Hostname: page-component-7c8c6479df-27gpq Total loading time: 0 Render date: 2024-03-28T13:26:13.351Z Has data issue: false hasContentIssue false

Epigenetics and maternal nutrition: nature v. nurture

Published online by Cambridge University Press:  29 November 2010

Rebecca Simmons*
Affiliation:
Department of Pediatrics, Children's Hospital Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
*
Corresponding author: Dr R. Simmons, fax +1 215 573 7627, email rsimmons@mail.med.upenn.edu
Rights & Permissions [Opens in a new window]

Abstract

Under- and over-nutrition during pregnancy has been linked to the later development of diseases such as diabetes and obesity. Epigenetic modifications may be one mechanism by which exposure to an altered intrauterine milieu or metabolic perturbation may influence the phenotype of the organism much later in life. Epigenetic modifications of the genome provide a mechanism that allows the stable propagation of gene expression from one generation of cells to the next. This review highlights our current knowledge of epigenetic gene regulation and the evidence that chromatin remodelling and histone modifications play key roles in adipogenesis and the development of obesity. Epigenetic modifications affecting processes important to glucose regulation and insulin secretion have been described in the pancreatic β-cells and muscle of the intrauterine growth-retarded offspring, characteristics essential to the pathophysiology of type-2 diabetes. Epigenetic regulation of gene expression contributes to both adipocyte determination and differentiation in in vitro models. The contributions of histone acetylation, histone methylation and DNA methylation to the process of adipogenesis in vivo remain to be evaluated.

Type
Conference on ‘Nutrition and health: cell to community’
Copyright
Copyright © The Author 2010

Abbreviations:
IUGR

intrauterine growth retarded

Pdx1

pancreatic duodenal homeobox-1

SGA

small for gestational age; T2D, type-2 diabetes

Link of low birth weight to later development of disease

It is becoming increasingly apparent that the in utero environment in which a fetus develops may have long-term effects on subsequent health and survival(Reference Hales and Barker1, Reference Kermack2). The landmark cohort study of 300 000 men by Ravelli and colleagues showed that exposure to the Dutch famine of 1944–45 during the first one-half of pregnancy resulted in significantly higher obesity rates at 19 years of age(Reference Ravelli, Stein and Susser3). Subsequent studies of English men demonstrated a relationship between low birth weight and the later development of CVD(Reference Barker4) and impaired glucose tolerance(Reference Valdez, Athens and Thompson5Reference Forsen, Eriksson and Tuomilehto8). Other studies of populations in the USA(Reference Rich-Edwards, Colditz and Stampfer9Reference Bavdekar, Sachdev and Fall11), Sweden(Reference Whincup, Cook and Adshead12), France(Reference Martin, McCarthy and Smith13, Reference Law, Shiell and Newsome14), Norway(Reference Li, Johnson and Goran15) and Finland(Reference Boney, Verma and Tucker16), have demonstrated a significant correlation between low birth weight and the later development of adult diseases. The associations with low birth weight and increased risk of CHD, stroke and type-2 diabetes (T2D) remain strong, even after adjusting for lifestyle factors (e.g. smoking, physical activity, occupation, income, dietary habits and childhood socio-economic status) and occur independent of the current level of obesity or exercise(Reference Clausen, Borch-Johnsen and Pedersen17, Reference Flanagan, Moore and Godsland18).

Low birth weight and insulin secretion

It remains controversial as to whether the adverse effects of intrauterine growth retardation on glucose homeostasis are mediated through programming of the fetal endocrine pancreas(Reference Hales and Barker1). Growth-retarded fetuses and newborns have been reported to have both a reduced population of pancreatic β-cells(Reference Van Assche, De Prins and Aerts19) or a normal percentage of pancreatic area occupied by β-cells(Reference Beringue, Blondeau and Castellotti20). Both of these studies were observational, morphometric analyses were not optimal, and only a small number of fetuses/newborns were examined. It is likely that a significant proportion, but not all growth-retarded fetuses will have reduced β-cell numbers. A more clinically relevant consideration is the impact of fetal growth retardation upon β-cell function.

Intrauterine growth-retarded (IUGR) fetuses have been found to exhibit lower insulin and glucose levels and higher glucose:insulin ratio in the third trimester as measured by cordocentesis(Reference Econimides, Proudler and Nicolaides21). Two recent studies showed that IUGR infants display decreased pancreatic β-cell function, but increased insulin sensitivity at birth(Reference Setia, Sridhar and Bhat22, Reference Bazaes, Salazar and Pittaluga23). Low birth weight has been associated with reduced insulin response after glucose ingestion in young non-diabetic men, whereas other studies have found no impact of low birth weight upon insulin secretion(Reference Clausen, Borch-Johnsen and Pedersen17, Reference Flanagan, Moore and Godsland18). However, none of these earlier studies adjusted for the corresponding insulin sensitivity, which has a profound impact upon insulin secretion. Therefore, Jensen et al.(Reference Jensen, Storgaard and Dela24) measured insulin secretion and insulin sensitivity in a well-matched Caucasian population of 19-year-old glucose-tolerant men with birth weights either below the 10th percentile (small for gestational age (SGA)) or between the 50th and 75th percentile (controls). To eliminate the major confounders such as ‘diabetes genes’, none of the participants had a family history of diabetes, hypertension, or IHD. There was no difference between the groups with regard to current weight, BMI, body composition and lipid profile. When controlled for insulin sensitivity, insulin secretion was reduced by 30%. Insulin sensitivity, however, was normal in the SGA subjects. The investigators hypothesised that defects in insulin secretion may precede defects in insulin action and that once SGA individuals accumulate body fat, they will develop insulin resistance(Reference Jensen, Storgaard and Dela24).

Genetics v. environment

Several epidemiological and metabolic studies of twins and first-degree relatives of patients with T2D have demonstrated an important genetic component of diabetes(Reference Barnett, Eff and Leslie25Reference Vaag, Henricksen and Madsbad28). The association between low birth weight and risk of T2D in some studies could theoretically be explained by a genetically determined reduced fetal growth rate. In other words, the genotype responsible for T2D may itself cause retarded fetal growth in utero. This forms the basis for the fetal insulin hypothesis, which suggests that genetically determined insulin resistance could result in low insulin-mediated fetal growth in utero as well as insulin resistance in childhood and adulthood(Reference Hattersley and Tooke29). Insulin is one of the major growth factors in fetal life, and monogenic disorders that affect fetal insulin secretion or fetal insulin resistance also affect fetal growth. Mutations in the gene encoding glucokinase have been identified that result in low birth weight and maturity onset diabetes of the young(Reference Froguel, Zouali and Vionnet30, Reference Hattersley, Beards and Ballantyne31).

