
Zhao, 2010 (53)
Observation,
cross-species
comparison
Candidate
gene
Healthy pregnancy (5),
pregnant mice, rhesus
monkeys
1st trimester, 3rd
trimester, at delivery
Maternal peripheral
blood, Placenta
-RASSF1A
hypermethylated in
placenta, unmethylated
in maternal blood cells
Chiu, 2007 (121)
Cross-sectional Candidate gene Healthy pregnancy (1160) 1st trimester, 2nd
trimester, At delivery
Maternal venous
blood
Venous cord blood
White blood
cells
LINE-1 elements
hypermethylation
Burris, 2012 (46)
Genome wide Healthy pregnancy (24) <12 weeks, at delivery Maternal peripheral
blood, cord blood
-PSMB7,B4GALT5
associated with DMG
concentrations in
cord blood
Knight, 2018 (79)
Longitudinal Genome wide Non-pregnant state, pregnant
state, postpartum (21)
1014 weeks,
2628 weeks,
24daysafter
delivery
Maternal peripheral
blood
- 91% of 196 significant
probes showed decrease
in methylation levels
across pregnancy.
Gruzieva, 2019 (47)
Multi-omics
integrative
study
Genome wide PTB (270 family trios), term
birth (521 family trios),
VEPTB, pPROM, PE,
uterine anomalies and
endometriosis, cervix
related, and idiopathic
PTB
14daysafter
delivery
Whole blood - RAB31 and RBPJ had
significant associations
found for all three data
types (WGS, DNA
methylation, and
mRNA expression)
Knijnenburg,
2019 (102)
Reprod. Sci.
a reference methylation data, based on which the sample cell
proportions are estimated [139]. Other bioinformatic tools for
reference-free cell type estimation mostly rely on prior infor-
mation on cell type-specific marks of specific cells from
existing database and infer cell proportion based on this infor-
mation [140,141]. These methods do not fully account for the
cell type diversity of study samples under question. DNAm-
based deconvolution of selected cell type-specific differential-
ly methylated regions (DMRs) from sorted and purified cell
populations of individual subjects are yet to be investigated.
Moreover, during pregnancy, maternal blood also contains
fetal genetic materials, and thus there remains a necessity to
account for both to exclusively parse the maternal component.
Studies have been conducted to investigate genetic as well as
other epigenetic processes such as histone modifications, non-
coding RNA, etc., on pregnancy and adverse pregnancy out-
comes [142144]. Integration of such approaches with DNAm
studies would enable holistic understanding of pregnancy phe-
notype at the cellular and systemic level. Analysis of
biospecimens collected prior to conception, during gestation
and after delivery will help to identify unique DNAm changes
occurring specifically due to pregnancy. Prospective cohorts
should therefore be used to capture dynamic epigenetic modifi-
cations during pregnancy [145]. Since pregnancy-related compli-
cations owe its etiology to various factors such as gene-gene,
gene-environment interaction, etc. a multidimensional approach
towards pregnancy can address the effect-size of epistatic as well
as pleiotropic effects of genes [146]. A multi-omics approach
should help unravel the complex events of pregnancy which
leads to diverse outcomes. Such an approach could lead to re-
purposing of existing as well as discovery of novel therapeutic
agents, for clinical intervention in case of adversity. Designing a
prospective study cohort which captures relevant clinical, SES,
and demographic data would help to develop a holistic under-
standing of pregnancy. Cohorts like that of the Group for
Advanced Research on Birth OutcomesDBT India Initiative
(GARBH-Ini) from India have taken into account the clinical,
SES, genomics, epigenomics, metagenomics, microbial and pro-
teomic correlates of pregnancy for integrated analysis [147].
Such longitudinal cohort studies can provide platforms which
can facilitate improved understanding of the pregnancy and its
outcomes. They may be better poised to address the epigenomics
of pregnancy, primarily DNAm landscape in mothers through
multiple time points during pregnancy and at postpartum, than
cross-sectional studies. Such strategy of following up mothers
would help identify pregnancy specific alterations, as well as
provide predictive markers for maternal health.
Conclusion
We have reviewed studies conducted on changes in maternal
DNA methylation during pregnancy and its associated
adverse conditions. Exploration of epigenomic changes can
provide improved understanding of the dynamic biological
processes that take place during pregnancy. This would allow
clinicians to identify women at increased risk for adverse
pregnancy outcomes and develop precise, individualized
risk-specific interventions.
Supplementary Information The online version contains supplementary
material available at https://doi.org/10.1007/s43032-020-00456-4.
Acknowledgments We thank Professor Partha Pratim Majumder for his
mentorship, advice and suggestions.
Funding JD is supported by the Research Fellowship (NET) of the
University Grants Commission (UGC), India.
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
Ethical Consents This is a reviewarticle and does not involve collection
of human biospecimens or analysis. The review is based on published
information. Hence no ethical consent is required.
References
1. Dadvand P, Parker J, Bell ML, BonziniM, Brauer M, Darrow LA,
et al. Maternal exposure to particulate air pollution and term birth
weight: a multi-country evaluation of effect and heterogeneity.
Environ Health Perspect. 2013;121:267373.
2. Barker DJ. The origins of the developmental origins theory. J
Intern Med. 2007;261:4127.
3. Carmichael SL, Yang W, Shaw GM, others. Maternal dietary
nutrient intake and risk of preterm delivery. Am J Perinatol.
2013;30:57988.
4. Rich-Edwards JW, Colditz GA, Stampfer MJ, Willett WC,
Gillman MW, Hennekens CH, et al. Birthweight and the risk for
type 2 diabetes mellitus in adult women. Ann Intern Med.
1999;130:27884.
5. Susser ES, Lin SP. Schizophrenia after prenatal exposure to the
Dutch Hunger Winter of 1944-1945. Arch Gen Psychiatry.
1992;49:9838.
6. Michels KB, Xue F. Role of birthweight in the etiology of breast
cancer. Int J Cancer. 2006;119:200725.
7. Stuebe AM, Forman MR, Michels KB. Maternal-recalled gesta-
tional weight gain, pre-pregnancy body mass index, and obesity in
the daughter. Int J Obes. 2009;33:74352.
8. Johnson MP, Brennecke SP, East CE, et al. Genome-wide associ-
ation scan identifies a risk locus for preeclampsia on 2q14, near
the inhibin, beta b gene. PLoS One. 2012;7:e33666.
9. Hong X, Hao K, Ji H, Peng S, Sherwood B, di Narzo A, et al.
Genome-wide approach identifies a novel gene-maternal pre-
pregnancy bmi interaction on preterm birth. Nat Commun.
2017;8:15608.
10. Rappoport N, Toung J, Hadley D, Wong RJ, Fujioka K, Reuter J,
et al. A genome-wide association study identifies only two ances-
try specific variants associated with spontaneous preterm birth. Sci
Rep. 2018;8:226.
Reprod. Sci.
11. Zhang YP, Liu XH, Gao SH, Wang JM, Gu YS, Zhang JY, et al.
Risk factors for preterm birth in five maternal and child health
hospitals in Beijing. PLoS One. 2012;7:e52780.
12. Nieuwenhuijsen MJ, Dadvand P, Grellier J, Martinez D, Vrijheid
M. Environmental risk factors of pregnancy outcomes: a summary
of recent meta-analyses of epidemiological studies. Environ
Health. 2013;12:6.
13. Rogac M, Peterlin B. Epigenetic signature of chronic maternal
stress load during pregnancy might be a potential biomarker for
spontaneous preterm birth. Balkan J Med Genet. 2018;21:2733.
14. Hoyo C, Murphy SK, Jirtle RL (2009) Imprint regulatory elements
as epigenetic biosensors of exposure in epidemiological studies.
15. Relton CL, Smith GD. Epigenetic epidemiology of common com-
plex disease: prospects for prediction, prevention, and treatment.
PLoS Med. 2010;7:e1000356.
16. Dolinoy DC, Weidman JR, Jirtle RL. Epigenetic gene regulation:
linking early developmental environment to adult disease. Reprod
Toxicol. 2007;23:297307.
