Anda di halaman 1dari 5

Molecular and Cellular Endocrinology 435 (2016) 2e6

Contents lists available at ScienceDirect

Molecular and Cellular Endocrinology


j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m/ l o c a t e / m c e

Maternal obesity and prenatal programming


a b, *
Summer Elshenawy , Rebecca Simmons
a Childrens Hospital of Philadelphia, 3516 Civic Center Boulevard, Philadelphia, PA 19104, USA
b Perelman School of Medicine at University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA

article info abstract

Article history: Obesity is a significant and increasing public health concern in the United States and worldwide. Clinical and epidemiological
Received 28 March 2016 evidence clearly shows that genetic and environmental factors contribute to the increased susceptibility of humans to obesity
Received in revised form and its associated comorbidities; the interplay of these factors is explained by the concept of epigenetics. The impact of
1 July 2016
maternal obesity goes beyond the newborn period; fetal programming during the critical window of pregnancy, can have long
Accepted 1 July 2016
term detrimental effects on the offspring as well as future generations. Emerging evidence is uncovering a link between the
Available online 5 July 2016
clinical and molecular findings in the offspring with epigenetic changes in the setting of maternal obesity. Research targeted
towards reducing the transgenerational propagation and develop-mental programming of obesity is vital in reducing the
Keywords:
increasing rates of disease.
Developmental programming
Maternal obesity Epigenetics
2016 Elsevier Ireland Ltd. All rights reserved.
Obesity in pregnancy
Fetal origins of adult disease

Obesity is a significant and increasing public health concern in the United understanding of the impact of maternal obesity on pregnancy and childhood
States and worldwide (Flegal et al., 2010; Ogden et al., 2006, 2012). In 2010 outcomes. This review will aim to describe clinical data as well as data from
almost one third of adults and 17% of chil-dren and adolescents were obese animal models, with a focus on fetal pro-gramming in the setting of maternal
(Ogden et al., 2012). Obesity in pregnancy is associated with complications obesity.
that include gestational diabetes, intrauterine growth restriction, infants born
large for gestational age, increased caesarian sections and other obstetric 1. Birth weight and obesity
interventions, as well as miscarriages (Schmatz et al., 2010; Lu et al., 2001;
Morin, 1998). Understanding obesity on an epidemiologic and molecular Maternal obesity has been linked to macrosomia (Gaudet et al., 2014),
level has become a significant area of focus within the scientific community. which is a risk factor for obesity and metabolic syndrome later in life. The
Particularly important, is the under-standing of how maternal obesity may variation in fetal growth in the setting of maternal obesity may be related to
affect the outcomes of offspring from the neonatal period to adulthood. The diet composition or vascular factors, and result from differing underlying
impact of maternal obesity goes beyond the newborn period; fetal pro- molecular mechanisms. Epidemi-ologic studies have shown a trend of
gramming during the critical window of pregnancy, can have long term increasing maternal obesity with an associated increase in prevalence of
detrimental effects on the offspring and future generations (Nicholas et al., infants who were born large for gestational age (LGA) (Lu et al., 2001). In a
2016; Simmons, 2008). Environmental exposures in utero, including systematic re-view by Yu et al., pre-pregnancy obesity in women correlated
alterations of the nutritional milieu are particularly important during a time of with an increased risk of having LGA baby, and increased risk of obesity later
such rapid growth (Simmons, 2008). The rising prevalence of obesity and its in life (Yu et al., 2013). A Mendelian randomization study performed by
associated comorbidities, (Bouret et al., 2015), has led to a need for a better Tyrell et al., established genetic alleles associated with maternal obesity and
understanding of the impact of obesity on population health, including an elevated blood glucose were linked to higher birth weight in the offspring
(Tyrrell et al., 2016). There is a clearly established link between birth weight
and obesity later in life. In the U.S. Growing Up Today Study, a cohort study
of over 14,000 adolescents, a 1-kg increment in birth weight in full term
infants was associated with an approximately 50% increase in the risk of
overweight at ages 9e14 year (Gillman et al., 2003).
When adjusted for
* Corresponding author. Hallam Hurt Professor Pediatrics, Perelman School of Medicine maternal BMI, the increase in risk remained
,Childrens Hospital of Philadelphia, USA.
E-mail address: rsimmons@mail.med.upenn.edu (R. Simmons).