The recently described T2D susceptibility gene transcription factor 7-like 2 confers a risk-allele frequency of approximately 30%(Reference Grant, Thorleifsson and Reynisdottir32). Studies of non-diabetic subjects show that transcription factor 7-like 2 diabetes-risk genotypes alter insulin secretion(Reference Damcott, Pollin and Reinhart33Reference Munoz, Lok and Gower35). A large study of 24 053 subjects combined from six studies demonstrated that transcription factor 7-like 2 is the first T2D gene to be reproducibly associated with altered birth weight. Each maternal copy of the T allele at re7903146 increased offspring birth weight by 30 g, and the investigators suggest that the most likely mechanism is through reduced maternal-insulin secretion resulting in maternal hyperglycemia and increased insulin-mediated fetal growth(Reference Freathy, Weedon and Bennett36).

Recent genetic studies suggest that the increased susceptibility to T2D of subjects who are born SGA also results from the combination of both genetic factors and an unfavourable fetal environment. Polymorphisms of PPAR-γ 2, a gene involved in the development and in the metabolic function of adipose tissue, have been shown to modulate the susceptibility of subjects who are born SGA to develop insulin resistance later in life(Reference Kubaszek, Markkanen and Eriksson37, Reference Eriksson, Lindi and Uusitupa38). The polymorphism is only associated with a higher risk of T2D if birth weight is reduced(Reference Kubaszek, Markkanen and Eriksson37, Reference Eriksson, Lindi and Uusitupa38). There is obviously a close relationship between genes and the environment. Not only can maternal gene expression alter the fetal environment, but the maternal intrauterine environment also affects fetal gene expression and both influence birth weight.

Association between obesity in pregnancy and later obesity in the offspring

Obesity is one of the most pervasive and burdensome public health problems in modern times. The steady increase in overweight reproductive-age women is correlated with increases in rates of childhood and infant obesity. A possible link between the abnormal intrauterine environment and abnormal growth and development of offspring must be considered. Maternal obesity significantly increases fetal and neonatal adiposity in the human subjects; thus, enhanced adipocyte development per se must play an important role in the genesis of obesity in the offspring(Reference Catalano39).

A number of epidemiological studies have shown that there is a direct relationship between birth weight and BMI in childhood and in adult life(Reference Sorensen, Sabroe and Rothman40, Reference Parsons, Power and Manor41). In the US 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 9–14 years(Reference Gillman, Rifas-Siman and Berkey42). When adjusted for maternal BMI, the increase in risk remained significantly elevated at 30%. A study of Danish military conscripts showed that even after controlling for birth length and maternal factors, BMI at ages 18–26 strongly correlated with birth weight(Reference Sorensen, Sabroe and Rothman40).

What animal models can tell us

Animal models have a normal genetic background upon which environmental effects during gestation or early post-natal life can be tested for their role in inducing disease later in life. Ontogeny of β-cell development in the rodent approximates what has been observed in human subjects(Reference Rahier, Wallon and Henquin43, Reference Von Dorsche, Reiher and Hahn44). The most commonly used animal models for IUGR are energetic or protein restriction, glucocorticoid administration, or induction of uteroplacental insufficiency in the pregnant rodent. In rats, maternal dietary protein restriction (approximately 40–50% of normal intake, termed LP) throughout gestation and lactation has been reported to alter insulin secretory capacity and reduce β-cell mass through a reduction in the β-cell proliferation rate and an increase in apoptosis(Reference Dahri, Reusens and Remacle45Reference Reusens, Remacle and Barker53). Expression of pancreatic duodenal homeobox-1 (Pdx1), a homeodomain-containing transcription factor that regulates early development of both endocrine and exocrine pancreas, and later differentiation and function of β-cells(Reference Arantes, Teixeira and Reis54), is also reduced in islets from pups of LP mothers(Reference Melloul55). In adulthood, rats born from LP mothers still have reductions in β-cell mass and insulin secretion and show glucose intolerance, but usually not overt diabetes(Reference Dahri, Reusens and Remacle45, Reference Dahri, Snoeck and Reusens-Billen46, Reference Reusens, Remacle and Barker53). In old age, LP offspring develop fasting hyperglycemia associated with insulin resistance(Reference Petry, Dorling and Pawlak56Reference Fernandez-Twinn, Wayman and Ekizoglou60).

Total energetic restriction during the last week of pregnancy and throughout lactation also reduces β-cell mass and impairs insulin secretion in the offspring(Reference Garofano, Czernichow and Bréant61, Reference Garofano, Czernichow and Bréant62). When maternal undernutrition is prolonged until weaning and normal nutrition is given to the offspring from weaning onwards, growth retardation and β-cell mass reduction persists in adulthood(Reference Garofano, Czernichow and Bréant62).

Treatment of pregnant rats with dexamethasone during the last week of gestation retards fetal growth(Reference Shen, Seckl and Slack63). Insulin content of fetal β-cells is reduced and is associated with a reduction in Pdx1(Reference Shen, Seckl and Slack63).

An ovine model of IUGR induced by placental insufficiency (heat induced) results in a significant reduction in β-cell mass in fetuses near term (0·9 of gestation) from decreased rates of β-cell proliferation and neogenesis(Reference Limesand, Jensen and Hutton64). Plasma insulin concentrations in the IUGR fetuses are lower at baseline and glucose-stimulated insulin secretion is impaired. Similar deficits occur with arginine-stimulated insulin secretion. A deficiency in islet glucose metabolism also occurs in the rate of islet glucose oxidation at maximal stimulatory glucose concentrations. Thus, pancreatic islets from nutritionally deprived IUGR fetuses caused by chronic placental insufficiency have impaired insulin secretion caused by reduced glucose-stimulated glucose oxidation rates, insulin biosynthesis and insulin content. This impaired glucose-stimulated insulin secretion occurs despite an increased fractional rate of insulin release from a greater proportion of releasable insulin as a result of diminished insulin stores(Reference Limesand, Rozance and Zerbe65).

To extend these experimental studies of growth retardation, we developed a model of IUGR in the rat that restricts fetal growth(Reference Ogata, Bussey and Finley66Reference Boloker, Gertz and Simmons68). Growth-retarded fetal rats have critical features of a metabolic profile characteristic of growth-retarded human fetuses: decreased levels of glucose, insulin, insulin-like growth factor-I, amino acids and oxygen(Reference Ogata, Bussey and Finley66Reference Unterman, Lascon and Gotway70). Birth weights of IUGR animals are significantly lower than those of controls until approximately 7 weeks of age, when IUGR rats catch up to controls. Between 7 and 10 weeks of age, the growth of IUGR rats accelerates and surpasses that of controls, and by 26 weeks of age, IUGR rats are obese(Reference Simmons, Templeton and Gertz67). No significant differences are observed in blood glucose and plasma insulin levels at 1 week of age. Between 7 and 10 weeks of age, however, IUGR rats develop mild fasting hyperglycemia and hyperinsulinemia. IUGR animals are glucose intolerant and insulin resistant at an early age. First-phase insulin secretion in response to glucose is also impaired early in life in IUGR rats, before the onset of hyperglycemia. There are no significant differences in β-cell mass, islet size or pancreatic weight between IUGR and control animals at 1 and 7 weeks of age. In 15-week-old IUGR rats, however, the relative β-cell mass is 50% that of controls, and by 26 weeks of age, β-cell mass is less than one-third that of controls. This loss of β-cell mass is accompanied by a reduction in Pdx1 expression that is greater than that in β-cell mass(Reference Stoffers, Desai and DeLeon71). By 6 months of age, IUGR rats develop diabetes with a phenotype remarkably similar to that observed in the human subject with T2D: progressive dysfunction in insulin secretion and insulin action(Reference Simmons, Templeton and Gertz67). Thus, despite different animal models of IUGR, these studies support the hypothesis that an abnormal intrauterine milieu can induce permanent changes in β-cell function after birth and lead to T2D in adulthood.

