Hostname: page-component-7c8c6479df-5xszh Total loading time: 0 Render date: 2024-03-19T05:55:01.639Z Has data issue: false hasContentIssue false

Nutrition in early life, and risk of cancer and metabolic disease: alternative endings in an epigenetic tale?

Published online by Cambridge University Press:  12 December 2008

Graham C. Burdge*
Affiliation:
Institute of Human Nutrition, Institute of Developmental Sciences Building, Southampton General Hospital, MP 887, Tremona Road, SouthamptonSO16 6YD, UK
Karen A. Lillycrop
Affiliation:
Developmental and Cell Biology, Biomedical Sciences Building, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, UK
Alan A. Jackson
Affiliation:
Institute of Human Nutrition, Institute of Developmental Sciences Building, Southampton General Hospital, MP 887, Tremona Road, SouthamptonSO16 6YD, UK
*
*Corresponding author: Dr Graham C. Burdge, fax +44 23 80795225, email g.c.burdge@southampton.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

There is substantial evidence which shows that constraints in the early life environment are an important determinant of risk of metabolic disease and CVD. There is emerging evidence that higher birth weight, which reflects a more abundant prenatal environment, is associated with increased risk of cancer, in particular breast cancer and childhood leukaemia. Using specific examples from epidemiology and experimental studies, this review discusses the hypothesis that increased susceptibility to CVD, metabolic disease and cancer have a common origin in developmental changes induced in the developing fetus by aspects of the intra-uterine environment including nutrition which involve stable changes to the epigenetic regulation of specific genes. However, the induction of specific disease risk is dependent upon the nature of the environmental challenge and interactions between the susceptibility set by the altered epigenome and the environment throughout the life course.

Type
Horizons in Nutritional Science
Copyright
Copyright © The Authors 2008

Cancer is the result of derangement of cellular processes which control cell division and apoptosis(1). There is a large body of information which shows that environmental exposures, including nutrition, play a significant role in the aetiology of the disease. Such exposures usually precede the appearance of clinical disease by a prolonged period of time, often several decades(1). While gene mutation has a role in the aetiology of cancer, there is increasing evidence which shows that epigenetic processes such as DNA methylation and covalent modifications to histones are also involved(Reference Vucic, Brown and Lam2). Such epigenetic changes represent potential for altered gene activity and hence cellular dysregulation, but these may only be manifest when the gene is exposed to an appropriate environmental signal which is enhanced or diminished as a consequence of the epigenetic change compared to normal cells. This suggestion is consistent with a life-course perspective on cancer risk. The early life environment has been shown to be an important factor in determining risk for some types of cancer. Measures of growth in early life show statistical associations with risk of specific cancers. In this context, there appear to be parallels between causal processes in cancer with metabolic disease and CVD in which the early life environment and epigenetic processes lead to a susceptibility which increases the risk to later specific environmental exposures(Reference Jackson3). The purpose of this review is to discuss the hypothesis that the early life environment, in particular nutrition, induces changes in the development of the offspring which lead to increased risk of cancer, or metabolic disease and CVD. The extent to which altered epigenetic processes established during development represent a common mechanism in cancer and metabolic disease, diseases which are generally considered as resulting from widely different pathological processes, will also be discussed.