17. Groom A, Elliott H, Embleton N, Relton C. Epigenetics and child
health: basic principles. Arch Dis Child. 2011;96:8639.
18. Cao-Lei L, Dancause KN, Elgbeili G, Laplante DP, Szyf M, King
S. DNA methylation mediates the effect of maternal cognitive
appraisal of a disaster in pregnancy on the childs c-peptide secre-
tion in adolescence: Project Ice Storm. PLoS One. 2018;13:
e0192199.
19. Suter MA, Aagaard-Tillery KM. Environmental influences on epi-
genetic profiles. Semin Reprod Med. 2009;27:38090.
20. Feinberg AP. Epigenetics at the epicenter of modern medicine.
Jama. 2008;299:134550.
21. Talens RP, Boomsma DI, Tobi EW, Kremer D, Jukema JW,
Willemsen G, et al. Variation, patterns, and temporal stability of
DNA methylation: considerations for epigenetic epidemiology.
FASEB J. 2010;24:313544.
22. Kim M, Costello J. DNA methylation: an epigenetic mark of cel-
lular memory. Exp Mol Med. 2017;49:e322.
23. Duncan EJ, Gluckman PD, Dearden PK. Epigenetics, plasticity,
and evolution: how do we link epigenetic change to phenotype? J
Exp Zool B Mol Dev Evol. 2014;322:20820.
24. McCarthy MI, Hirschhorn JN. Genome-wide association studies:
potential next steps on a genetic journey. Hum Mol Genet.
2008;17:R15665.
25. Maher B. Personal genomes: the case of the missing heritability.
Nature News. 2008;456:1821.
26. Huh SJ, Clement K, Jee D, Merlini A, Choudhury S, Maruyama
R, et al. Age-and pregnancy-associated dna methylation changes
in mammary epithelial cells. Stem cell reports. 2015;4:297311.
27. Lumey L, Stein AD, Kahn HS, Van der Pal-de Bruin KM, Blauw
G, Zybert PA, et al. Cohort profile: the Dutch Hunger Winter
families study. Int J Epidemiol. 2007;36:1196204.
28. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser
ES, et al. Persistent epigenetic differences associated with prenatal
exposure to famine in humans. Proc Natl Acad Sci. 2008;105:
170469.
29. Smith F, Garfield A, Ward A. Regulation of growth and metabo-
lism by imprinted genes. Cytogenetic and genome research.
2006;113:27991.
30. Jirtle RL, Skinner MK. Environmental epigenomics and disease
susceptibility. Nat Rev Genet. 2007;8:25362.
31. (2020) DNA methylation, an epigenetic mode of gene expression
regulation in reproductive science. Harvard Catalyst Profiles
Harvard Catalyst.
32. Bischof P. Trophoblast differentiation and invasion: its signifi-
cance for human embryo implantation. Early Pregnancy. 1997;3:
81.
33. Kim S-M, Kim J-S. A review of mechanisms of implantation.
Development & reproduction. 2017;21:3519.
34. Müller HM, Ivarsson L, Schröcksnadel H, et al. DNA methylation
changes in sera of women in early pregnancy are similar to those
in advanced breast cancer patients. Clin Chem. 2004;50:10658.
35. De Craene B, Berx G. Regulatory networks defining EMT during
cancer initiation and progression. Nat Rev Cancer. 2013;13:97
110.
36. Xiao F-H, Wang H-T, Kong Q-P (2019) Dynamic DNA methyl-
ation during aging: a prophetof age-related outcomes. Frontiers
in genetics 10:
37. Mor G, Cardenas I, Abrahams V, Guller S. Inflammation and
pregnancy: the role of the immune system at the implantation site.
Ann N Y Acad Sci. 2011;1221:807.
38. Erlebacher A. Immunology of the maternal-fetal interface. Annu
Rev Immunol. 2013;31:387411.
39. Svensson-Arvelund J, Ernerudh J, Buse E, Cline JM, Haeger J-D,
Dixon D, Markert UR, Pfarrer C, Vos PD, Faas MM (2014) The
placenta in toxicology. Part ii: systemic and local immune adap-
tations in pregnancy. Toxicologic pathology 42:327338.
40. Romero R. Novel aspects of neutrophil biology in human preg-
nancy. Am J Reprod Immunol. 2005;53:275.
41. PetrušićV, ŽivkovićI, Muhandes L, DimitrijevićR, Stojanović
M, DimitrijevićL. Infection-induced autoantibodies and pregnan-
cy related pathology: an animal model. Reprod Fertil Dev.
2014;26:57886.
42. Veenstra van Nieuwenhoven A, Heineman M, Faas M. The im-
munology of successful pregnancy. Hum Reprod Update. 2003;9:
34757.
43. White WM, Brost BC, Sun Z, Rose C, Craici I, Wagner SJ, et al.
Normal early pregnancy: a transient state of epigenetic change
favoring hypomethylation. Epigenetics. 2012;7:72934.
44. Herrmann-Lavoie C, Rao C, Akoum A. Chorionic gonadotropin
down-regulates the expression of the decoy inhibitory interleukin
1 receptor type ii in human endometrial epithelial cells.
Endocrinology. 2007;148:537784.
45. Kuhajda FP, Piantadosi S, Pasternack GR. Haptoglobin-related
protein (hpr) epitopes in breast cancer as a predictor of recurrence
of the disease. N Engl J Med. 1989;321:63641.
46. Hoffman L, Winfrey V, Blaeuer G, Olson G. A haptoglobin-like
glycoprotein is produced by implantation-stage rabbit endometri-
um. Biol Reprod. 1996;55:17684.
47. Wongpaiboonwattana W, Tosukhowong P, Dissayabutra T,
Mutirangura A, Boonla C. Oxidative stress induces hypomethy-
lation of line-1 and hypermethylation of the runx3 promoter in a
bladder cancer cell line. Asian Pac J Cancer Prev. 2013;14:3773
8.
48. Burris HH, Rifas-Shiman SL, Baccarelli A, Tarantini L, Boeke
CE, Kleinman K, et al. Associations of LINE-1 DNA methylation
with preterm birth in a prospective cohort study. J Dev Orig Health
Dis. 2012;3:17381.
49. Gruzieva O, Merid SK, Chen S, et al. DnA methylation trajecto-
ries during pregnancy. Epigenetics insights. 2019;12:
2516865719867090.
50. Poon LL, Leung TN, Lau TK, Chow KC, Lo YD. Differential
DNA methylation between fetus and mother as a strategy for de-
tecting fetal DNA in maternal plasma. Clin Chem. 2002;48:35
41.
51. Chim SS, Tong YK, Chiu RW, Lau TK, Leung TN, Chan LY,
et al. Detection of the placental epigenetic signature of the maspin
gene in maternal plasma. Proc Natl Acad Sci. 2005;102:147538.
52. Chiu RW, Chim SS, Wong IH, et al. Hypermethylation of rassf1a
in human and rhesus placentas. Am J Pathol. 2007;170:94150.
53. Ramo-Fernández L, Boeck C, Koenig AM, Schury K, Binder EB,
Gündel H, et al. The effects of childhood maltreatment on epige-
netic regulation of stress-response associated genes: an intergen-
erational approach. Sci Rep. 2019;9:112.
Reprod. Sci.
54. Bellido ML, Radpour R, Lapaire O, Bie ID, Hösli I, Bitzer J, et al.
MALDI-tof mass array analysis of rassf1a and serpinb5 methyla-
tion patterns in human placenta and plasma. Biol Reprod.
2010;82:74550.
55. Kulkarni A, Chavan-Gautam P, Mehendale S, Yadav H, Joshi S.
Global DNA methylation patterns in placenta and its association
with maternal hypertension in pre-eclampsia. DNA Cell Biol.
2011;30:7984.
56. Zhao A, Cheng Y, Li X, Li Q, Wang L, Xu J, et al. Zhao X. Mol
Hum Reprod. 2010;17:199206.
57. Katari S, Turan N, Bibikova M, Erinle O, Chalian R, Foster M,
et al. DNA methylation and gene expression differences in chil-
dren conceived in vitro or in vivo. Hum Mol Genet. 2009;18:
376978.