http://dx.doi.org/10.1016/j.mce.2016.07.002
0303-7207/ 2016 Elsevier Ireland Ltd. All rights reserved.
S. Elshenawy, R. Simmons / Molecular and Cellular Endocrinology 435 (2016) 2e6 3

significantly elevated at 30%. A study of Danish military conscripts showed islands are CG-rich sequences located near coding sequences and they serve
that even after controlling for birth length and maternal factors, BMI at ages as promoters for their associated genes. Approximately half of mammalian
18e26 strongly correlated with birth weight (Sorensen et al., 1997). Both genes have CpG islands. The methylation status of CpG islands within
paternal and maternal adiposity are correlated with a higher birth weight of promoter sequences works as an essential regulatory element by modifying
the offspring. However, the association is much stronger for the mother the binding affinity of transcrip-tion factors to DNA binding sites. In normal
compared to the father (Parsons et al., 2001; Guillaume et al., 1995; Whitaker cells, most CpG islands remain unmethylated; however, under circumstances
et al., 2010; Oken, 2009) suggesting that the intrauterine environment plays such as oxidative stress, they can become methylated de novo. This aber-rant
an important role in the later development of obesity. In addition to birth methylation is accompanied by local changes in histone modification and
weight, several clinical studies have shown a direct relation-ship between chromatin structure, such that the CpG island and its embedded promoter take
maternal obesity and childhood obesity (Parsons et al., 2001; Boerschmann et on a repressed conformation that is incompatible with gene transcription. It is
al., 2010). In a retrospective cohort study of 8500 low income children in the not known why partic-ular CpG islands are susceptible to aberrant
US, Whitaker et al. demonstrated a two-fold increase in prevalence of early methylation. DNA methylation is commonly associated with gene silencing
childhood obesity in children who were born to obese mothers (Whitaker, and con-tributes to X-chromosomal inactivation, genomic imprinting, and
2004). Smith et al. looked at metabolic features of children born before or transcriptional regulation of tissue-specific genes during cellular
after mothers underwent bariatric surgery. They found that children born after differentiation (Pinney and Simmons, 2012). In the case of maternal obesity
maternal bariatric surgery had had lower birth weight, lower prevalence of and fetal programming, differential methylation of retinoid X receptor- a
severe obesity adjusted for age and gender, greater insulin sensitivity, (RXRA) gene in umbilical cord tissue, was shown to be associated with
improved lipid profile, lower C-reactive protein, and leptin and increased childhood fat mass, when adjusted for sex (Godfrey et al., 2011).
ghrelin than offspring born before maternal bariatric surgery (Smith et al.,
2009). Guenard et al. also looked at offspring before and after bariatric
surgery, finding epigenetic changes in genes involved in glucoregulatory,
including insulin sensitivity, inflammatory, and vascular disease genes
conferring a more favorable cardiometabolic profile in the offspring born after 2.2. Histone modifications
maternal bariatric surgery (Guenard et al., 2013). Catalano et al. specifically
discusses that pre-pregnancy obesity has a stronger association with metabolic In eukaryotes, the nucleosome consists of DNA wrapped around an
alterations of the fetus, than gestational weight gain. In one study, they octameric complex of two molecules of each of the four his-tones: H2A,
2 H2B, H3, and H4. The amino termini of histones can be modified by
showed the maternal pre-pregnancy BMI greater than 30 kg/m was asso-
ciated with increased body fat percentage at age 8. Furthermore, they acetylation, methylation, sumoylation, phosphoryla-tion, glycosylation, and