Early life environment and risk of metabolic disease

Studies in man

Epidemiological studies show that the environment experienced before birth and shortly afterwards has long-term effects on the subsequent risk of metabolic diseases including CVD, type 2 diabetes mellitus and obesity. Barker(Reference Barker4) and Godfrey & Barker(Reference Godfrey and Barker5) showed that adults who were small at birth had a higher risk of developing metabolic disease and CVD, and that increasing birth weight was associated with a graded decrease in risk. Such induction of increased risk as a result of intra-uterine constraint has been termed ‘fetal programming’. Importantly, these effects occur over the usual range of birth weights rather than being the result of pathological states such as intra-uterine growth retardation or, at the opposite end of the spectrum, macrosomy(Reference Barker4). These findings have been replicated in a substantial number of studies in different populations world wide, although there are important variations in specific outcome measures (see Gluckman & Hanson(Reference Gluckman and Hanson6) for examples). Size at birth reflects the growth of the fetus and small size reflects a developmental constraint imposed by the intra-uterine environment. Nutrition, together with hormones and oxygen supply, is an important determinant of fetal growth and has been studied extensively with respect to its role in the induction of risk of metabolic disease. Perhaps the most persuasive example of a specific role for prenatal nutrition in determining subsequence risk of metabolic disease comes from studies of adults who were in utero during the Dutch Hunger Winter which occurred during a discrete period of 1944–5(Reference Roseboom, van der Meulen and Ravelli7). These studies show that compared to adults born before or conceived after the famine, adults who were fetuses during the famine showed increased risk of hypertension, obesity and insulin resistance. Furthermore, the severity of such effects was graded according to the stage of development of the fetus at the time of famine(Reference Roseboom, van der Meulen and Ravelli7). This suggests that different organ systems are susceptible to nutritional constraint during specific periods of development.

The paradigm that environmental constraint in early life determines subsequent risk on non-communicable diseases has been extended by Gluckman & Hanson(Reference Gluckman and Hanson8). They suggest that this association is an example of a wider spectrum of developmental responses to environmental stimuli which through developmental plasticity induce adaptations to the phenotype of the fetus which predict the postnatal environment. If correct, such predictions confer a survival advantage. This hypothesis has been supported by recent observations between fatness at birth in women, and their subsequent reproductive capacity(Reference Jasienska, Thune and Ellison9). It is postulated that a mismatch between the predicted environment and that which the offspring experiences after birth results in a disadvantage that in man leads to increased risk of disease(Reference Gluckman and Hanson8). The extent and nature of the mismatch remains to be determined.

There is emerging data that the effects of prenatal nutrition on health in adulthood can be transmitted to more than one subsequent generation(Reference Gluckman, Hanson and Beedle10). For example, examination of historical records of the population in Overkalix, Sweden showed that mortality from diabetes increased in men if the paternal grandfather was exposed to abundant nutrition during their pre-pubertal period of slow growth(Reference Kaati, Bygren and Edvinsson11). The children born to women who were in their first trimester during the Dutch Hunger Winter were heavier and those born to women in their third trimester were lighter than the children of women born before the famine(Reference Stein and Lumey12). This suggests the effects of the famine during the grandmother's pregnancy were transmitted to their grandchildren.

Studies in animal models

The findings of epidemiological studies have been largely supported by the results of studies of the effects of specific nutritional interventions during pregnancy and/or lactation in laboratory animals, mainly rats and mice (for detailed reviews see Armitage et al. (Reference Armitage, Khan and Taylor13) and Bertram & Hanson(Reference Bertram and Hanson14)), although studies have also been conducted in other species. The nutritional interventions include feeding a diet with reduced protein content within the physiological range for rodents(Reference Langley and Jackson15), global reduction in food intake(Reference Vickers, Gluckman and Coveny16) or a high-fat diet(Reference Khan, Dekou and Douglas17). In general terms, feeding these diets to pregnant rodents induced in the offspring aspects of metabolic disease and CVD which in man have been associated with a poor prenatal environment. The offspring of these animal models show, albeit to different degrees, impaired control of blood pressure (hypertension and endothelial dysfunction), insulin resistance, dyslipidaemia, obesity and altered locomotor behaviour. In contrast to man, birthweight in rodents is generally unaffected by nutritional constraint during pregnancy, possibly because of the relative immaturity of the pups at birth and the difference in the timing of fat deposition during development. This emphasises that birthweight is a proxy marker for the effects of the intra-uterine environment on the development of the fetus rather than necessarily being a causal component of the mechanism which links environmental exposure to later risk of disease (also consider Roseboom et al. (Reference Roseboom, van der Meulen and Ravelli7)).