58. Bouchard L, Thibault S, Guay S-P, Santure M, Monpetit A, St-
Pierre J, et al. Leptin gene epigenetic adaptation to impaired glu-
cose metabolism during pregnancy. Diabetes Care. 2010;33:
243641.
59. Velegrakis A, Sfakiotaki M, Sifakis S. Human placental growth
hormone in normal and abnormal fetal growth. Biomed Rep.
2017;7:11522.
60. Masuyama H, Hiramatsu Y. Potential role of estradiol and proges-
terone in insulin resistance through constitutive androstane recep-
tor. J Mol Endocrinol. 2011;47:22939.
61. de Barros MC, Lopes MA, Francisco RP, Sapienza AD, Zugaib
M. Resistance exercise and glycemic control in women with ges-
tational diabetes mellitus. Am J Obstet Gynecol. 2010;203:556
e1.
62. Marion D (2008) Screening and diagnosis of gestational diabetes
mellitus. UpToDate web site.
63. Weng X, Liu F, Zhang H, Kan M, Wang T, Dong M, et al.
Genome-wide DNA methylation profiling in infants born to ges-
tational diabetes mellitus. Diabetes Res Clin Pract. 2018;142:10
8.
64. Enquobahrie DA, Moore A, Muhie S, Tadesse MG, Lin S,
Williams MA. Early pregnancy maternal blood DNA methylation
in repeat pregnancies and change in gestational diabetes mellitus
statusa pilot study. Reprod Sci. 2015;22:90410.
65. Loeffen J, Smeets R, Smeitink J, Triepels R, Sengers R, Trijbels F,
et al. The human NADH: ubiquinone oxidoreductase ndufs5
(15kDa) subunit: CDNA cloning, chromosomal localization, tis-
sue distribution and the absence of mutations in isolated complex
i-deficient patients. J Inherit Metab Dis. 1999;22:1928.
66. Patti M-E, Corvera S. The role of mitochondria in the pathogene-
sis of type 2 diabetes. Endocr Rev. 2010;31:36495.
67. Qiu C, Enquobahrie DA, Frederick IO, Sorensen TK, Fernandez
MAL, David RM, et al. Early pregnancy urinary biomarkers of
fatty acid and carbohydrate metabolism in pregnancies complicat-
ed by gestational diabetes. Diabetes Res Clin Pract. 2014;104:
393400.
68. Kang J, Lee C-N, Li H-Y, Hsu K-H, Lin S-Y. Genome-wide DNA
methylation variation in maternal and cord blood of gestational
diabetes population. Diabetes Res Clin Pract. 2017;132:12736.
69. Butte NF. Carbohydrate and lipid metabolism in pregnancy: nor-
mal compared with gestational diabetes mellitus. Am J Clin Nutr.
2000;71:1256S61S.
70. Milano W, De Rosa M, Milano L, Capasso A. Antipsychotic
drugs opposite to metabolic risk: neurotransmitters, neurohormon-
al and pharmacogenetic mechanisms underlying with weight gain
and metabolic syndrome. The open neurology journal. 2013;7:23
31.
71. Wendland EM, Torloni MR, Falavigna M, Trujillo J, Dode MA,
Campos MA, et al. Gestational diabetes and pregnancy outcomes
a systematic review of the World Health Organization (WHO) and
the International Association of Diabetes in Pregnancy Study
Groups (IADPSG) diagnostic criteria. BMC Pregnancy
Childbirth. 2012;12:23.
72. Ruchat S-M, Houde A-A, Voisin G, St-Pierre J, Perron P,
Baillargeon J-P, et al. Gestational diabetes mellitus epigenetically
affects genes predominantly involved in metabolic diseases.
Epigenetics. 2013;8:93543.
73. Finer S, Mathews C, Lowe R, Smart M, Hillman S, Foo L, et al.
Maternal gestational diabetes is associated with genome-wide
DNA methylation variation in placenta and cord blood of exposed
offspring. Hum Mol Genet. 2015;24:30219.
74. Baron Gaillard CL, Pallesi-Pocachard E, Massey-Harroche D,
Richard F, Arsanto J-P, Chauvin J-P, et al. Hook2 is involved in
the morphogenesis of the primary cilium. Mol Biol Cell. 2011;22:
454962.
75. Haeseleer F, Jang G-F, Imanishi Y, Driessen CA, Matsumura M,
Nelson PS, et al. Dual-substrate specificity short chain retinol
dehydrogenases from the vertebrate retina. J Biol Chem.
2002;277:4553746.
76. Wu P, Farrell WE, Haworth KE, EmesRD, Kitchen MO, Glossop
JR, et al. Maternal genome-wide DNA methylation profiling in
gestational diabetes shows distinctive disease-associated changes
relative to matched healthy pregnancies. Epigenetics. 2018;13:
1228.
77. De Benoist B, Cogswell M, Egli I, McLean E (2008) Worldwide
prevalence of anaemia 1993-2005; who global database of
anaemia.
78. Greenberg JA, Bell SJ, Guan Y, Yu Y-h. Folic acid supplementa-
tion and pregnancy: more than just neural tube defect prevention.
Rev Obstet Gynecol. 2011;4:52.
79. Pritchard JA, Adams RH. Erythrocyte production and destruction
during pregnancy. American Journal of Obstetrics & Gynecology.
1960;79:7507.
80. Sharma JB, Shankar M. Anemia in pregnancy. JIMSA. 2010;23:
25360.
81. Tamura T, Picciano MF. Folate and human reproduction. Am J
Clin Nutr. 2006;83:9931016.
82. Joubert BR, Herman T, Felix JF, et al. Maternal plasma folate
impacts differential DNA methylation in an epigenome-wide me-
ta-analysis of newborns. Nat Commun. 2016;7:10577.
83. Knight AK, Park HJ, Hausman DB, Fleming JM, Bland VL, Rosa
G, et al. Association between one-carbon metabolism indices and
DNA methylation status in maternal and cord blood. Sci Rep.
2018;8:16873.
84. Blencowe H, Cousens S, Chou D, Oestergaard M, Say L, Moller
A-B, et al. Born too soon: the global epidemiology of 15 million
preterm births. Reprod Health. 2013;10:S2.
85. Liu L, Oza S, Hogan D, Chu Y, Perin J, Zhu J, et al. Global,
regional, and national causes of under-5 mortality in 200015:
an updated systematic analysis with implications for the sustain-
able development goals. Lancet. 2016;388:302735.
86. Menon R. Spontaneous preterm birth, a clinical dilemma: etiolog-
ic, pathophysiologic and genetic heterogeneities and racial dispar-
ity. Acta Obstet Gynecol Scand. 2008;87:590600.
87. Frey HA, Stout MJ, Pearson LN, Tuuli MG, Cahill AG, Strauss JF
III, et al. Genetic variation associated with preterm birth in
African-American women. Am J Obstet Gynecol. 2016;215:
235e1.
88. Menon R, Fortunato SJ, Velez Edwards DR, Williams SM.
Association of genetic variants, ethnicity and preterm birth with
amniotic fluid cytokine concentrations. Ann Hum Genet.
2010;74:16583.
89. Zhang G, Feenstra B, Bacelis J, Liu X, Muglia LM, Juodakis J,
et al. Genetic associations with gestational duration and spontane-
ous preterm birth. N Engl J Med. 2017;377:115667.
90. Hong X, Sherwood B, Ladd-Acosta C, Peng S, Ji H, Hao K, et al.
Genome-wide DNA methylation associations with spontaneous
Reprod. Sci.
preterm birth in US blacks: findings in maternal and cord blood
samples. Epigenetics. 2018;13:16372.
91. Chen Q, Coffey A, Bourgoin SG, Gadina M. Cytohesin binder
and regulator augments t cell receptor-induced nuclear factor of
activated t cells ap-1 activation through regulation of the jnk path-
way. J Biol Chem. 2006;281:1998594.