Animal model of obesity in pregnancy induces obesity in the offspring

Several investigators have used animal models of high-fat or western style-diet-induced obesity (a diet that has increased fat and carbohydrate content) and have shown that maternal over-nutrition induces increased adiposity and induces permanent changes in metabolism in the offspring(Reference Levin and Govek72Reference Han, Xu and Epstein84). Using a similar Western diet, female Sprague–Dawley rats were started on the designated diet at 4 weeks of age. The rats were mated at 12–14 weeks of age, and all pups were weaned onto a control diet(Reference Sen and Simmons85). Offspring from dams fed the Western diet had significantly increased adiposity as early as 2 weeks as well as impaired glucose tolerance compared to offspring of dams fed a control diet. Inflammation and oxidative stress were increased in pre-implantation embryos, fetuses and newborns of obese dams. Oxidative stress was correlated with increased expression of pro-adipogenic and lipogenic genes in fat tissue and in pre-implantation embryos. These results suggest that obesity is programmed as early as the pre-implantation stage of development.

Chromatin structure, DNA methylation and gene expression

Epigenetic modifications of the genome provide a mechanism that allows the stable propagation of gene expression from one generation of cells to the next. Epigenetic states can be modified by environmental factors, which may contribute to the development of abnormal phenotypes. There are at least two distinct mechanisms through which epigenetic information can be inherited: histone modifications and DNA methylation(Reference Berger86, Reference Reik87).

In eukaryotes, the nucleosome is formed when DNA is wrapped around an octameric complex of two molecules of each of the four histones: H2A, H2B, H3 and H4. The N-termini of histones can be modified by acetylation, methylation, sumoylation, phosphorylation, glycosylation and ADP ribosylation. The most common histone modifications involve acetylation and methylation of lysine residues in the N-termini of H3 and 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(Reference Berger86, Reference Reik87). Moreover, lysine residues can be mono-, di-, or trimethylated in vivo, providing an additional mechanism of regulation(Reference Berger86, Reference Reik87).

The second class of epigenetic regulation is DNA methylation, in which a cytosine base is modified by a DNA methyltransferase at the C5 position of cytosine, a reaction that is carried out by various members of a single family of enzymes(Reference Reik87). Approximately 70% of CpG dinucleotides in human DNA are constitutively methylated, whereas most of the unmethylated CpG are located in CpG islands. CpG islands are CG-rich sequences located near coding sequences, and serve as promoters for their associated genes. Approximately half of mammalian genes have CpG islands(Reference Reik87). The methylation status of CpG islands within promoter sequences works as an essential regulatory element by modifying the binding affinity of transcription factors to DNA-binding sites. In normal cells, most CpG islands remain unmethylated; however, under circumstances such as cancer(Reference Yoshida, Shigematsu and Shivapurkar88Reference Takahashi, Shigematsu and Shivapurkar90) and oxidative stress, they can become methylated de novo. This aberrant methylation is accompanied by local changes in histone modification and chromatin structure, such that the CpG island and its embedded promoter take on a repressed conformation that is incompatible with gene transcription. It is not known why particular CpG islands are susceptible to aberrant methylation.

DNA methylation is commonly associated with gene silencing and contributes to X-chromosomal inactivation, genomic imprinting as well as transcriptional regulation of tissue-specific genes during cellular differentiation (reviewed in(Reference Cedar and Bergman91Reference Gopalakrishnan, Van Emburgh and Robertson93)). It is not known why some genes are able to undergo aberrant DNA methylation; however, a study by Feltus et al.(Reference Feltus, Lee and Costello94) suggests that there is a ‘DNA sequence signature associated with aberrant methylation’. Of major significance to T2D is their finding that Pdx1, a pancreatic homeobox transcription factor, was one of only fifteen genes (of 1749 examined) with CpG islands within the promoter that were methylation-susceptible (which was induced by over-expression of a DNA methyltransferase). This study demonstrates that genes essential to pancreatic development, like Pdx1, are susceptible to epigenetic modifications, which could ultimately affect gene expression.

Histone methylation can influence DNA methylation patterns and vice versa(Reference Cedar and Bergman91). For example, methylation of lysine 9 on histone 3 (H3) promotes DNA methylation, while CpG methylation stimulates methylation of lysine 9 on H3(Reference Schubeler, Lorincz and Cimbora92). Recent evidence indicates that this dual relationship between histone methylation and DNA methylation might be accomplished by direct interactions between histone and DNA methyltransferases(Reference Cedar and Bergman91). Thus, chromatin modifications induced by adverse stimuli are self-reinforcing and can propagate.

Maternal nutritional supplementation and epigenetic modifications in the offspring

The role of environmental regulation of epigenetic phenomena in the offspring has been established by experiments performed in agouti mice (reviewed in(Reference Martin, Cropley and Suter95)). Wild-type expression of the agouti protein results in a phenotypic brown coat colour in the mouse. In this mouse model, an endogenous retrovirus-like transposon sequence is inserted close to the gene coding for the agouti protein. An unmethylated retrotransposon promoter overrides the wild-type agouti promoter, resulting in ectopic agouti expression and a yellow coat colour. A methylated retrotransposon is silenced and results in a wild-type agouti (brown) coat. Wolff et al. have investigated whether maternal diet can alter the phenotype of the agouti mouse(Reference Cooney, Dave and Wolff96) and found that when pregnant females are fed a diet supplemented with methyl donors, a larger proportion of offspring have a wild-type agouti coat colour as compared to the offspring of mothers fed a standard diet. These studies indicate that the maternal methyl donor diet leads to increased methylation of the offspring's retrotransposon. Methylation silences the offspring's retrotransposon allowing the wild-type agouti promoter to be expressed, thus resulting in a mouse with a wild-type (brown) coat colour. These results suggest that a maternal nutritional environmental exposure can change the stable expression of genes in the offspring through an epigenetic modification that takes place in utero.