Animal models also provide a means of testing the effects of interventions to prevent or reverse the effects of the prenatal environment on the development of the fetus. Two interventions have so far been shown to prevent fetal programming. First, administration of leptin during the neonatal period in rats prevented excessive weight gain in response to a high-fat diet in the offspring of dams which received a 70 % reduction in global nutrition during pregnancy(Reference Vickers, Gluckman and Coveny16). Secondly, increasing the availability of metabolites involved in one-carbon metabolism, specifically glycine and folic acid, to pregnant rats consuming a protein-restricted (PR) diet prevented induction of hypertension and vascular dysfunction in the offspring(Reference Jackson, Dunn and Marchand18Reference Torrens, Brawley and Anthony20). These findings suggest that one-carbon metabolism plays a central role in the induction of an altered phenotype.

Prevention of hypertension by increasing the glycine content of the PR diet(Reference Jackson, Dunn and Marchand18) or impaired lipid and glucose homeostasis by increasing the folic acid content of the PR diet by a similar amount to the daily intake of pregnant women using folic acid supplements(Reference Dunn, Burdge and Jackson21) decreased the growth of the offspring, which suggests normalisation of vascular function and metabolism may occur at the expense of growth and so represent a developmental trade-off. Although increasing the folic acid content of the maternal PR diet prevented hypertension in the offspring, increasing the folic acid content of the protein-sufficient control diet by the same amount increased blood pressure in the offspring(Reference Dunn, Burdge and Jackson21, Reference Burdge, Lillycrop and Jackson22). The relative protein and folic acid content of the diet fed to pregnant rats induces in the offspring opposing changes in fasting blood lipid and glucose concentrations(Reference Burdge, Lillycrop and Jackson22). However, the magnitude of such effects also depends upon the amount of fat fed to the offspring after weaning(Reference Burdge, Lillycrop and Jackson22). Transmission between generations of obesity associated with the epigenetically regulated agouti phenotype in mice was also prevented by dietary supplementation with methyl donors(Reference Waterland, Travisano and Tahiliani23). Thus the physiological phenotype of the offspring is dependent upon interactions between prenatal nutrition and diet in later life. One implication of these findings is that altered susceptibility to disease may reflect the early life environment, but risk may be modified by environmental exposures throughout the life course.

Growth before birth and risk of cancer

Information regarding the effect of the prenatal environment on later risk of cancer is rather more limited than that on the early life origins of CVD and metabolic disease. With the exception of the well-known effects of prenatal exposure to endocrine-disrupting agents such as diethylstilbesterol(Reference Greenwald, Barlow and Nasca24) or ionising radiation(Reference Kato, Yoshimoto and Schull25) on subsequent cancer risk, the precise environmental exposure which leads to cancer is often difficult to define and epidemiological studies are dependent upon testing the strength of associations with proxy measures such as birth weight. Since cancer encompasses a highly heterogeneous group of diseases, the strength of any association between markers of the prenatal environment and subsequent disease risk might be expected to be weak. Nevertheless, associations have been demonstrated between proxy markers of the intra-uterine environment and risk of specific cancers including breast cancer, and leukaemia and some other non-reproductive cancers. To allow comparison between markers of the prenatal environment, and risk of cancer and risk of metabolic disease and CVD, the following discussion will focus on associations with birth weight. However, there are studies which report associations based on other markers of the intra-uterine environment such as length at birth. Many of these are cited in the references listed later.