92. OBrien M, Morrison JJ, Smith TJ. Upregulation of pscdbp, tlr2,
twist1, flj35382, ednrb, and rgs12 gene expression in human
myometrium at labor. Reprod Sci. 2008;15:38293.
93. Kropf P, Baud D, Marshall SE, et al. Arginase activity mediates
reversible T cell hyporesponsiveness in human pregnancy. Eur J
Immunol. 2007;37:93545.
94. Jeanty C, Derderian SC, Mackenzie TC. Maternal-fetal cellular
trafficking: clinical implications and consequences. Curr Opin
Pediatr. 2014;26:37782.
95. Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: func-
tional surprises from the RNA world. Genes Dev. 2009;23:1494
504.
96. Consortium F, II Team RGERGPI &, others (2002) Analysis of
the mouse transcriptome based on functional annotation of 60,770
full-length cDNAs. Nature 420:563, 573.
97. Chen SJ, Liu YL, Sytwu HK. Immunologic regulation in pregnan-
cy: from mechanism to therapeutic strategy for
immunomodulation. Clin Dev Immunol. 2012;2012:258391.
98. Parets SE, Conneely KN, Kilaru V, Menon R, Smith AK. DNA
methylation provides insight into intergenerational risk for pre-
term birth in African Americans. Epigenetics. 2015;10:78492.
99. Foster HA, Davies J, Pink RC, Turkcigdem S, Goumenou A,
Carter DR, et al. The human myometrium differentially expresses
mTOR signalling components before and during pregnancy: evi-
dence for regulation by progesterone. J Steroid Biochem Mol
Biol. 2014;139:16672.
100. Laplante M, Sabatini DM. MTOR signaling at a glance. J Cell Sci.
2009;122:358994.
101. Jaffer S, Shynlova O, Lye S. Mammalian target of rapamycin is
activated in association with myometrial proliferation during preg-
nancy. Endocrinology. 2009;150:467280.
102. McRae AF, Powell JE, Henders AK, Bowdler L, Hemani G, Shah
S, et al. Contribution of genetic variation to transgenerational in-
heritance of DNA methylation. Genome Biol. 2014;15:R73.
103. Fujimoto T, Parry S, Urbanek M, Sammel M, Macones G,
Kuivaniemi H, et al. A single nucleotide polymorphism in the
matrix metalloproteinase-1 (MMP-1) promoter influences amnion
cell MMP-1 expression and risk for preterm premature rupture of
the fetal membranes. J Biol Chem. 2002;277:6296302.
104. Wang H, Ogawa M, Wood JR, Bartolomei MS, Sammel MD,
Kusanovic JP, et al. Genetic and epigenetic mechanisms combine
to control mmp1 expression and its association with preterm pre-
mature rupture of membranes. Hum Mol Genet. 2008;17:1087
96.
105. Moutquin J-M. Classification and heterogeneity of preterm birth.
BJOG Int J Obstet Gynaecol. 2003;110:303.
106. Knijnenburg TA, Vockley JG, Chambwe N, Gibbs DL,
Humphries C, Huddleston KC, et al. Genomic and molecular
characterization of preterm birth. Proc Natl Acad Sci. 2019;116:
581927.
107. Winn VD, Haimov-Kochman R, Paquet AC, Yang YJ,
Madhusudhan MS, Gormley M, et al. Gene expression profiling
of the human maternal-fetal interface reveals dramatic changes
between midgestation and term. Endocrinology. 2007;148:
105979.
108. Denis M, Enquobahrie DA, Tadesse MG, Gelaye B, Sanchez SE,
Salazar M, et al. Placental genome and maternal-placental genetic
interactions: a genome-wide and candidate gene association study
of placental abruption. PLoS One. 2014;9:e116346.
109. Han H, Tanigaki K, Yamamoto N, Kuroda K, Yoshimoto M,
Nakahata T, et al. Inducible gene knockout of transcription factor
recombination signal binding protein-j reveals its essential role in t
versus b lineage decision. Int Immunol. 2002;14:63745.
110. Cunningham FG, Leveno K, Bloom S, Hauth J, Gilstrap L,
Wenstrom K (2005) Gestational trophoblastic disease. Williams
Obstetrics. 22nd ed. New York: McGraw Hill.
111. Small MJ, Kershaw T, Frederic R, Blanc C, Neale D, Copel J,
et al. Characteristics of preeclampsia-and eclampsia-related mater-
nal death in rural Haiti. J Matern Fetal Neonatal Med. 2005;18:
3438.
112. Manandhar T, Prashad B, Nath Pal M. Risk factors for intrauterine
growth restriction and its neonatal outcome. Gynecol Obstet.
2018;8:21610932.
113. Walsh SW. Obesity: a risk factor for preeclampsia. Trends in
Endocrinology & Metabolism. 2007;18:36570.
114. Burton GJ, Jauniaux E. Placental oxidative stress: from miscar-
riage to preeclampsia. J Soc Gynecol Investig. 2004;11:34252.
115. Hing B, Braun P, Cordner ZA, Ewald ER, Moody L, McKane M,
et al. Chronic social stress induces DNA methylation changesat an
evolutionary conserved intergenic region in chromosome X.
Epigenetics. 2018;13:62741.
116. He F, Berg A, Kawasawa YI, Bixler EO, Fernandez-Mendoza J,
Whitsel EA, et al. Association between DNA methylation in
obesity-related genes and body mass index percentile in adoles-
cents. Sci Rep. 2019;9:2079.
117. Wilson SL, Leavey K, Cox B, Robinson WP. The value of DNA
methylation profiling in characterizing preeclampsia and intrauter-
ine growth restriction. BioRxiv. 2017;151290.
118. Leavey K, Wilson SL, Bainbridge SA, Robinson WP, Cox BJ.
Epigenetic regulation of placental gene expression in transcrip-
tional subtypes of preeclampsia. Clin Epigenetics. 2018;10:28.
119. Wilson SL, Leavey K, Cox BJ, Robinson WP. Mining DNA
methylation alterations towards a classification of placental pa-
thologies. Hum Mol Genet. 2018;27:13546.
120. Tsui DW, Chan KA, Chim SS, L-w C, T-y L, T-k L, et al.
Quantitative aberrations of hypermethylated rassf1a gene se-
quences in maternal plasma in pre-eclampsia. Prenatal
Diagnosis: Published in Affiliation With the International
Society for Prenatal Diagnosis. 2007;27:12128.
121. Bianchi DW (2004) Circulating fetal DNA: its origin and diagnos-
tic potential-a review. Placenta 25 Suppl A:S93S101.
122. Emlen W, Mannik M. Effect of DNA size and strandedness on the
in vivo clearance and organ localization of DNA. Clin Exp
Immunol. 1984;56:18592.
123. TSUMITA T, IWANAGA M. Fate of injected deoxyribonucleic
acid in mice. Nature. 1963;198:10889.
124. Hesson LB, Cooper WN, Latif F. The role of RASSF1A methyl-
ation in cancer. Dis Markers. 2007;23:7387.
125. Chiu RW, Chim SS, Wong IH, et al. Hypermethylation of
RASSF1A in human and rhesus placentas. Am J Pathol.
2007;170:94150.
126. Lee SB, Wong AP, Kanasaki K, Xu Y, Shenoy VK, McElrath TF,
et al. Preeclampsia: 2-methoxyestradiol induces cytotrophoblast
invasion and vascular development specifically under hypoxic
conditions. Am J Pathol. 2010;176:71020.
127. Mousa AA, Archer KJ, Cappello R, Estrada-Gutierrez G, Isaacs
CR, Strauss JF III, et al. DNA methylation is altered in maternal
blood vessels of women with preeclampsia. Reprod Sci. 2012;19:
133242.
128. Tanabe T, Ullrich V. Prostacyclin and thromboxane synthases. J
Lipid Mediat Cell Signal. 1995;12:24355.
129. Walsh SW. Preeclampsia: an imbalance in placental prostacyclin
and thromboxane production. Am J Obstet Gynecol. 1985;152:
33540.
Reprod. Sci.