Epigenetic regulation of gene expression in fetal growth retardation

A number of studies suggest that uteroplacental insufficiency, a common cause of IUGR, induces epigenetic modifications in offspring(Reference MacLennan, James and Melnyk97Reference Raychaudhuri, Raychaudhuri and Thamotharan100). Epigenetic modifications affecting processes important to glucose regulation and insulin secretion, characteristics essential to the pathophysiology of T2D have been described in the IUGR liver, pancreatic β-cells and muscle(Reference MacLennan, James and Melnyk97Reference Raychaudhuri, Raychaudhuri and Thamotharan100).

Chromatin remodelling in the β-cell of intrauterine growth retarded rats

Pdx1 is a homeodomain-containing transcription factor that plays a critical role in the early development of both the endocrine and exocrine pancreas, and in the later differentiation and function of the β-cell. As early as 24 h after the onset of growth retardation, Pdx1 mRNA levels are reduced by more than 50% in IUGR fetal rats. Suppression of Pdx1 expression persists after birth and progressively declines in the IUGR animal, implicating an epigenetic mechanism.

Changes in histone acetylation are the first epigenetic modification found in β-cells of IUGR animals. Islets isolated from IUGR fetuses show a significant decrease in H3 and H4 acetylation at the proximal promoter of Pdx1 (Reference Park, Stoffers and Nicholls99). These changes in H3 and H4 acetylation are associated with a loss of binding of upstream stimulatory factor-1 to the proximal promoter of Pdx1 (Reference Park, Stoffers and Nicholls99). Upstream stimulatory factor-1 is a critical activator of Pdx1 transcription, and its decreased binding markedly decreases Pdx1 transcription(Reference Qian, Kaytor and Towle101, Reference Sharma, Leonard and Lee102). After birth, histone deacetylation progresses and is followed by a marked decrease in H3K4 trimethylation and a significant increase in dimethylation of H3K9 in IUGR islets(Reference Park, Stoffers and Nicholls99). H3K4 trimethylation is usually associated with active gene transcription, whereas H3K9 dimethylation is usually a repressive chromatin mark. Progression of these histone modifications parallels the progressive decrease in Pdx1 expression that manifests as a deterioration in glucose homeostasis and increased oxidative stress in the aging IUGR animals(Reference Park, Stoffers and Nicholls99). Nevertheless, at 2 weeks of age, the silencing histone modifications in the IUGR pup are responsible for the suppression of Pdx1 expression since there is no appreciable methylation of CpG islands in mice at this age(Reference Park, Stoffers and Nicholls99). Reversal of histone deacetylation in IUGR islets at 2 weeks of age, is sufficient to nearly normalize Pdx1 mRNA levels permanently, perhaps due to active β-cell replication present in the neonatal rodent(Reference Park, Stoffers and Nicholls99).

In IUGR, Pdx1 is first silenced due to the recruitment of co-repressors, including histone deacetylase 1 and mSin3A (mammalian Sin 3)(Reference Park, Stoffers and Nicholls99). These repressors catalyse histone deacetylation. Binding of these deacetylases facilitates loss of trimethylation of H3K4, further repressing Pdx1 expression(Reference Park, Stoffers and Nicholls99). We found that inhibition of histone deacetylase activity by trichostatin A treatment normalises H3K4me3 levels at Pdx1 in IUGR islets(Reference Park, Stoffers and Nicholls99). These data suggest that the association of histone deacetylase 1 at Pdx1 in IUGR islets likely serves as a platform for the recruitment of a demethylase, which catalyses demethylation of H3K4.

The molecular mechanism responsible for DNA methylation in IUGR islets is likely dependent on the methylation status of lysine 9 on H3 (H3K9). Previous studies have shown that changes in methylation of H3K9 precede changes in DNA methylation(Reference Li, Rauch and Chen103, Reference Bachman, Park and Rhee104). It has also been suggested that DNA methyltransferases may act only on chromatin that is methylated at H3K9(Reference Kouzarides105). Histone methyltransferases specifically DNA methyltransferase 3A and DNA methyltransferase 3B, bind to DNA methylases, thereby initiating DNA methylation(Reference Kouzarides105).

These results demonstrate that IUGR induces a self-propagating epigenetic cycle in which the mSin3A–histone deacetylase complex is first recruited to the Pdx1 promoter, histone tails are subjected to deacetylation and Pdx1 transcription is repressed. At the neonatal stage, this epigenetic process is reversible and may define an important developmental window for therapeutic approaches. However, as dimethylated H3K9 accumulates, DNA methyltransferase 3A is recruited to the promoter and initiates de novo DNA methylation, which locks in the silenced state in the IUGR adult pancreas resulting in diabetes.

How do these epigenetic events lead to diabetes? Targeted homozygous disruption of Pdx1 in mice results in pancreatic agenesis, and homozygous mutations yield a similar phenotype in human subjects(Reference Bernardo, Hay and Docherty106). Milder reductions in Pdx1 protein levels, as occurs in the Pdx±mice, allow for the development of a normal mass of β-cells(Reference Bernardo, Hay and Docherty106), but result in the impairment of several events in glucose-stimulated insulin secretion(Reference Bernardo, Hay and Docherty106). These results indicate that Pdx1 plays a critical role in the normal function of β-cells(Reference Bernardo, Hay and Docherty106) in addition to its role in β-cell lineage development. This may be the reason that human subjects with heterozygous missense mutations in Pdx1 exhibit early- and late-onset forms of T2D(Reference Bernardo, Hay and Docherty106).

The discovery of a critical developmental stage during which aberrant epigenetic modifications may be reversed represents a therapeutic window for the use of novel agents that could prevent common diseases with late-onset phenotypes. T2D is one such disease, where predisposed individuals could be treated with agents that normalize the epigenetic programming of key genes, thus providing protection against development of the adult diabetic phenotype.

Genome-wide DNA methylation is disrupted in intrauterine growth-retarded islets

Epigenetic modifications are not confined to the Pdx1 locus in the IUGR rat. We mapped DNA methylation across approximately 1 000 000 loci using the HELP assay(Reference Thompson, Fazzari and Niu107). Comparison of IUGR with normal rats at 7 weeks of age prior to the onset of diabetes, revealed changes in DNA methylation at a number of novel loci, not limited to canonical CpG islands or promoters. We found that IUGR in the rat causes consistent and non-random changes in cytosine methylation, affecting <1% of HpaII sites in the genome in the islet. The majority of these changes take place not at promoters but at intergenic sequences, many of which are evolutionarily conserved. Furthermore, some of these loci are in proximity to genes manifesting concordant changes in gene expression and are enriched near genes that regulate processes that are markedly impaired in IUGR islets (e.g. vascularization, proliferation, insulin secretion and cell death).