Breast cancer

The findings of studies which have investigated the association between breast cancer and birth weight are summarised in Table 1. These studies differ markedly in design, the extent to which other variables were included in the analysis, the number of cases studied and whether pre- and post-menopausal women were analysed together or separately. The possible impact of such differences in study design on the reported outcomes have been discussed in detail elsewhere(Reference Ekbom, Gluckman and Hanson26, Reference Michels and Xue27). Where associations between birth weight and risk of breast cancer were found they suggest that, in contrast to CVD or metabolic disease, lower birth weight tended to be associated with lower risk of breast cancer, and that cancer risk increased in a graded manner with increasing birth weight (see references in Table 1). While it has been reported that the association between birth weight and cancer risk is strongest in women who develop the disease before their menopause(1), not all studies support this (Table 1). The extent of association between breast cancer risk and maternal age, parity and maternal smoking are less clear as studies have reported conflicting findings(Reference Ekbom, Hsieh and Lipworth32, Reference Innes, Byers and Schymura34, Reference Sanderson, Shu and Jin39, Reference Okasha, McCarron and Gunnell53) and some have reported J-shaped associations(Reference Innes, Byers and Schymura34). While in some studies the highest birth weights are above 4·5 kg (Table 1), the majority fall below this level. Thus any effect of pathological changes associated with macrosomy cannot account entirely for the positive association between birth weight and risk of breast cancer. A recent meta-analysis supports the overall conclusion that risk of breast cancer is increased in individuals with higher birth weight(Reference Xue and Michels54). Higher birth weight was associated with 12 % increase in relative risk of breast cancer, while higher birth length was associated with 28 % increased risk of disease. Overall, these studies support the suggestion that the intra-uterine environment exerts a persistent effect on the risk of women developing breast cancer.

Table 1 Associations between breast cancer and birth weight

* Studies in which high birth weight was defined as ≥ 4500 g.

Two recent studies have investigated the effect of maternal diet on mammary tumorigenesis in their offspring. de Assis et al. (Reference de Assis, Khan and Hilakivi-Clarke55) found that the offspring of rats fed high-fat diet had structural changes to mammary tissue, lower oestrogen receptor-α expression and increased levels of activated mitogen-activated protein kinase. These offspring showed altered mammary gland structure, increased oestrogen receptor-α, insulin receptor and insulin-like growth factor (IGF)-1 receptor expression and increased sensitivity to tumour induction with 7,12-dimethylbenz[a]anthracene. Fernandez-Twinn et al. (Reference Fernandez-Twinn, Ekizoglou and Gusterson56) reported that the offspring of rats fed a PR diet during pregnancy and lactation had a 2-fold increased sensitivity to mammary tumour induction by nitrosomethylurea. Thus, although these studies used opposing maternal dietary insults, the overall outcome was to increase the sensitivity of mammary tissue to tumorigenic agents. An assessment of maternal folic acid intake in mice on tumour burden in the adult offspring genetically predisposed to intestinal tumours failed to show an effect of prenatal folate exposure or an interaction between prenatal and post-weaning folate exposure(Reference McKay, Williams and Mathers57). However, since all mice produced tumours and the effects of varying maternal folic acid intake were not assessed in wild-type mice, it is possible that any effects of prenatal folate supply on tumorigenesis were masked by the genetic defect.

Leukaemia and hepatoblastoma in childhood

A number of studies have reported a positive association between birth weight and risk of both acute myeloid and lymphoblastic leukaemia in childhood (see references in Table 2). As with the reports that show a positive association between birth weight and breast cancer, the majority of studies report associations within the normal range of birth weights. In contrast, risk of hepatoblastoma has been reported to be increased in children born below the normal range of birth weights(Reference Slovis and Roebuck75). This association is strongest in children born below 2500 g, particularly in those born below 1500 g(Reference Reynolds, Urayama and Von Behren76Reference Feusner and Plaschkes79). It is difficult to explain the opposing relationship between the intra-uterine environment and hepatoblastoma, compared to other cancers. One possible explanation is that the children at risk of hepatoblastoma were born at the lower extreme of birth weight which suggests intra-uterine constraint outside of the normal range which influences patterns of development in the fetus.

Table 2 Associations between childhood leukaemia and birth weight

ALL, acute lymphoblastic leukaemia; AML, acute myeloid leukaemia.

* Studies in which high birth weight was defined as ≥ 4500 g.