130. Chavarrı a ME, Lara-González L, González-Gleason A, Garcı a-
Paleta Y, Vital-Reyes VS, Reyes A (2003) Prostacyclin/
thromboxane early changes in pregnancies that are complicated
by preeclampsia. Am J Obstet Gynecol 188:986992.
131. Anderson CM, Ralph JL, Wright ML, Linggi B, Ohm JE. DNA
methylation as a biomarker for preeclampsia. Biological research
for nursing. 2014;16:40920.
132. Schulz LC, Widmaier EP, Qiu J, Roberts RM. Effect of leptin on
mouse trophoblast giant cells. Biol Reprod. 2009;80:41524.
133. Liu AX, Jin F, Zhang WW, Zhou TH, Zhou CY, Yao WM, et al.
Proteomic analysis on the alteration of protein expression in the
placental villous tissue of early pregnancy loss. Biol Reprod.
2006;75:41420.
134. Abumaree MH, Chamley LW, Badri M, El-Muzaini MF.
Trophoblast debris modulates the expression of immune proteins
in macrophages: a key to maternal tolerance of the fetal allograft?
J Reprod Immunol. 2012;94:13141.
135. Liu B, Xu Y, Voss C, Qiu FH, Zhao MZ, Liu YD, et al. Altered
protein expression in gestational diabetes mellitus placentas pro-
vides insight into insulin resistance and coagulation/fibrinolysis
pathways. PLoS One. 2012;7:e44701.
136. Sharma D, Shastri S, Sharma P (2016) Intrauterine growth restric-
tion: antenatal and postnatal aspects. Clinical Medicine Insights:
Pediatrics 10:CMPedS40070.
137. Martin EM, Fry RC. Environmental influences on the epigenome:
exposure-associated DNA methylation in human populations.
Annu Rev Public Health. 2018;39:30933.
138. Koh W, Pan W, Gawad C, Fan HC, Kerchner GA, Wyss-Coray T,
et al. Noninvasive in vivo monitoring of tissue-specific global
gene expression in humans. Proc Natl Acad Sci U S A.
2014;111:73616.
139. Houseman EA, Accomando WP, Koestler DC, Christensen BC,
Marsit CJ, Nelson HH, et al. DNA methylation arrays as surrogate
measures of cell mixture distribution. BMC bioinformatics.
2012;13:86.
140. Rahmani E, Schweiger R, Shenhav L, Wingert T, Hofer I, Gabel
E, et al. BayesCCE: a Bayesian framework for estimating cell-type
composition from dna methylation without the need for methyla-
tion reference. Genome Biol. 2018;19:118.
141. Li Z, Wu H. TOAST: improving reference-free cell composition
estimation by cross-cell type differential analysis. Genome Biol.
2019;20:190.
142. Michalczyk AA, Janus ED, Judge A, Ebeling PR, Best JD,
Ackland MJ, et al. Transientepigenomic changes during pregnan-
cy and early postpartum in women with and without type 2 dia-
betes. Epigenomics. 2018;10:419.
143. Moen G-H, Sommer C, Prasad RB, Sletner L, Groop L, Qvigstad
E, et al. MECHANISMS IN ENDOCRINOLOGY: epigenetic
modifications and gestational diabetes: a systematic review of
published literature. Eur J Endocrinol. 2017;176:R24767.
144. Jia N, Li J. Noncoding RNAs in unexplained recurrent spontane-
ous abortions and their diagnostic potential. Dis Markers. 2019.
https://doi.org/10.1155/2019/7090767.
145. Wang G, Divall S, Radovick S, Paige D, Ning Y, Chen Z, et al.
Preterm birth and random plasma insulin levels at birth and in
early childhood. Jama. 2014;311:58796.
146. Parets SE, Knight AK, Smith AK. Insights into genetic suscepti-
bility in the etiology of spontaneous preterm birth. Appl Clin
Genet. 2015;8:283.
147. Bhatnagar S, Majumder PP, Salunke DM. A pregnancy cohort to
study multidimensional correlates of preterm birth in India: study
design, implementation, and baseline characteristics of the partic-
ipants. Am J Epidemiol. 2019;188:62131.
PublishersNoteSpringer Nature remains neutral with regard to jurisdic-
tional claims in published maps and institutional affiliations.
Reprod. Sci.
... In contrast to cfDNA yield/fractions, the assessment of cfDNA methylation patterns potentially involves evaluation of both the cause and/or effect of HDP 22,28 . Recent systematic reviews summarized the role of DNA methylation in physiological and pathophysiological pregnancies including PE 29,30 . The majority of methylation studies conducted so far have focused on the placental compartment and mostly on selected candidate genes, emphasizing on outcomes and co-morbidities of pregnancy 30 . ...
... Recent systematic reviews summarized the role of DNA methylation in physiological and pathophysiological pregnancies including PE 29,30 . The majority of methylation studies conducted so far have focused on the placental compartment and mostly on selected candidate genes, emphasizing on outcomes and co-morbidities of pregnancy 30 . Reports on cfDNA genome-wide differential methylation regions (DMRs) or patterns, especially in early pregnancy, are absent 29 . ...
Article
Full-text available
Hypertensive disorders of pregnancy (HDP) contribute substantially to perinatal morbidity and mortality. Epigenetic changes point towards cardio-metabolic dysregulation for these vascular disorders. In early pregnancy, epigenetic changes using cell free DNA (cfDNA) are largely unexplored. We aimed to investigate these in HDP between 11 and 14 weeks of gestation by analysis of cfDNA methylation profiles in patients with hypertensive disorders. We identified patients without chronic hypertension but with subsequent development of preeclampsia (PE) (n = 11), with chronic hypertension (HT) but without PE development (n = 14), and lacking both PE and HT (n = 422). We matched patients according to PE risk factors into three groups (n = 5 each group): (1) PE: no HT but PE development, (2) HT: chronic hypertension but no PE and (3) Control: no PE or HT. We successfully optimized our cfDNA isolation process prior to whole genome bisulfite sequencing. Analysis of cfDNA methylation changes indicate a common predisposition in PE and HT groups, chiefly of maternal origin. Assessment of significant differentially methylated regions and annotated genes point towards a common cardiovascular predisposition in preeclampsia and hypertension groups in the first trimester. We postulate the pivotal role of the maternal cardiovascular system in HDP, which is already evident in the first trimester.
... Epigenetic mechanisms can increase the risk of becoming obese and diabetic, and can be influenced by the nutritional status and physical activity patterns of the parent. Many environmental abuses such as restriction or unhealthy nutrition, lack of exercise, tobacco smoking, alcohol consumption, exposure to environmental pollutants, psychological stress, ethnicity, and hypertension have been recognized to increase an individual's risk of metabolic disorder during their lifetime in around 50% of cases [95][96][97][98]. The prevalence of GDM has risen dramatically by over 30% within the last two decades in several countries, including developing countries [99]. ...
... Studies established on whole blood DNAm are generally limited. Whole blood is composed of multiple cell types, and each cell type contributes to the CpG locus-specific DNA methylation signal [96]. Studies on DNAm in whole blood have not topically addressed the issue of cellular heterogeneity. ...
Article
Full-text available
Epigenetics modification such as DNA methylation can affect maternal health during the gestation period. Furthermore, pregnancy can drive a range of physiological and molecular changes that have the potential to contribute to pathological conditions. Pregnancy-related risk factors include multiple environmental, behavioral, and hereditary factors that can impact maternal DNA methylation with long-lasting consequences. Identification of the epigenetic patterns linked to poor pregnancy outcomes is crucial since changes in DNA methylation patterns can have long-term effects. In this review, we provide an overview of the epigenetic changes that influence pregnancy-related molecular programming such as gestational diabetes, immune response, and pre-eclampsia, in an effort to close the gap in current understanding regarding interactions between the environment, the genetics of the fetus, and the pregnant woman.