Summary

The studies described above clearly show that environmental effects can induce epigenetic alterations, ultimately effecting expression of key genes linked to the development of T2D including genes critical for pancreatic development and β-cell function, peripheral glucose uptake and insulin resistance and atherosclerosis. Recent progress in understanding the epigenetic programming of gene function has led to the development of novel therapeutic agents with epigenetic targets in diseases such as cancer. Understanding the role of developmental programming of genes crucial to the development of T2D may unveil a critical window during which epigenetic therapeutic agents could be used as a means to prevent the later development of a disease. Prior to the use of such therapeutic agents there remains much to be learned about the programming of the epigenetic code, especially on a genome-wide scale. Much of the recent progress in understanding epigenetic phenomena is directly attributable to technologies that allow researchers to pinpoint the genomic location of proteins that package and regulate access to the DNA. The advent of DNA microarrays and inexpensive DNA sequencing has allowed many of those technologies to be applied to the whole genome. It is now possible that epigenetic profiling of CpG islands in the human genome can be used as a tool to identify genomic loci that are susceptible to DNA methylation. Aberrant methylation may then be used as a biomarker for disease. The genome-wide mapping of histone modifications by ChIP (chromatin immunoprecipitation)-chip and ChIP-seq has led to important insights regarding the mechanism of transcriptional and epigenetic memory, and how different chromatin states are propagated through the genome in yeast and in mammalian cells(Reference Lieb, Beck and Bulyk108, Reference Kim, Barrera and Zheng109). Although CHIP-seq experiments are currently being performed in human tissue, obstacles such as intrinsic human epigenetic variability (including age-related changes), and tissue-specific epigenetic variability must be characterized and mapped in the healthy, non-diseased state before this information can be applied to diseases such as T2D. Eventually genome-wide epigenetic characterization will lead to specific therapies with epigenetic targets and also will allow monitoring of genome-wide epigenetic consequences of these therapies once they are applied.

Acknowledgements

The author declares no conflict of interest. This work was supported by NIH grants: DK55704 and DK078761.