demonstrated that at age 8, children of obese mothers had higher systolic ADP ribosylation. The most common his-tone modifications involve
blood pressures, waist circumference, triglycerides, insulin resistance and acetylation and methylation of lysine residues in the amino termini of H3 and
leptin levels (Catalano et al., 2009). H4. Increased acetylation induces transcription activation, whereas decreased
acetylation usually induces transcription repression. Methylation of histones,
on the other hand, is associated with both transcription repression and
activation. Moreover, lysine residues can be mono-, di-, or trimethylated in
vivo, providing an additional level of regulation (Pinney and Simmons, 2012).
Histone methylation can influence DNA methylation patterns and vice versa
2. Epigenetics (Cedar and Bergman, 2009). For example, methylation of lysine 9 on histone
3 (H3) promotes DNA methylation, while CpG methylation stimulates
Clinical and epidemiological evidence clearly shows that genetic and methylation of lysine 9 on H3 (Schubeler et al., 2000). Recent evi-dence
environmental factors contribute to the increased suscepti-bility of humans to indicates that this reciprocal relationship between histone methylation and
obesity and its associated comorbidities; the interplay of these factors is DNA methylation might be accomplished by direct interactions between
explained by the concept of epigenetics (Bouret et al., 2015). Epigenetics histone and DNA methyltransferases (Cedar and Bergman, 2009). Thus,
explains, as Barker describes, developmental plasticity in which chromatin modifications induced by adverse stimuli are self-reinforcing and
environment impacts gene expression, particularly during vulnerable times of can propagate.
development (Barker, 2004). Epigenetics controls differentiation and develop-
ment of different cell types by modulating chromatin architecture. It is a
dynamic process that is influenced by environmental factors. The mechanisms
include DNA methylation, histone modification and the presence of 2.3. Noncoding RNAs
noncoding RNAs and microRNAs (Nicholas et al., 2016; Bouret et al., 2015;
Pinney and Simmons, 2012). The epigenetic modifications can lead to stable Noncoding RNAs such as microRNAs (miRNA), small RNAs, and long
propagation with transgenerational effects. Although human data is still or large RNAs, play a significant role in epigenetic gene regu-lation and
limited, emerging evidence is uncovering a link between the clinical and chromosomal dynamics and transcription (Bernstein and Allis, 2005). With
molecular findings in the offspring with epigenetic changes in the setting of the discovery that most of the eukaryotic ge-nomes are transcribed into RNAs
maternal obesity. that have no protein-coding po-tential, evidence has emerged of the different
regulatory functions of noncoding RNAs. Studies have shown differential
expression of miRNAs in the amnion, plasma, and other tissues of obese
mothers (Nardelli et al., 2014; Yan et al., 2013; Carreras-Badosa et al., 2015).
2.1. DNA methylation Yan et al. demonstrated that the offspring of obese pregnant ewes had
decreased expression of miRNA let-7g in skeletal muscle, which correlated
DNA methylation is a class of epigenetic regulation, in which a cytosine with increased adipose deposition in skeletal muscle (Yan et al., 2013). This
base is modified by DNA methyltransferase at the C5 po-sition of cytosine, a demonstrates the role non-coding RNAs may play in regulation of
reaction that is carried out by various members of a single family of enzymes. adipogenesis through differential gene expression. Carreras-Badosa et al.
Approximately 70% of CpG di-nucleotides in human DNA are constitutively conducted a study in which plasma circulating miRNAs were measured in
methylated, whereas most of the unmethylated CpGs are located in CpG normal pregnancies
islands. CpG
4 S. Elshenawy, R. Simmons / Molecular and Cellular Endocrinology 435 (2016) 2e6