... Pregnancy has also been linked to changes in DNA methylation (DNAm) -the addition or removal of methyl groups to cytosine-phosphate-guanine (CpG) sites on DNA, frequently in promoter regions -which regulate gene expression. DNAm changes throughout pregnancy are considered as an important part of gene regulation and normal cellular control mechanisms [14,16]. Fradin et al. investigated DNAm changes in maternal blood of a longitudinal cohort of pregnant women from early pregnancy (up to 18 weeks) to late pregnancy (35 weeks and later) and found a gain of methylation in genes involved in morphogenesis, such as ezrin, and a loss of methylation in genes promoting maternal-infant bonding [22]. ...
Article
Full-text available
Background Pregnancy and childbirth have been connected to modified risk of a wide variety of conditions in later life, including neurodegenerative disorders and cancers. The presence, extent, and direction of the effect that childbearing status has on decreasing or increasing the risk of these conditions differs depending on the disease. The mechanisms by which pregnancy and childbirth modify the risk of diseases are still unknown. DNA methylation (DNAm) alterations that occur during pregnancy and persist after childbirth may help us understand this phenomenon. Results Blood DNAm was available from 89 women (28 parous; 61 nulliparous) at ages 18 and 26 years in the Isle of Wight birth cohort; no significant differences in the population characteristics were present between the analyzed population and the full cohort. We performed an epigenome-wide association study on 389,355 CpGs and identified 184 CpGs to be significantly differentially methylated between parous and nulliparous women after adjusting for confounders and multiple testing. Of these CpGs, 105 had regression coefficients in the same direction in an independent Mexico City based ELEMENT cohort, of which 13 were significant (replication P < 0.05). These 13 CpGs were associated with 16 unique genes. DNAm levels tracked with gene expression in 3 of the replicated genes, one of which (TM2D3) was differentially expressed in parous vs nulliparous women. Gene disease association analysis identified a network of parous-associated diseases. Conclusions Our results suggest that pregnancy and childbirth lead to DNAm changes in parous women and these changes persist at least 6 months and up to 8 years postpartum. Parous-related CpG sites may play a role in how childbearing status modifies risk of later life diseases in women. Further studies are needed to explore the linkage and mechanism.
... The majority of these APBOs are associated with maternal exposure to genetic and environmental factors during pregnancy. Pregnancy is a vital period of plasticity wherein maternal exposure to multiple environmental, behavioral, and hereditary factors may significantly affect fetal development as well as the mother's health [5,6]. ...
Article
Full-text available
Background Deoxyribonucleic acid (DNA) methylation is one of the epigenetic modifications that has gained a lot of interest as a factor influencing fetal programming and as a biomarker for adverse pregnancy and birth outcomes (APBOs). Epidemiological studies have demonstrated that DNA methylation can result in adverse pregnancy and birth outcomes (APBOs) including miscarriage, intrauterine growth restriction (IUGR), low birth weight (LBW), sepsis, and preterm birth (PTB), which may later result in diseases in adulthood. However, the mechanism by which DNA methylation influences these APBOs remains unclear. The systematic review will assess the association between global and gene-specific DNA methylation with adverse pregnancy outcomes. Method The Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) 2020 checklist will be followed when conducting this systematic review. To develop the search strategy the PI(E)COS (population, intervention/exposure, comparator/control, outcome, and study designs) framework will be followed. Thus far, the research team has retrieved 4721 from Cochrane Library, PubMed, Web of Sciences, and MEDLINE. Out of these, 584 studies have been screened for eligibility, and approximately 124 studies meet the inclusion criteria. Pending the search results identified from the grey literature. For identification of unpublished studies in journals indexed in electronic databases, Google Scholar will be used. I.M and A.S will separately extract data from the articles and screen them, if there are any disagreements between I.M and A.S, then the L.M will resolve them. The methodological quality and bias risk of the included studies will be evaluated using the Critical Appraisal Skill Programme CASP) checklist. I2\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$${I}^{2}$$\end{document} and χ2\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\chi 2{}$$\end{document} alpha = 0.10 statistic will be used for assessing statistical heterogeneity between studies. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) approach will be used to assess and grade the overall quality of extracted data. Ethics and dissemination Ethical approval is not required. The systematic review will assess available literature on possible associations between DNA methylation with adverse pregnancy and birth outcomes (APBOs) including LBW, IUGR, miscarriage, sepsis, and PTB. The findings could help guide future research assessing DNA methylation and other APBOs. Systematic review registration PROSPERO CRCRD42022370647.
... DNA methylation changes even at single CpG loci can lead to altered gene expression and manifest a particular phenotype [6][7][8][9][10]. Emerging evidence suggests the ability of DNA methylation changes during pregnancy to impact the health of the pregnancy as well as birth outcomes [11]. However, most studies have compared adverse and normal pregnancies, and very little effort has been made to understand the epigenetic alterations involved in term pregnancy itself [12][13][14]. ...
Article
Full-text available
Background: We performed an epigenome-wide longitudinal DNA methylation study on an Indian cohort of pregnant women, GARBH-Ini, at three time points during pregnancy and at delivery. Aim & objective: Our aim was to identify temporal DNA methylation changes in maternal peripheral blood during the period of gestation and assess their impact on biological pathways critical for term delivery. Results: Significantly differentially methylated CpGs were identified by linear mixed model analysis (Bonferroni p < 0.01) and classified into two distinct temporal methylation trends: increasing and decreasing during gestation. Genes with upward methylation trend were enriched for T-cell activity, while those with a downward trend were enriched for solute transport and cell structure organization functions. Conclusion: Consistent trends of DNA methylation in maternal peripheral blood point to the sentinel function of T cells in the maintenance of pregnancy, and the importance of coordinated cellular remodeling to facilitate term delivery.
Article
Full-text available
For much of human evolution, the average lifespan was <40 years, due in part to disease, infant mortality, predators, food insecurity, and, for females, complications of childbirth. Thus, for much of evolution, many females did not reach the age of menopause (45–50 years of age) and it is mainly in the past several hundred years that the lifespan has been extended to >75 years, primarily due to public health advances, medical interventions, antibiotics, and nutrition. Therefore, the underlying biological mechanisms responsible for disease risk following menopause must have evolved during the complex processes leading to Homo sapiens to serve functions in the pre-menopausal state. Furthermore, as a primary function for the survival of the species is effective reproduction, it is likely that most of the advantages of having such post-menopausal risks relate to reproduction and the ability to address environmental stresses. This opinion/perspective will be discussed in the context of how such post-menopausal risks could enhance reproduction, with improved survival of offspring, and perhaps why such risks are preserved. Not all post-menopausal females exhibit risk for this set of diseases, and those who do develop such diseases do not have all of the conditions. The diseases of the post-menopausal state do not operate as a unified complex, but as independent variables, with the potential for some overlap. The how and why there would be such heterogeneity if the risk factors serve essential functions during the reproductive years is also discussed and the concept of sets of reversible epigenetic changes associated with puberty, pregnancy, and lactation is offered to explain the observations regarding the distribution of post-menopausal conditions and their potential roles in reproduction. While the involvement of an epigenetic system with a dynamic “modification-demodification-remodification” paradigm contributing to disease risk is a hypothesis at this point, validation of it could lead to a better understanding of post-menopausal disease risk in the context of reproduction with commonalities may also lead to future improved interventions to control such risk after menopause.