References

1.Hales, CN & Barker, DJP (1992) Type 2 diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia 35, 595601.CrossRefGoogle ScholarPubMed
2.Kermack, WO (1934) Death rates in Great Britain and Sweden. Lancet 1, 698703.CrossRefGoogle Scholar
3.Ravelli, GP, Stein, ZA & Susser, MW (1976) Obesity in young men after famine exposure in utero and early infancy. N Engl J Med 295, 349353.CrossRefGoogle ScholarPubMed
4.Barker, DJP (2004) The developmental origins of adult disease. J Am Coll Nutr 23, 588S595S.CrossRefGoogle ScholarPubMed
5.Valdez, R, Athens, MA, Thompson, GH et al. (1994) Birthweight and adult health outcomes in a biethnic population in the USA. Diabetologia 37, 624631.CrossRefGoogle Scholar
6.Jaquet, D, Gaboriau, A, Czernichow, P et al. (2000) Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J Clin Endocrinol Metab 85, 14011406.Google ScholarPubMed
7.Egeland, GM, Skjaerven, R & Irgrens, LM (2000) Birth characteristics of women who develop gestational diabetes: population based study. BMJ 321, 546547.CrossRefGoogle ScholarPubMed
8.Forsen, T, Eriksson, J, Tuomilehto, J et al. (2000) The fetal and childhood growth of persons who develop type 2 diabetes. Ann Intern Med 133, 176182.CrossRefGoogle ScholarPubMed
9.Rich-Edwards, JW, Colditz, GA, Stampfer, MJ et al. (1999) Birthweight and the risk for type 2 diabetes mellitus in adult women. Ann Intern Med 130, 278284.CrossRefGoogle ScholarPubMed
10.Eriksson, J, Forsen, T, Tuomilehto, J et al. (2000) Fetal and childhood growth and hypertension in adult life. Hypertension 36, 790794.CrossRefGoogle ScholarPubMed
11.Bavdekar, A, Sachdev, HS, Fall, CHD et al. (2004) Relation of serial changes in childhood body-mass index to impaired glucose tolerance in young adulthood. N Engl J Med 350, 865875.Google Scholar
12.Whincup, PH, Cook, DG, Adshead, T et al. (1997) Childhood size is more strongly related than size at birth to glucose and insulin levels in 10–11 year-old children. Diabetologia 40, 319326.CrossRefGoogle ScholarPubMed
13.Martin, RM, McCarthy, A, Smith, GD et al. (2003) Infant nutrition and blood pressure in early adulthood: the Barry Caerphilly Growth study. Am J Clin Nutr 77, 14891497.CrossRefGoogle ScholarPubMed
14.Law, CM, Shiell, AW, Newsome, CA et al. (2002) Fetal, infant, and childhood growth and adult blood pressure: a longitudinal study from birth to 22 years of age. Circulation 105, 10881092.CrossRefGoogle ScholarPubMed
15.Li, C, Johnson, MS & Goran, MI (2001) Effects of low birth weight on insulin resistance syndrome in Caucasion and African–American children. Diabetes Care 24, 20352042.CrossRefGoogle Scholar
16.Boney, CM, Verma, A, Tucker, R et al. (2005) Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics 115, 290296.CrossRefGoogle ScholarPubMed
17.Clausen, JO, Borch-Johnsen, K & Pedersen, O (1997) Relation between birth weight and the insulin sensitivity index in a population sample of 331 young, healthy Caucasians. Am J Epidemiol 146, 2331.CrossRefGoogle Scholar
18.Flanagan, DE, Moore, VM, Godsland, IF et al. (2000) Fetal growth and the physiological control of glucose tolerance in adults: a minimal model analysis. Am J Physiol Endocrinol Metab 278, E700E706.CrossRefGoogle ScholarPubMed
19.Van Assche, FA, De Prins, F, Aerts, L et al. (1977) The endocrine pancreas in small-for dates infants. Br J Obstet Gynaecol 84, 751753.CrossRefGoogle ScholarPubMed
20.Beringue, F, Blondeau, B, Castellotti, MC et al. (2005) Endocrine pancreas development in growth-retarded human fetuses. Diabetes 51, 385391.CrossRefGoogle Scholar
21.Econimides, DL, Proudler, A & Nicolaides, KH (1989) Plasma insulin in appropriate and small for gestational age fetuses. Am J Obstet Gynecol 160, 10911094.CrossRefGoogle Scholar
22.Setia, S, Sridhar, MG, Bhat, V et al. (2006) Insulin sensitivity and insulin secretion at birth in intrauterine growth retarded infants. Pathology 38, 236238.CrossRefGoogle ScholarPubMed
23.Bazaes, RA, Salazar, TE & Pittaluga, E (2003) Glucose and lipid metabolism in small for gestational age infants at 48 hours of age. Pediatrics 111, 804809.CrossRefGoogle ScholarPubMed
24.Jensen, CB, Storgaard, H, Dela, F et al. (2002) Early differential defects of insulin secretion and action in 19-year-old Caucasian men who had low birth weight. Diabetes 51, 12711280.CrossRefGoogle ScholarPubMed
25.Barnett, AH, Eff, C, Leslie, RDG et al. (1981) Diabetes in identical twins. Diabetologia 20, 8793.CrossRefGoogle ScholarPubMed
26.Newman, B, Selby, JV, King, MC et al. (1987) Concordance for type 2 diabetes mellitus in male twins. Diabetologia 30, 763768.CrossRefGoogle ScholarPubMed
27.Warram, JH, Martin, BC, Krolewski, AS et al. (1990) Slow glucose removal rate and hyperinsulinemia precede the development of type II diabetes in the offspring of diabetic parents. Ann Intern Med 113, 909915.CrossRefGoogle ScholarPubMed
28.Vaag, A, Henricksen, JE, Madsbad, S et al. (1995) Insulin secretion, insulin action, and hepatic glucose production in identical twins discordant for NIDDM. J Clin Invest 95, 690698.CrossRefGoogle Scholar
29.Hattersley, AT & Tooke, JE (1999) The fetal insulin hypothesis: an alternative explanation of the association of low birthweight with diabetes and vascular disease. Lancet 353, 17891792.CrossRefGoogle ScholarPubMed
30.Froguel, P, Zouali, H & Vionnet, N (1993) Familial hyperglycemia due to mutations in glucokinase: definition of a subtype of diabetes mellitus. New Engl J Med 328, 697702.CrossRefGoogle ScholarPubMed
31.Hattersley, AT, Beards, F, Ballantyne, E et al. (1998) Mutations in the glucokinase gene of the fetus result in reduced birth weight. Nat Genet 19, 268270.CrossRefGoogle ScholarPubMed
32.Grant, SF, Thorleifsson, G, Reynisdottir, I et al. (2006) Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38, 320323.CrossRefGoogle ScholarPubMed
33.Damcott, CM, Pollin, TI, Reinhart, LJ et al. (2006) Polymorphisms in the transcription factor 7-like 2 (TCF7L2) gene are associated with type 2 diabetes in the Amish: replication and evidence for a role in both insulin secretion and insulin resistance. Diabetes 55, 26542659.CrossRefGoogle ScholarPubMed
34.Saxena, R, Gianniny, L, Burtt, NP et al. (2006) Common single nucleotide polymorphisms in TCF7L2 are reproducibly associated with type 2 diabetes and reduce the insulin response to glucose in nondiabetic individuals. Diabetes 55, 28902895.CrossRefGoogle ScholarPubMed
35.Munoz, J, Lok, KH, Gower, BA et al. (2006) Polymorphism in the transcription factor 7-like 2 (TCF7L2) gene is associated with reduced insulin secretion in nondiabetic women. Diabetes 55, 36303634.CrossRefGoogle ScholarPubMed
36.Freathy, RM, Weedon, MN, Bennett, A et al. (2007) Type 2 diabetes TCF7L2 risk genotypes alter birth weight: a study of 24,053 individuals. Am J Hum Genet 80, 11501161.CrossRefGoogle Scholar
37.Kubaszek, A, Markkanen, A, Eriksson, JG et al. (2004) The association of the K121Q polymorphism of the plasma cell glycoprotein-1 gene with type 2 diabetes and hypertension depends on size at birth. J Clin Endocrinol Metab 89, 20442047.CrossRefGoogle ScholarPubMed
38.Eriksson, JG, Lindi, V, Uusitupa, M et al. (2002) The effects of the Pro12Ala polymorphism of the peroxisome proliferator-activated receptor-gamma2 gene on insulin sensitivity and insulin metabolism interact with size at birth. Diabetes 517, 23212324.CrossRefGoogle Scholar