as well as pregnancies complicated by pre-gestational and gesta-tional and insulin resistance has been studied in the setting of maternal obesity and
obesity. In this study 13 circulating miRNAs were differen-tially expressed in as a separate entity. Insulin sensitivity is reduced in the pregnant state, and
gestational obesity when compared to normal pregnancies, many of which are while placental factors may contribute, emerging evidence has implicated
associated with altered meta-bolism in the mother including weight gain, other factors such as elevated cytokines, including TNF alpha, and altered
glucose tolerance, in-sulin sensitivity, and serum lipid levels. Specific lipid metabolism as culprits to this change during gestation (Catalano, 2010;
miRNAs were also associated with placental weight and birth weight, as well Schaefer-Graf et al., 2008). These factors are exacerbated in the setting of
as growth during infancy, after controlling maternal factors (Carreras-Badosa maternal obesity leading to more significant insulin resistance. Insulin also
et al., 2015). plays an important role on neuronal development with implications in central
regulation of energy homeostasis (Bouret et al., 2015; Puro and Agardh, 1984;
Heidenreich and Toledo, 1989a, 1989b). A causal relationship has been
3. Macronutrient impact on fetal development clearly established between maternal glucose intolerance and macrosomia.
Increased maternal concentrations of glucose and amino acids stimulate the
The adverse effects of maternal obesity on the fetus are likely fetal pancreas to secrete exaggerated amounts of insulin, and the fetal liver to
multifactorial, involving hormonal, inflammatory, and metabolic alterations. produce higher levels of insulin dependent growth factors. Fetal
Macronutrient intake influenced by maternal diet and body composition likely hyperinsulinism stimulates the growth of fetal adi-pose tissue and of other
plays a role in the metabolic conditioning of the fetus in utero. Metabolomic insulin-responsive tissues, often leading to macrosomia (Simmons, 2008).
analysis of umbilical cord blood showed that accelerated early childhood Furthermore, a gene expression study by Radaelli et al., demonstrated
weight gain was associ-ated with differential regulation of metabolites related significant alterations in genes involved in lipid metabolism in the placentas
to food and plant component (Isganaitis et al., 2015). Furthermore, while of mothers with gestational and type 1 diabetes mellitus (Radaelli et al.,
there is a physiologic increase in fasting triglycerides throughout preg-nancy, 2009). In a large international multicenter observational study consisting of
mothers with higher pre-pregnancy BMIs see a greater in-crease in fasting 25,000 women, maternal GDM and obesity were independently associated
triglycerides and free fatty acids, demonstrating alteration in lipid metabolism with increased birth weights as well as other adverse pregnancy outcomes.
in the obese mother (Di Cianni et al., 2005). DiCianni et al. showed maternal Their combined effect was greater than either alone (Catalano et al., 2012;
fasting triglycerides corre-lated with birth weight (Di Cianni et al., 2005). In a Group, 2010). Macrosomia results from growth of excess fetal adipose tissue
study looking at differential gene expression in maternal obesity, Carty et al. and other insulin responsive tissue in response to increased insulin secretion
found reduced expression of COX7A2 in subcutaneous tissue of obese from fetal pancreas (Simmons, 2008). Macrosomia can lead to increased
pregnant women at term. COX7A2 is a mitochondrial protein, with key roles obstetric complications including perinatal asphyxia, shoulder dystocia, and
in steroidogenesis and oxidative stress regulation and could provide a link increased rate of C-sections. Furthermore, in utero exposure to maternal
between inflammation and obesity-related pregnancy complications (Carty et diabetes is also associated with increased risk of devel-opment of obesity,
al., 2014). diabetes, and metabolic syndrome in childhood and adulthood. Even in
patients with similar birth weights, infants born to mothers with GDM had
increased body fat based anthro-prometric measurements, compared to infants
Maternal obesity and fetal programming has been studied in multiple born to mothers with normal glucose tolerance (Catalano et al., 2003; Sewell
animal models. Various models evaluate the impact of different types of diet, et al., 2006). A study conducted by Dabalea et al. looked at families that
and may indicate that the macronutrient content of the diet, may impact the consisted of at least one sibling before and one sibling born after maternal
mechanism of fetal program-ming. In animal experiments, high fat diet leads diagnosis of type 2 diabetes. This study showed increased BMI and increased
to poor glycemic control and increased adiposity in offspring (Simmons, risk of diabetes in the siblings born after mothers were diagnosed. By looking
2008; Bouret et al., 2015). In a unique study design, Sasson et al. per-formed at siblings of the same parents, this study controlled for genetic factors,
a reciprocal early two-cell embryo transfer between mice fed different diets suggesting that the intra-uterine environment predisposes offspring to obesity
prior to and during pregnancy. Pre-gestational exposure to high fat diet and diabetes (Dabelea et al., 2000). In the human infant, birth weight and
resulted in growth restricted fetuses and newborn pups but no effect on adult infant adiposity is positively correlated with leptin levels. Both cord blood
weight, body fat content or leptin levels. They observed differential gene and plasma concentrations are increased in infants of diabetic mothers (Law
expression of several imprinted genes in the placenta of the pre-pregnancy et al., 1992; Fall et al., 1995). In a study of a cohort consisting of 64 mothers,
high fat diet animals suggesting an epigenetic alteration in the germ line in the 33 GDM and 31 controls together with their 9-year-old offspring, an elevated
setting of maternal obesity. Gestational exposure to maternal high fat diet child leptin was highly correlated with elevated maternal leptin in GDM
(HFD) resulted in increased body fat, elevated leptin levels, and impaired mothers (Shekhawat et al., 1998). It is conceivable that programming of leptin
glucose tolerance in offspring. In contrast, the offspring of mothers who were regulatory pathways may be another causal mechanism linking obesity to
fed a HFD during pre-conceptional period and throughout gestation, had exposure to diabetes in pregnancy (Simmons, 2008).
significantly higher body fat content, but while the females were not
inherently glucose intol-erant, the males were. There were many more genes
differentially expressed in the HFD/HFD group suggesting a compound effect
of the pre-conceptional and gestational exposure. Many of the genes that were
differentially expressed in the placenta play a role in inflammation (Sasson et
al., 2015).