Preprint
Full-text available
Background Deoxyribonucleic acid (DNA) methylation is one the epigenetic modifications which has gain a lot of interest as a factor influencing fetal programming and as a biomarker for adverse pregnancy and birth outcomes (APBOs). Epidemiological studies have demonstrated that DNA methylation can result in adverse pregnancy and birth outcomes (APBOs) including miscarriage, intrauterine growth restriction (IUGR), low birth weight (LBW), sepsis as well as preterm birth (PB), which may later result in diseases in adulthood. However, the mechanism at which DNA methylation influences these APBOs remain unclear. The systematic review will assess the association between global and gene specific DNA methylation with adverse pregnancy outcomes. Method The Preferred Reporting Items for Systematic review and Meta-Analysis Protocol (PRISMA-P) 2015 checklist will be followed when conducting this systematic review. To develop the search strategy the PECOS (population, exposure, comparator/control, outcome, and study designs) framework will be followed. A systematic search will be conducted on electronic databases such as Cochrane library, PubMed, as well as MEDLINE. For identification of unpublished studies in journals indexed in electronic databases, Google Scholar will be used. Two reviewers will separately extract data from the articles and screen them, if there are any disagreements between reviewer one and two, then the third reviewer will resolve them. The methodological quality and bias risk of the included studies will be evaluated using Critical Appraisal Skill Programme CASP) checklist. \({I}^{2}\) and \({X}^{2}\) alpha = 0.10 statistic will be used for assessing statistical heterogeneity between studies. The Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach will be used to assess and grade the overall quality of extracted data. Discussion The systematic review will assess available literature on possible associations between DNA methylation with adverse pregnancy and birth outcomes (APBOs) including LBW, IUGR, miscarriage, sepsis and PB. The findings could help guide future research assessing DNA methylation and other APBOs. PROSPERO registration number: CRD42022370647
Article
Maternal sleep and circadian health during pregnancy are emerging as important predictors of pregnancy outcomes, but examination of potential epigenetic mechanisms is rare. We investigated links between maternal leukocyte DNA methylation of circadian genes and birth outcomes within a pregnancy cohort. Women (n = 96) completed a questionnaire and provided a blood sample at least once during early-to-mid pregnancy (average gestation weeks = 14.2). Leukocyte DNA was isolated and DNA methylation (average percent of methylation) at multiple CpG sites within BMAL1, PER1, and MTNR1B genes were quantified by pyrosequencing. Birth outcomes including gestational age at delivery, birthweight, and head circumference were abstracted from medical charts. Linear regression analyses were run between each CpG site with birth outcomes, adjusting for important confounders. Sleep duration and timing were assessed as secondary exposures. Higher methylation of a CpG site in PER1 was associated with smaller log-transformed head circumference (β=-0.02 with 95% CI -0.02 to 0.01; P, trend = 0.04). Higher methylation of MTNR1B (averaged across sites) was associated with lower log-transformed birthweight (-0.08 with 95% CI -0.16 to -0.01; P, trend = 0.0495). In addition, longer sleep duration was associated with higher birthweight (0.10 with 95% CI 0.02 to 0.18 comparing > 9 h to < 8 h; P, trend = 0.04). This pilot investigation revealed that higher methylation of PER1 and MTNR1B genes, and sleep duration measured in early-to-mid pregnancy were related to birth outcomes.
Article
Full-text available
Maternal smoking in pregnancy (MSP) affects the offspring's DNA methylation (DNAm). There is a lack of knowledge regarding individual differences in susceptibility to exposure to MSP. Glutathione S-transferase (GST) genes are involved in protection against harmful oxidants such as those found in cigarette smoke. This study aimed to test whether polymorphisms in GST genes influence the effect of MSP on offspring DNAm. Using data from the Isle of Wight birth cohort, we assessed the association of MSP and offspring DNAm in 493 mother-child dyads (251 male, 242 female) with the effect-modifying role of GST gene polymorphism (at rs506008, rs574344, rs12736389, rs3768490, rs1537234, and rs1695). MSP was assessed by levels of nicotine and its downstream metabolites (coti-nine, norcotinine, and hydroxycotinine) in maternal sera. In males, associations of hydroxycotinine with DNAm at cg18473733, cg25949550, cg11647108, and cg01952185 and norcotinine with DNAm at cg09935388 were modified by GST gene polymorphisms (p-values < 0.05). In females, associations of hydroxycotinine with DNAm at cg12160087 and norcotinine with DNAm at cg18473733 were modified by GST gene polymorphisms (p-values < 0.05). Our study emphasizes the role of genetic polymorphism in GST genes in DNAm's susceptibility to MSP.
Article
Introduction: Gestational diabetes mellitus (GDM) is the first occurrence of diabetes due to abnormal maternal sugar metabolism after pregnancy, which may lead to adverse pregnancy outcomes. Hesperidin is known to decrease in the cord blood of GDM with obesity, but its role is unknown. This study aims to explore the potential function of hesperidin in GDM with obesity to develop new therapeutic ideas. Methods: Peripheral blood and placental tissues from GDM and GDM with obesity patients were collected to isolate human villous trophoblasts and detection. Bioinformatics was used to analyze the differential methylation genes between GDM and GDM with obesity. Immunofluorescence was applied for the detection of CK7 expression. Cells vitality was detected by CCK8 and transwell. Molecular docking was applied to predict the binding of hesperidin and ATG7 protein. Inflammation and m6A levels was analyzed by ELISA. ATG7, LC3, TLR4 and P62 proteins was analyzed by Western blot. Results: The methylation of ATG7 gene was up-regulated in GDM with obesity compared with GDM. The m6A and autophagy proteins levels in GDM with obesity were higher than that in GDM. LPS with 2.5-25 mM glucose induced the increase of autophagy proteins, inflammation and m6A levels in human villous trophoblasts. Hesperidin formed hydrogen bonds and hydrophobic interactions with ATG7 proteins. Hesperidin (0.25 μM) inhibited the autophagy proteins and m6A level in LPS and 25 mM glucose-induced human villous trophoblasts. Discussion: GDM with obesity followed the increase of autophagy proteins and m6A levels. Hesperidin inhibited the autophagy proteins and m6A level in LPS and glucose-induced human villous trophoblasts.
Preprint
Full-text available
Placental health is a key component to healthy pregnancy. Placental insufficiency (PI), inadequate nutrient delivery to the fetus, is associated with preeclampsia (PE), a maternal hypertensive disorder, and intrauterine growth restriction (IUGR), pathologically poor fetal growth. PI is more common in early-onset PE (EOPE) than late-onset PE (LOPE). However, the relationship between these disorders remains unclear. While DNA methylation (DNAm) alterations have been identified in PE and IUGR, these entities can overlap and few studies have analyzed these separately. This study aims to identify altered DNAm in EOPE, LOPE, and normotensive IUGR, validate these alterations, and use them to better understand the relationships between these related disorders. Placental samples from a discovery cohort (43 controls, 22 EOPE, 18 LOPE, 11 IUGR) and validation cohort (15 controls, 22 EOPE, 11 LOPE) were evaluated using the Illumina HumanMethylation450 array. To minimize gestational age (GA) effects, EOPE samples were compared to pre-term controls (GA <37 weeks), while LOPE and IUGR were compared to term controls (GA >37 weeks). There were 1703 differentially methylated (DM) sites (FDR<0.05, Δβ>0.1) in EOPE, 5 in LOPE, and 0 in IUGR. Of the 1703 EOPE sites, 599 were validated in the second cohort. These sites cluster samples from both cohorts into 3 distinct methylation clusters. Interestingly, LOPE samples diagnosed between 34-36 weeks with co-occurring IUGR clustered with the EOPE methylation cluster. DNAm profiling may provide an independent tool to refine clinical diagnoses into subgroups with more uniform pathology. The challenges in reproducing genome-wide DNAm studies are also discussed.
Article
Full-text available
Unexplained recurrent spontaneous abortion (URSA) is defined as the loss of two or more consecutive pregnancies before the 20th week of gestation with normal findings on routine screening tests. Our understanding of the cellular and molecular pathogenesis of URSA is still far from complete. Noncoding RNAs (ncRNAs) play a pivotal role in transcription and expression. The functions of ncRNAs may also improve understanding of URSA pathogenesis. Because of their stability in the circulatory system and at the maternal-fetal interface, it may be possible to use ncRNAs as biomarkers for certain disease states. Here, we provide a narrative review of the current state of knowledge about ncRNAs associated with URSA. The possibility of developing a diagnostic tool using ncRNAs is discussed. The underlying mechanisms of how ncRNAs may lead to the onset of URSA are explored in this review.
Article
Full-text available
In the analysis of high-throughput data from complex samples, cell composition is an important factor that needs to be accounted for. Except for a limited number of tissues with known pure cell type profiles, a majority of genomics and epigenetics data relies on the “reference-free deconvolution” methods to estimate cell composition. We develop a novel computational method to improve reference-free deconvolution, which iteratively searches for cell type-specific features and performs composition estimation. Simulation studies and applications to six real datasets including both DNA methylation and gene expression data demonstrate favorable performance of the proposed method. TOAST is available at https://bioconductor.org/packages/TOAST.