Other cancers

McCormack et al. (Reference McCormack, dos Santos Silva and Koupil80) reported detailed statistical analysis of the associations between a number of different cancers and birth weight. When adjusted for birth order and socioeconomic factors at birth and in adulthood, the hazard ratio for each standard deviation increase in birth weight adjusted for gestational age (HRad) for endometrial cancer was 0·79, while the HRad for ovarian cancer was not significantly related to birth weight. There was no significant association between birth weight and prostate cancer in men. This study also described the effect of birth weight on risk of non-reproductive cancers. The HRad for colorectal cancer was 1·16, and lymphatic and haematopoietic tissues 1·19. However, there were no significant associations between birth weight and stomach, liver, pancreatic, respiratory, urinary, neurological, skin and endocrinological cancers. One study has shown an increased OR for osteosarcoma in adults born ≥ 4000 g compared to those born between 3000 and 3500 g(Reference Troisi, Masters and Joshipura81). Also, the OR for all brain tumours in children was 1·05 in those born ≥ 4000 g compared to those born between 2500 and 3990 g, but was higher (OR 1·40) when astrocytomas were considered separately.

For all cancers, the HRad was 1·23 in women less than 50 years, but not related to birth weight in older women, while the HRad for men of all ages was 1·08(Reference McCormack, dos Santos Silva and Koupil80).

Summary

There is a growing body of evidence which supports the suggestion that the intra-uterine environment modifies risk of specific cancers in children and adults. Despite the wide variability of the disease processes including histological origin, time of onset of disease, sex and age of the patients and the means of data collection there is an overall trend towards an increase in risk of specific cancers associated with higher birth weight, particularly at the upper end of the normal range.

Epigenetics and the developmental origins of metabolic disease

Prenatal environment, phenotype induction and gene transcription

The induction of changes to the phenotype of the offspring in response to the prenatal environment that persist throughout the lifespan implies stable changes to gene transcription resulting in altered activities of metabolic pathways and homeostatic control processes, and in differences in the structure of tissues. There are several studies which have investigated the effect of maternal nutrition during pregnancy and/or lactation on gene expression in the offspring in animal models. For example, feeding a PR diet to pregnant rats induced changes to the expression of genes involved in energy balance and glucocorticoid activity in the adult offspring (Table 3). Although the number of genes studied so far is limited, these demonstrate stable effects of nutrient restriction on transcription. Importantly, some of the genes which show altered expression following prenatal under-nutrition are transcription factors which affect multiple pathways in development and nutrient homeostasis; for example PPAR and the glucocorticoid receptor (GR) (Table 3). Thus modifying the regulation of expression of a few key transcription factors may alter the activities of a large number of metabolic and developmental pathways.

Table 3 The effects of maternal dietary protein restriction during pregnancy, or pregnancy and lactation in the rat on the expression of genes associated with energy balance in the adult offspring

Epigenetic regulation of transcription

The methylation of CpG dinucleotides, which are clustered at the 5′ promoter regions of genes, confers stable silencing of transcription(Reference Bird89). Methylation patterns are largely established during embryogenesis or in early postnatal life. Following fertilisation, maternal and paternal genomes undergo extensive demethylation. This is followed by de novo methylation just prior to implantation(Reference Bird89, Reference Reik, Dean and Walter90). About 70 % of CpG are methylated, mainly in repressive heterochromatin regions and in repetitive sequences such as retrotransposable elements(Reference Yoder, Soman and Verdine91). Promoter methylation is important for asymmetrical silencing of imprinted genes(Reference Li, Beard and Jaenisch92) and retrotransposons(Reference Walsh, Chaillet and Bestor93, Reference Waterland and Jirtle94). DNA methylation also plays a key role in cell differentiation by silencing the expression of specific genes during the development and differentiation of individual tissues, and thus the timing of gene methylation is tissue- and gene-specific(Reference Gidekel and Bergman95, Reference Hershko, Kafri and Fainsod96). For some genes, for example δ-crystallin II and phosphoenolpyruvate carboxykinase, there also appear to be gradations of promoter demethylation associated with developmental changes in the role of the gene product(Reference Grainger, Hazard-Leonards and Samaha97, Reference Benvenisty, Mencher and Meyuhas98).