More recently, animal studies have also looked at a western diet model, 4. Inflammation and developmental programming
which is high in simple sugars to more closely mimic human diets, which
result in alterations in glucose metabolism, increased adiposity, as well as Obesity triggers a chronic low-grade state of inflammation characterized
alterations in central appetite reg-ulatory systems in the offspring (Bouret et by abnormal cytokines and adipokine production (Hotamisligil, 2006).
al., 2015). Thus varia-tions in different obesogenic diets could explain the Inflammatory markers are systemically elevated in both human and animal
variable presentation seen in population studies. In addition to fat, alter-ations studies of obese subjects (Segovia et al., 2014). The health of a pregnancy
in glucose metabolisms in the setting of gestational diabetes relies on a delicate balance of pro and anti-inflammatory factors. There
is evidence of
S. Elshenawy, R. Simmons / Molecular and Cellular Endocrinology 435 (2016) 2e6 5

inflammation triggered by obesity in the placenta, in the form of macrophage


infiltration and elevated cytokine expression (Challier et al., 2008). The
placenta is a vital organ in sustaining the fetus and maintaining a health
maternal-fetal interface, thus the inflamma-tory state in the setting of obesity
can lead to detrimental effects that can explain adverse outcomes of
pregnancy as well as long-term effects on the fetus (Schmatz et al., 2010;
Madan et al., 2009). Obesity results in adipocyte hypertrophy, which can lead
to cell hypoxia and necrosis, triggering an inflammatory cascade of
chemokine release and metabolic dysregulation including the transport of
fatty acids (Sun et al., 2011). Free fatty acids have been shown to activate the
TLR4 signaling, thus creating a link between obesity and the innate immune
system (Schaeffler et al., 2009). Zhu et al. observed elevated free fatty acids,
cholesterol, and tri-glycerides in fetal circulation from obese ewes which were
accompanied by upregulation of toll-like receptor 4 (TLR4), NF- kB, and JNK
(Zhu et al., 2010a) as well as increased mRNA expression fatty acid transport
proteins (FATPs) in the placenta, peroxisome proliferator-activated receptor
(PPAR)-g, which regulates the expression of FATPs (Zhu et al., 2010b).
Additional studies in ro-dents have also implicated (PPAR)- g in metabolic
dysregulation in adipose tissue as well as skeletal muscle the setting of
maternal obesity (Shankar et al., 2008; Samuelsson et al., 2008; Bayol et al.,
2005).

Figure 1. The effect of maternal obesity on the offspring is multifactorial and may result from
alterations in macronutrient availability (elevated fats, carbohydrates and decreased insulin
sensitivity), epigenetic modifications (DNA methylation, histone modification, and noncoding
4.1. Adipokines RNAs) and inflammation (adipokine and cytokine release). There is often interplay between
these factors resulting in a compounded effect on the offspring. The impact is evident in short
term outcomes such as fetal growth as well as long term outcomes including increased risk of
Adipokines are cytokines released by adipocytes that have im-plications
childhood obesity and ultimately metabolic syndrome and obesity in the adult. There are also
in regulation of a number of diverse processes including lipogenesis, trans-generational effects with an ongoing cycle of offspring of obese mothers going on to have
angiogenesis inflammation, and metabolic processes. Adipose tissue serves to offspring with exposure to the same environmental and epigenetic factors leading to an ongoing
store fat, and had recently been thought to be relatively inert. However, the epidemic of obesity.
recognition of adipokines has clearly established the role of adipocyte in
maintaining energy homeostasis (Simmons, 2008; Pinney and Simmons,
2012; Segovia et al., 2014; Sun et al., 2011). Some of these adipokines
regulators in the fetal liver and placental labyrinth at term. Notably they found
include adiponectin, leptin, and resistin among others (Pinney and Simmons,
alterations in components of the histone acetylation pathway. Transcript levels
2012) with ongoing research resulting in identification of novel adipokines
of arginine methyltransferases Prmt1 and Prmt7 were upregulated in the liver
(Segovia et al., 2014). Leptin is a well-studied adipokine, that serves as a
of fetuses born to obese mothers. Prmts catalyze the methylation of arginine
satiety factor with receptors in the hy-pothalamus; mutations in the leptin
histone resi-dues and are implicated in hepatic gluconeogenesis (Panchenko et
gene can lead to an obese phenotype. It can be used as a circulating signal of
al., 2016). They also saw alterations in the lysine acetylation pathway in
fat mass, and an indicator of the important regulatory role played by
offspring of obese mothers. They also found differential expression in KATs
adipocytes (Zhang et al., 1994). Furthermore, leptin and its receptor are
and HDACs, which are lysine demethylases, with implications in metabolic
expressed in murine placenta, indicating it may have a role in fetal growth
processes including adipogenesis, he-patic lipid metabolism, regulation of
and development (Hoggard et al., 1997). Levels of leptin in cord blood are
circadian rhythm, energy expenditure, gluconeogenesis and lipogenesis
related to birth weight in offspring (Pinney and Simmons, 2012). Resistin is
(Panchenko et al., 2016).
highly expressed in visceral fat; it an-tagonizes insulin action leading to
insulin resistance. In the setting of maternal obesity, dietary intake has been
shown to permanently influence metabolism of offspring through epigenetic
mechanisms, with transgenerational effects. Studies have shown alterations of
acetylation and methylation of histone H3K9 in the promoter re-gion of 5. Conclusion
adiponectin, an adipokine that possesses anti-inflammatory and insulin
sensitizing properties. Adiponectin possesses anti-inflammatory and insulin The adverse outcomes associated with maternal obesity arise from
sensitizing properties and is reduced in obese patients with insulin resistance genetic, epigenetic, metabolic and inflammatory alterations with a lasting
and type 2 DM. In rodent models, maternal high fat diet lead to epigenetic impact on fetal development (See Fig. 1). Research targeted towards reducing
changes in fat and skeletal muscle. Alterations were seen in the acetylation the transgenerational propagation and developmental programming of obesity
and methylation of H3K9 of the adiponectin promoter and changes in is vital in reducing the increasing rates of disease. Recognizing the risk
methylation of histone H4K20 within the leptin promoter (Nicholas et al., factors for obesity and its associated comorbidities will allow clinicians to
2016). Panchenko et al. performed a study looking at the epigenetic effects of intervene prior to some of the critical windows of development. These in-
obesity and weight loss during gestation in a mouse model fed a high fat diet terventions may be able to reverse the programming during these vulnerable
with a phenotype of fetal growth restriction (Panchenko et al., 2016). They periods, in order to attenuate the effects on future generations (Segovia et al.,
found that maternal obesity was associated with altered expression of some 2014).
epigenetic