39.Catalano, PM (2003) Obesity and pregnancy-The propagation of a viscous cycle? J Clin Endocrinol Metab 88, 35003506.CrossRefGoogle ScholarPubMed
40.Sorensen, HT, Sabroe, S, Rothman, KJ et al. (1997) Relation between weight and length at birth and body mass index in young adulthood: cohort study. Bone Miner J 315, 1137.Google ScholarPubMed
41.Parsons, TJ, Power, C & Manor, O (2001) Fetal and early life growth and body mass index from birth to early adulthood in 1958 British cohort: longitudinal study. BMJ 323, 13311335.CrossRefGoogle ScholarPubMed
42.Gillman, MW, Rifas-Siman, SL, Berkey, CS et al. (2003) Maternal gestational diabetes, and adolescent obesity. Pediatrics 111, E221E226.CrossRefGoogle ScholarPubMed
43.Rahier, J, Wallon, J & Henquin, J-C (1981) Cell populations in the endocrine pancreas of human neonates and infants. Diabetologia 20, 540546.CrossRefGoogle ScholarPubMed
44.Von Dorsche, H, Reiher, H & Hahn, H-J (1988) Phases in the early development of the human islet organ. Anat Anz 166, 6976.Google Scholar
45.Dahri, S, Reusens, B, Remacle, C et al. (1995) Nutritional influences on pancreatic development and potential links with non-insulin-dependent diabetes. Proc Nutr Soc 54, 345356.CrossRefGoogle ScholarPubMed
46.Dahri, S, Snoeck, A, Reusens-Billen, B et al. (1991) Islet function in off-spring of mothers on low-protein diet during gestation. Diabetes 40, 115120.CrossRefGoogle Scholar
47.Snoeck, A, Remacle, C, Reusens, B, et al. (1990) Effect of a low protein diet during pregnancy on the fetal rat endocrine pancreas. Biol Neonate 57, 107118.CrossRefGoogle ScholarPubMed
48.Berney, DM, Desai, M, Palmer, DJ et al. (1997) The effects of maternal protein deprivation on the fetal rat pancreas: major structural changes and their recuperation. J Pathol 183, 109115.3.0.CO;2-B>CrossRefGoogle ScholarPubMed
49.Wilson, MR & Hughes, SJ (1997) The effect of maternal protein deficiency during pregnancy and lactation on glucose tolerance and pancreatic islet function in adult rat offspring. J Endocrinol 154, 177185.CrossRefGoogle ScholarPubMed
50.Bertin, E, Gangnerau, MN, Bellon, G et al. (2002) Development of beta-cell mass in fetuses of rats deprived of protein and/or energy in last trimester of pregnancy. Am J Physiol Regul Integr Comp Physiol 283, R623R630.CrossRefGoogle ScholarPubMed
51.Boujendar, S, Reusens, B, Merezak, S et al. (2002) Taurine supplementation to a low protein diet during foetal and early postnatal life restores a normal proliferation and apoptosis of rat pancreatic islets. Diabetologia 45, 856866.CrossRefGoogle ScholarPubMed
52.Petrik, J, Reusens, B, Arany, E et al. (1999) A low protein diet alters the balance of islet cell replication and apoptosis in the fetal and neonatal rat, and is associated with a reduced pancreatic expression of insulin-like growth factor-II. Endocrinology 140, 48614873.CrossRefGoogle ScholarPubMed
53.Reusens, B & Remacle, C (2000) Effects of maternal nutrition and metabolism on the developing endocrine pancreas. In Fetal origins of cardiovascular and lung disease, pp. 339358 [Barker, DJP, editor]. New York: Marcel Dekker Inc.Google Scholar
54.Arantes, VC, Teixeira, VPA, Reis, MAB et al. (2002) Expression of PDX-1 is reduced in pancreatic islets from pups of rat dams fed a low protein diet during gestation and lactation. J Nutr 132, 30303035.CrossRefGoogle ScholarPubMed
55.Melloul, D (2004) Transcription factors in islet development and physiology: role of PDX-1 in beta-cell function. Ann N Y Acad Sci 1014, 2837.CrossRefGoogle ScholarPubMed
56.Petry, CJ, Dorling, MW, Pawlak, DB et al. (2001) Diabetes in old male offspring of rat dams fed a reduced protein diet. Int J Exp Diabetes Res 2, 139143.CrossRefGoogle ScholarPubMed
57.Ozanne, SE, Wang, CL, Coleman, N et al. (1996) Altered muscle insulin sensitivity in the male offspring of protein-malnourished rats. Am J Physiol 271, E1128E1134.Google ScholarPubMed
58.Ozanne, SE, Jensen, CB, Tingey, KJ et al. (2005) Low birthweight is associated with specific changes in muscle insulin-signaling protein expression. Diabetologia 48, 547552.CrossRefGoogle Scholar
59.Ozanne, SE, Olsen, GS, Hansen, LL et al. (2003) Early growth restriction leads to down regulation of protein kinase C zeta and insulin resistance in skeletal muscle. J Endocrinol 177, 235241.CrossRefGoogle ScholarPubMed
60.Fernandez-Twinn, DS, Wayman, A, Ekizoglou, S et al. (2005) Maternal protein restriction leads to hyperinsulinemia and reduced insulin-signaling protein expression in 21-mo-old female rat offspring. Am J Physiol Regul Integr Comp Physiol 288, R368R373.CrossRefGoogle ScholarPubMed
61.Garofano, A, Czernichow, P & Bréant, B (1997) In utero undernutrition impairs rat beta-cell development. Diabetologia 40, 12311234.CrossRefGoogle ScholarPubMed
62.Garofano, A, Czernichow, P & Bréant, B (1999) Effect of ageing on beta-cell mass and function in rats malnourished during the perinatal period. Diabetologia 42, 711718.CrossRefGoogle ScholarPubMed
63.Shen, CN, Seckl, JR, Slack, JM et al. (2003) Glucocorticoids suppress beta-cell development and induce hepatic metaplasia in embryonic pancreas. Biochem J 375 (Pt 1), 4150.CrossRefGoogle ScholarPubMed
64.Limesand, SW, Jensen, J, Hutton, JC et al. (2005) Diminished β-cell replication contributes to reduced β-cell mass in fetal sheep with intrauterine growth restriction. Am J Physiol Regul Integr Comp Physiol 288, R1297R1305.CrossRefGoogle ScholarPubMed
65.Limesand, SW, Rozance, PJ, Zerbe, GO et al. (2006) Attenuated insulin release and storage in fetal sheep pancreatic islets with intrauterine growth restriction. Endocrinology 147, 14881497.CrossRefGoogle ScholarPubMed
66.Ogata, ES, Bussey, M & Finley, S (1986) Altered gas exchange, limited glucose, branched chain amino acids, and hypoinsulinism retard fetal growth in the rat. Metabolism 35, 950977.CrossRefGoogle ScholarPubMed
67.Simmons, RA, Templeton, L, Gertz, S et al. (2001) Intrauterine growth retardation leads to type II diabetes in adulthood in the rat. Diabetes 50, 22792286.CrossRefGoogle ScholarPubMed
68.Boloker, J, Gertz, S & Simmons, RA (2002) Offspring of diabetic rats develop obesity and type II diabetes in adulthood. Diabetes 51, 14991506.CrossRefGoogle Scholar
69.Simmons, RA, Gounis, AS, Bangalore, SA et al. (1991) Intrauterine growth retardation: fetal glucose transport is diminished in lung but spared in brain. Pediatr Res 31, 5963.CrossRefGoogle Scholar
70.Unterman, T, Lascon, R, Gotway, M et al. (1990) Circulating levels of insulin-like growth factor binding protein-1 (IGFBP-1) and hepatic mRNA are increased in the small for gestational age fetal rat. Endocrinology 127, 20352037.CrossRefGoogle Scholar
71.Stoffers, DA, Desai, BM, DeLeon, DD et al. (2003) Neonatal exendin-4 prevents the development of diabetes in the intrauterine growth retarded rat. Diabetes 52, 734740.CrossRefGoogle ScholarPubMed
72.Levin, BE & Govek, E (1998) Gestational obesity accentuates obesity in obesity-prone progeny. Am J Physiol Regul Integr Comp Physiol 44, R1374R1379.CrossRefGoogle Scholar