Article
Full-text available
There is emerging evidence on DNA methylation (DNAm) variability over time; however, little is known about dynamics of DNAm patterns during pregnancy. We performed an epigenome-wide longitudinal DNAm study of a well-characterized sample of young women from the Swedish Born into Life study, with repeated blood sampling before, during and after pregnancy (n = 21), using the Illumina Infinium MethylationEPIC array. We conducted a replication in the Isle of Wight third-generation birth cohort (n = 27), using the Infinium HumanMethylation450k BeadChip. We identified 196 CpG sites displaying intra-individual longitudinal change in DNAm with a false discovery rate (FDR) P < .05. Most of these (91%) showed a decrease in average methylation levels over the studied period. We observed several genes represented by ⩾3 differentially methylated CpGs: HOXB3, AVP, LOC100996291, and MicroRNA 10a. Of 36 CpGs available in the replication cohort, 17 were replicated, all but 2 with the same direction of association (replication P < .05). Biological pathway analysis demonstrated that FDR-significant CpGs belong to genes overrepresented in metabolism-related pathways, such as adipose tissue development, regulation of insulin receptor signaling, and mammary gland fat development. These results contribute to a better understanding of the biological mechanisms underlying important physiological alterations and adaptations for pregnancy and lactation.
Article
Full-text available
Abstract While biological alterations associated with childhood maltreatment (CM) have been found in affected individuals, it remains unknown to what degree these alterations are biologically transmitted to the next generation. We investigated intergenerational effects of maternal CM on DNA methylation and gene expression in N = 113 mother-infant dyads shortly after parturition, additionally accounting for the role of the FKBP5 rs1360780 genotype. Using mass array spectrometry, we assessed the DNA methylation of selected stress-response-associated genes (FK506 binding protein 51 [FKBP5], glucocorticoid receptor [NR3C1], corticotropin-releasing hormone receptor 1 [CRHR1]) in isolated immune cells from maternal blood and neonatal umbilical cord blood. In mothers, CM was associated with decreased levels of DNA methylation of FKBP5 and CRHR1 and increased NR3C1 methylation, but not with changes in gene expression profiles. Rs1360780 moderated the FKBP5 epigenetic CM-associated regulation profiles in a gene × environment interaction. In newborns, we found no evidence for any intergenerational transmission of CM-related methylation profiles for any of the investigated epigenetic sites. These findings support the hypothesis of a long-lasting impact of CM on the biological epigenetic regulation of stress-response mediators and suggest for the first time that these specific epigenetic patterns might not be directly transmitted to the next generation.
Article
Full-text available
Preterm birth is the leading cause of mortality in newborn infants and can lead to significant neonatal morbidities. Spontaneous preterm birth accounts for at least 50.0% of all preterm births. We argue that chronic maternal stress load, which is an important risk factor for spontaneous preterm birth, could be represented by epigenetic signature of several specific genetic loci in the mother’s blood. A literature search was done in PubMed with the following keywords: “DNA methylation,” “epigenetics,” “maternal stress” and “preterm birth” from year 2000 to 2017. We suggest that these genetic loci might be related to vulnerability and hypersensibility of stress response during pregnancy in women with preterm births. The mother’s epi-genetic stress bioprofile was supposed to be a result of chronic maternal stress load since her birth. This epigenetic bioprofile might also be a potential biomarker for spontaneous preterm birth. DNA methylation changes are tissue-specific and human stress response manifests mostly through the central nervous system (CNS). Nevertheless, we found evidence that methylation changes of DNA isolated from blood leucocytes might be a reliable measure of stress-related epigenetic changes that occur in the CNS. Evaluating biological mechanisms through the development of simple assays based on epigenetic changes to measure chronic stress loads in expectant mothers can lead to our ability to prepare more effective measures for the prevention of preterm births, as well as leading to more effective treatment strategies for both expectant mothers and their newborns.
Article
Full-text available
Significance Preterm birth (PTB) complications are the leading cause of long-term morbidity and mortality in children. The genetic and molecular characteristics of PTB and related disorders remain unclear. In this study, a family-based cohort of 791 family trios, including 270 PTB and 521 control families, was investigated by using whole-genome sequencing, RNA sequencing, and DNA methylation data. Integrative analysis identified 160 genomic variants associated with PTB-related phenotypes and led to the discovery of 72 candidate biomarker genes for very early PTB (VEPTB). The genes associated with VEPTB involve growth signaling and inflammation- and immunity-related pathways. With these data, and by stratifying PTB by subphenotype, we have identified PTB genes and pathways that can be used as a starting point in further clinical studies.
Article
Full-text available
Childhood obesity remains an epidemic in the U.S. and worldwide. However, little is understood regarding the epigenetic basis of obesity in adolescents. To investigate the cross-sectional association between DNA methylation level in obesity-related genes and body mass index (BMI) percentile, data from 263 adolescents in the population-based Penn State Child Cohort follow-up exam was analysed. Using DNA extracted from peripheral leukocytes, epigenome-wide single nucleotide resolution of DNA methylation in cytosine-phosphate-guanine (CpG) sites and surrounding regions was obtained. We used multivariable-adjusted linear regression models to assess the association between site-specific methylation level and age- and sex-specific BMI percentile. Hypergeometric and permutation tests were used to determine if obesity-related genes were significantly enriched among all intragenic sites that achieved a p < 0.05 throughout the epigenome. Among the 5,669 sites related to BMI percentile with p < 0.05, 28 were identified within obesity-related genes. Obesity-related genes were significantly enriched among 103,466 intragenic sites (Phypergeometric = 0.006; Ppermutation = 0.006). Moreover, increased methylation on one site within SIM1 was significantly related to higher BMI percentile (P = 4.2E-05). If externally validated, our data would suggest that DNA methylation in obesity-related genes may relate to obesity risk in adolescents.
Article
Full-text available
The biological markers of aging used to predict physical health status in older people are of great interest. Telomere shortening, which occurs during the process of cell replication, was initially considered a promising biomarker for the prediction of age and age-related outcomes (e.g., diseases, longevity). However, the high instability in detection and low correlation with age-related outcomes limit the extension of telomere length to the field of prediction. Currently, a growing number of studies have shown that dynamic DNA methylation throughout human lifetime exhibits strong correlation with age and age-related outcomes. Indeed, many researchers have built age prediction models with high accuracy based on age-dependent methylation changes in certain CpG loci. For now, DNA methylation based on epigenetic clocks, namely epigenetic or DNA methylation age, serves as a new standard to track chronological age and predict biological age. Measures of age acceleration (Δage, DNA methylation age – chronological age) have been developed to assess the health status of a person. In addition, there is evidence that an accelerated epigenetic age exists in patients with certain age-related diseases (e.g., Alzheimer’s disease, cardiovascular disease). In this review, we provide an overview of the dynamic signatures of DNA methylation during aging and emphasize its practical utility in the prediction of various age-related outcomes.
Article
Globally, preterm birth is a major public health problem. In India, 3.6 million of the 27 million infants born annually are preterm. Risk stratification of women based on multidimensional risk factors assessed during pregnancy is critical for prevention of preterm birth. A cohort study of pregnant women was initiated in May 2015 at the civil hospital in Gurugram, Haryana, India. Women are enrolled within 20 weeks of gestation and are followed until delivery and once postpartum. The objectives are to identify clinical, epidemiologic, genomic, epigenomic, proteomic, and microbial correlates; discover molecular-risk markers by using an integrative-omics approach; and generate a risk-prediction algorithm for preterm birth. We describe here the longitudinal study design, methodology of data collection, and the repositories of data, biospecimens, and ultrasound images being created. A total of 4,326 pregnant women, with documented evidence of recruitment before 20 weeks of gestation, have been enrolled through March 2018. We report baseline characteristics and outcomes of the first 2,000 enrolled participants. A high frequency of preterm births (14.9% among 1,662 live births) is noteworthy. The cohort database and the repositories will become global resources to answer critical questions on preterm birth and other birth outcomes.