Funding

T32 (GM008638) NIH/NIGMS Medical Genetics Research


6 S. Elshenawy, R. Simmons / Molecular and Cellular Endocrinology 435 (2016) 2e6

Training Grant. Nardelli, C., et al., 2014. Characterization and predicted role of the microRNA expression
profile in amnion from obese pregnant women. Int. J. Obes. (Lond) 38 (3), 466e469.

Reference Nicholas, L.M., et al., 2016. The early origins of obesity and insulin resistance:
timing, programming and mechanisms. Int. J. Obes. (Lond) 40 (2), 229e238.
Barker, D.J., 2004. The developmental origins of adult disease. J. Am. Coll. Nutr. 23 (6 Suppl), Ogden, C.L., et al., 2006. Prevalence of overweight and obesity in the United States, 1999-
588Se595S. 2004. JAMA 295 (13), 1549e1555.
Bayol, S.A., Simbi, B.H., Stickland, N.C., 2005. A maternal cafeteria diet during gestation and Ogden, C.L., et al., 2012. Prevalence of obesity in the United States, 2009-2010. NCHS Data
lactation promotes adiposity and impairs skeletal muscle development and metabolism in Brief. (82), 1e8.
rat offspring at weaning. J. Physiol. 567 (Pt 3), 951e961. Oken, E., 2009. Maternal and child obesity: the causal link. Obstet. Gynecol. Clin.
North Am. 36 (2), 361e377 ix-x.
Bernstein, E., Allis, C.D., 2005. RNA meets chromatin. Genes Dev. 19 (14), 1635e1655. Panchenko, P.E., et al., 2016. Expression of epigenetic machinery genes is sensitive to maternal
Boerschmann, H., et al., 2010. Prevalence and predictors of overweight and insulin resistance in obesity and weight loss in relation to fetal growth in mice. Clin. Epigenetics 8, 22.
offspring of mothers with gestational diabetes mellitus. Diabetes
Care 33 (8), 1845e1849. Parsons, T.J., Power, C., Manor, O., 2001. Fetal and early life growth and body mass index from
Bouret, S., Levin, B.E., Ozanne, S.E., 2015. Gene-environment interactions controlling energy birth to early adulthood in 1958 British cohort: longitudinal study. BMJ 323 (7325),
and glucose homeostasis and the developmental origins of obesity. Physiol. Rev. 95 (1), 1331e1335.
47e82. Pinney, S.E., Simmons, R.A., 2012. Metabolic programming, epigenetics, and gesta- tional
Carreras-Badosa, G., et al., 2015. Altered Circulating miRNA expression Pro file in diabetes mellitus. Curr. Diab Rep. 12 (1), 67e74.
Pregestational and gestational obesity. J. Clin. Endocrinol. Metab. 100 (11), E1446eE1456. Puro, D.G., Agardh, E., 1984. Insulin-mediated regulation of neuronal maturation.
Science 225 (4667), 1170e1172.
Carty, D., et al., 2014. Differential gene expression in obese pregnancy. Pregnancy Hypertens. 4 Radaelli, T., et al., 2009. Differential regulation of genes for fetoplacental lipid pathways in
(3), 232e233. pregnancy with gestational and type 1 diabetes mellitus. Am. J. Obstet. Gynecol. 201 (2),
Catalano, P.M., 2010. Obesity, insulin resistance, and pregnancy outcome. Repro- duction 140 209 e1-209 e10.
(3), 365e371. Samuelsson, A.M., et al., 2008. Diet-induced obesity in female mice leads to offspring
Catalano, P.M., et al., 2003. Increased fetal adiposity: a very sensitive marker of abnormal in hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of
utero development. Am. J. Obstet. Gynecol. 189 (6), 1698e1704. developmental programming. Hypertension 51 (2), 383e392.
Catalano, P.M., et al., 2009. Perinatal risk factors for childhood obesity and meta- bolic Sasson, I.E., et al., 2015. Pre-gestational vs gestational exposure to maternal obesity