73.Howie, GJ, Sloboda, DM, Kamal, T et al. (2009) Maternal nutritional history predicts obesity in adult offspring independent of postnatal diet. J Physiol 587, 905915.CrossRefGoogle ScholarPubMed
74.Bayol, SA, Simbi, BH & Stickland, NC (2005) A maternal cafeteria diet during gestation and lactation promotes adiposity and impairs skeletal muscle development and metabolism in rat offspring at weaning. J Physiol 567, 951961.CrossRefGoogle ScholarPubMed
75.Buckley, AJ, Keseru, B, Briody, J et al. (2005) Altered body composition and metabolism in the male offspring of high fat-fed rats. Metabolism 54, 500507.CrossRefGoogle ScholarPubMed
76.Srinivasan, M, Katewa, SD, Palaniyappan, A et al. (2006) Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab 291, E792E799.CrossRefGoogle Scholar
77.Muhlhausler, BS, Adam, CL, Findlay, PA et al. (2006) Increased maternal nutrition alters development of the appetite-regulating network in the brain. FASEB J 20, 12571259.CrossRefGoogle ScholarPubMed
78.Caluwaerts, S, Lambin, S, van Bree, R et al. (2007) Diet-induced obesity in gravid rats engenders early hyperadiposity in the offspring. Metabolism 56, 14311438.CrossRefGoogle ScholarPubMed
79.Shankar, K, Harrekk, A, Liu, X et al. (2008) Maternal obesity at conception programs obesity in the offspring. Am J Physiol Regul Integr Comp Physiol 294, R528R538.CrossRefGoogle ScholarPubMed
80.Samuelsson, AM, Matthews, PA, Argenton, M et al. (2008) Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension 51, 383392.CrossRefGoogle ScholarPubMed
81.Mitra, A, Albers, KM, Crump, EM et al. (2008) Effect of high-fat diet during gestation, lactation or postweaning on physiological and behavioral indexes in borderline hypertensive rats. Am J Physiol Regul Integr Comp Physiol 296, R20R28.CrossRefGoogle ScholarPubMed
82.Gniuli, D, Calcagno, A, Caristo, ME et al. (2008) Effects of high-fat diet exposure during fetal life on type 2 diabetes development in the progeny. J Lipid Res 49, 19361945.CrossRefGoogle ScholarPubMed
83.Yan, X, Zhu, MJ, Xu, W et al. (2010) Up-regulation of toll-like receptor 4/nuclear factor-kB signaling is associated with enhanced adipogenesis and insulin resistance in fetal skeletal muscle of obese sheep at late gestation. Endocrinology 151, 380387.CrossRefGoogle Scholar
84.Han, J, Xu, J, Epstein, PN et al. (2005) Long-term effect of maternal obesity on pancreatic beta cells of offspring: reduced beta cell adaptation to high glucose and high-fat diet challenges in adult female mouse offspring. Diabetologia 48, 18101818.CrossRefGoogle ScholarPubMed
85.Sen, S & Simmons, RA (2010) Maternal antioxidant supplementation prevents obesity in the offspring of obese dams. Diabetes (In the Press).CrossRefGoogle Scholar
86.Berger, SL (2007) The complex language of chromatin regulation during transcription. Nature 447, 407412.CrossRefGoogle ScholarPubMed
87.Reik, W (2007) Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425432.CrossRefGoogle ScholarPubMed
88.Yoshida, H, Shigematsu, H, Shivapurkar, N et al. (2006) Deregulation of the HOXA10 homeobox gene in endometrial carcinoma: role in epithelial-mesenchymal transition. Cancer Res 66, 889897.CrossRefGoogle ScholarPubMed
89.So, K, Tamura, G, Honda, T et al. (2006) Multiple tumor suppressor genes are increasingly methylated with age in non-neoplastic gastric epithelia. Cancer Sci 97, 11551158.CrossRefGoogle ScholarPubMed
90.Takahashi, T, Shigematsu, H, Shivapurkar, N et al. (2006) Aberrant promoter methylation of multiple genes during multistep pathogenesis of colorectal cancers. Int J Cancer 118, 924931.CrossRefGoogle ScholarPubMed
91.Cedar, H & Bergman, Y (2009) Linking DNA methylations and histone modifications: patterns and paradigms. Nat Rev Genet 10, 295304.CrossRefGoogle Scholar
92.Schubeler, D, Lorincz, MC, Cimbora, DM et al. (2000) Genomic targeting of methylated DNA: influence of methylation on transcription, replication, chromatin structure, and histone acetylation. Mol Cell Biol 20, 9103–1912.CrossRefGoogle ScholarPubMed
93.Gopalakrishnan, S, Van Emburgh, BO, Robertson, KD et al. (2008) DNA methylation in development and human disease. Mutat Res 647, 3083.CrossRefGoogle ScholarPubMed
94.Feltus, FA, Lee, EK, Costello, JF et al. (2003) Predicting aberrant CpG island methylation. Proc Natl Acad Sci USA 100, 1225312258.CrossRefGoogle ScholarPubMed
95.Martin, DI, Cropley, JE, Suter, CM et al. (2008) Environmental influence on epigenetic inheritance at the Avy allele. Nutr Rev 66, Suppl. 1, S12S14.CrossRefGoogle ScholarPubMed
96.Cooney, CA, Dave, AA, Wolff, GL et al. (2002) Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J Nutr 132, 2393S2400S.CrossRefGoogle ScholarPubMed
97.MacLennan, NK, James, SJ, Melnyk, S et al. (2004) Uteroplacental insufficiency alters DNA methylation, one-carbon metabolism, and histone acetylation in IUGR rats. Physiol Genomics 18, 4350.CrossRefGoogle ScholarPubMed
98.Fu, Q, McKnight, RA, Yu, X et al. (2004) Uteroplacental insufficiency induces site-specific changes in histone H3 covalent modifications and affects DNA-histone H3 positioning in day 0 IUGR rat liver. Physiol Genomics 20, 108116.CrossRefGoogle Scholar
99.Park, JH, Stoffers, DA, Nicholls, RD et al. (2008) Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J Clin Invest 118, 23162324.Google ScholarPubMed
100.Raychaudhuri, N, Raychaudhuri, S, Thamotharan, M et al. (2008) Histone code modifications repress glucose transporter 4 expression in the intrauterine growth-restricted offspring. J Biol Chem 283, 1361113626.CrossRefGoogle ScholarPubMed
101.Qian, J, Kaytor, EN, Towle, HC et al. (1999) Upstream stimulatory factor regulates Pdx−1 gene expression in differentiated pancreatic β-cells. Biochem J 341, 315322.CrossRefGoogle ScholarPubMed
102.Sharma, S, Leonard, J, Lee, S et al. (1996) Pancreatic islet expression of the homeobox factor STF-1 (Pdx−1) relies on an E-box motif that binds USF. J Biol Chem 271, 22942299.CrossRefGoogle Scholar
103.Li, H, Rauch, T, Chen, ZX et al. (2006) The histone methyltransferase SETDB1 and the DNA methyltransferase DNMT3A interact directly and localize to promoters silenced in cancer cells. J Biol Chem 281, 1948919500.CrossRefGoogle ScholarPubMed
104.Bachman, KE, Park, BH, Rhee, I et al. (2003) Histone modifications and silencing prior to DNA methylation of a tumor suppressor gene. Cancer Cell 3, 8995.CrossRefGoogle ScholarPubMed
105.Kouzarides, T (2002) Histone methylation in transcriptional control. Curr Opin Genet Dev 12, 198209.CrossRefGoogle ScholarPubMed
106.Bernardo, AS, Hay, CW, Docherty, K et al. (2008) Pancreatic transcription factors and their role in the birth, life and survival of the pancreatic beta cell. Mol Cell Endocrinol 294, 19.CrossRefGoogle ScholarPubMed
107.Thompson, RF, Fazzari, MJ, Niu, H et al. (2010) Experimental IUGR induces alterations in DNA methylation and gene expression in pancreatic islets of rats. J Biol Chem 285, 1511115118.CrossRefGoogle ScholarPubMed
108.Lieb, JD, Beck, S, Bulyk, ML et al. (2006) Applying whole-genome studies of epigenetic regulation to study human disease. Cytogenet Genome Res 114, 115.CrossRefGoogle ScholarPubMed
109.Kim, TH, Barrera, LO, Zheng, M et al. (2005) A high-resolution map of active promoters in the human genome. Nature 436, 876880.CrossRefGoogle ScholarPubMed