dysregulation. Am. J. Clin. Nutr. 90 (5), 1303e1313. differentially programs the offspring in mice. Diabetologia 58 (3), 615e624.
Catalano, P.M., et al., 2012. The hyperglycemia and adverse pregnancy outcome study: Schaefer-Graf, U.M., et al., 2008. Maternal lipids as strong determinants of fetal environment
associations of GDM and obesity with pregnancy outcomes. Diabetes Care 35 (4), and growth in pregnancies with gestational diabetes mellitus. Diabetes Care 31 (9),
780e786. 1858e1863.
Cedar, H., Bergman, Y., 2009. Linking DNA methylation and histone modification: Schaeffler, A., et al., 2009. Fatty acid-induced induction of Toll-like receptor-4/nu- clear factor-
patterns and paradigms. Nat. Rev. Genet. 10 (5), 295e304. kappaB pathway in adipocytes links nutritional signalling with innate immunity.
Challier, J.C., et al., 2008. Obesity in pregnancy stimulates macrophage accumula- tion and Immunology 126 (2), 233e245.
inflammation in the placenta. Placenta 29 (3), 274e281. Schmatz, M., et al., 2010. Maternal obesity: the interplay between in flammation, mother and
Dabelea, D., et al., 2000. Intrauterine exposure to diabetes conveys risks for type 2 diabetes and fetus. J Perinatol 30 (7), 441e446.
obesity: a study of discordant sibships. Diabetes 49 (12), 2208e2211. Schubeler, D., et al., 2000. Genomic targeting of methylated DNA: in fluence of methylation on
transcription, replication, chromatin structure, and histone acetylation. Mol. Cell Biol. 20
Di Cianni, G., et al., 2005. Maternal triglyceride levels and newborn weight in pregnant women (24), 9103e9112.
with normal glucose tolerance. Diabet. Med. 22 (1), 21e25. Segovia, S.A., et al., 2014. Maternal obesity, in flammation, and developmental programming.
Fall, C.H., et al., 1995. Fetal and infant growth and cardiovascular risk factors in women. BMJ Biomed. Res. Int. 2014, 418975.
310 (6977), 428e432. Sewell, M.F., et al., 2006. Increased neonatal fat mass, not lean body mass, is associated with
Flegal, K.M., et al., 2010. Prevalence and trends in obesity among US adults, 1999- 2008. maternal obesity. Am. J. Obstet. Gynecol. 195 (4), 1100e1103.
JAMA 303 (3), 235e241. Shankar, K., et al., 2008. Maternal obesity at conception programs obesity in the offspring. Am.
Gaudet, L., et al., 2014. Maternal obesity and occurrence of fetal macrosomia: a systematic J. Physiol. Regul. Integr. Comp. Physiol. 294 (2). R528-38.
review and meta-analysis. Biomed. Res. Int. 2014, 640291. Shekhawat, P.S., et al., 1998. Neonatal cord blood leptin: its relationship to birth weight, body
Gillman, M.W., et al., 2003. Maternal gestational diabetes, birth weight, and adolescent obesity. mass index, maternal diabetes, and steroids. Pediatr. Res. 43 (3), 338e343.
Pediatrics 111 (3), e221-6.
Godfrey, K.M., et al., 2011. Epigenetic gene promoter methylation at birth is asso- ciated with Simmons, R., 2008. Perinatal programming of obesity. Semin. Perinatol. 32 (5), 371e374.
childs later adiposity. Diabetes 60 (5), 1528e1534.
Group, H.S.C.R., 2010. Hyperglycaemia and Adverse Pregnancy Outcome (HAPO) Study: Smith, J., et al., 2009. Effects of maternal surgical weight loss in mothers on intergenerational
associations with maternal body mass index. BJOG 117 (5), 575e584. transmission of obesity. J. Clin. Endocrinol. Metab. 94 (11), 4275e4283.
Guenard, F., et al., 2013. Differential methylation in glucoregulatory genes of offspring born
before vs. after maternal gastrointestinal bypass surgery. Proc. Natl. Acad. Sci. U S A 110 Sorensen, H.T., et al., 1997. Relation between weight and length at birth and body mass index in
(28), 11439e11444. young adulthood: cohort study. BMJ 315 (7116), 1137.
Guillaume, M., et al., 1995. Familial trends of obesity through three generations: the Belgian- Sun, K., Kusminski, C.M., Scherer, P.E., 2011. Adipose tissue remodeling and obesity.
Luxembourg child study. Int. J. Obes. Relat. Metab. Disord. 19 (3), S5eS9. J. Clin. Invest 121 (6), 2094e2101.
Tyrrell, J., et al., 2016. Genetic evidence for causal relationships between maternal obesity-
Heidenreich, K.A., Toledo, S.P., 1989. Insulin receptors mediate growth effects in cultured fetal related traits and birth weight. JAMA 315 (11), 1129e1140.
neurons. I. Rapid stimulation of protein synthesis. Endocrinology 125 (3), 1451e1457. Whitaker, R.C., 2004. Predicting preschooler obesity at birth: the role of maternal obesity in
early pregnancy. Pediatrics 114 (1), e29-36.
Heidenreich, K.A., Toledo, S.P., 1989. Insulin receptors mediate growth effects in cultured fetal Whitaker, K.L., et al., 2010. Comparing maternal and paternal intergenerational transmission of
neurons. II. Activation of a protein kinase that phosphorylates ribosomal protein S6. obesity risk in a large population-based sample. Am. J. Clin. Nutr. 91 (6), 1560e1567.
Endocrinology 125 (3), 1458e1463.
Hoggard, N., et al., 1997. Leptin and leptin receptor mRNA and protein expression in the Yan, X., et al., 2013. Maternal obesity downregulates microRNA let-7g expression, a possible
murine fetus and placenta. Proc. Natl. Acad. Sci. U S A 94 (20), 11073e11078. mechanism for enhanced adipogenesis during ovine fetal skeletal muscle development. Int.
Hotamisligil, G.S., 2006. Inflammation and metabolic disorders. Nature 444 (7121), 860e867. J. Obes. (Lond) 37 (4), 568e575.
Yu, Z., et al., 2013. Pre-pregnancy body mass index in relation to infant birth weight and
Isganaitis, E., et al., 2015. Associations of cord blood metabolites with early child- hood obesity offspring overweight/obesity: a systematic review and meta-analysis. PLoS One 8 (4),
risk. Int. J. Obes. (Lond) 39 (7), 1041e1048. e61627.
Law, C.M., et al., 1992. Early growth and abdominal fatness in adult life. J. Epidemiol. Zhang, Y., et al., 1994. Positional cloning of the mouse obese gene and its human homologue.
Community Health 46 (3), 184e186. Nature 372 (6505), 425e432.
Lu, G.C., et al., 2001. The effect of the increasing prevalence of maternal obesity on perinatal Zhu, M.J., et al., 2010. Maternal obesity up-regulates in flammatory signaling path-ways and
morbidity. Am. J. Obstet. Gynecol. 185 (4), 845e849. enhances cytokine expression in the mid-gestation sheep placenta. Placenta 31 (5),
Madan, J.C., et al., 2009. Maternal obesity and markers of in flammation in preg-nancy. 387e391.
Cytokine 47 (1), 61e64. Zhu, M.J., et al., 2010. Maternal obesity markedly increases placental fatty acid transporter
Morin, K.H., 1998. Perinatal outcomes of obese women: a review of the literature. expression and fetal blood triglycerides at midgestation in the ewe. Am. J. Physiol. Regul.
J. Obstet. Gynecol. Neonatal Nurs. 27 (4), 431e440. Integr. Comp. Physiol. 299 (5). R1224-31.

Anda mungkin juga menyukai