DNA methylation can induce transcriptional silencing by blocking the binding of transcription factors and/or through promoting the binding of the methyl CpG binding protein (MeCP2). The latter binds to methylated cytosines and, in turn, recruits histone modifying complexes composed of deacetylases and histone methyl transferases to the DNA resulting in a closed chromatin structure and transcriptional silencing(Reference Fuks, Hurd and Wolf99, Reference Turner100). The precise effect on transcription depends on the nature of the covalent modification and which N-terminal lysine residue is altered(Reference Strahl, Ohba and Cook101Reference Nakayama, Rice and Strahl105).

Early life environment and epigenetic regulation of transcription in the offspring

Since epigenetic regulation of gene promoters is established during development and is responsible for patterns of transcriptional expression and silencing in adults, perturbations to this process represent a candidate molecular mechanism for induction of persistent alterations in phenotype by the environment experienced in early life. In an elegant study of the effect of maternal behaviour during suckling on the development of stress response in the offspring, Weaver et al. (Reference Weaver, Cervoni and Champagne106) showed that pups raised by rat dams which showed poorer nurturing had an increased stress response. The effect was due to hypermethylation of specific CpG dinucleotides within the promoter of the GR gene in the hippocampus of the offspring which were reversed in the adult offspring by intra-cranial administration of Trichostatin A and l-methionine(Reference Weaver, Cervoni and Champagne106Reference Weaver, Meaney and Szyf108). Uterine artery ligation in the rat decreases p53 expression in the kidney of the offspring, which was associated with increased apoptosis and reduced nephron number(Reference Pham, MacLennan and Chiu109).

Embryo culture and epigenetic regulation of transcription

Nutrition in early life has been shown to alter the epigenetic regulation of transposable elements and of imprinted genes, including IGF-2. These will be summarised here as they are described in detail elsewhere(Reference Waterland and Jirtle110). The composition of the culture medium used to grow mouse embryos alters the expression of IGF-2 and H19 genes by changing the methylation status of their respective promoters(Reference Doherty, Mann and Tremblay111, Reference Khosla, Dean and Brown112). In man, in vitro fertilisation using the intracytoplasmic sperm injection technique is associated with increased risk of Angelman's syndrome(Reference Cox, Burger and Lip113, Reference Orstavik, Eiklid and van der Hagen114) and Beckwith–Weidemann syndrome(Reference DeBaun, Niemitz and Feinberg115) due to loss of methylation of regulatory regions of the UBE3A, and H19 and IGF-2 genes, respectively(Reference Cox, Burger and Lip113, Reference DeBaun, Niemitz and Feinberg115). While such alterations to the epigenetic regulation of imprinted genes produce dramatic alterations to the phenotype of the offspring which are evident in early life, these contrast with the phenotypes induced by variations in maternal nutrition during pregnancy which are more subtle and only become clinically apparent after the neonatal period in childhood or adulthood.

The agouti mouse model

Differences in maternal intake of nutrients involved in one-carbon metabolism, betaine, choline, folic acid and vitamin B12, during pregnancy in the agouti mouse changed the offsprings' coat colour from yellow (agouti) to brown (pseudo-agouti)(Reference Wolff, Kodell and Moore116). This shift is due to increased methylation of seven CpG dinucleotides 600 bp downstream of the Avy intracisternal-A particle insertion site which acts as a cryptic promoter directing the expression of the agouti gene(Reference Waterland and Jirtle110). Differential methylation of the seven CpG dinucleotides was associated with a change in the proportions of mice with the agouti or pseudo-agouti coat colour.

Epigenetic regulation of genes in the offspring of the rat maternal dietary protein restriction mode

Feeding a PR diet to rats during pregnancy induces hypomethylation of the PPARα and GR promoters and increased expression of GR and PPARα in the liver of the recently weaned offspring(Reference Lillycrop, Phillips and Jackson83). This shows that stable changes to the epigenetic regulation of the expression of transcription factors, and hence a phenotype, can be induced in the offspring by modest changes to maternal intake of a macronutrient during pregnancy. However, there is evidence which shows that nutrients involved in one-carbon metabolism play an important role in this process (see later). The expression of the PPARα and GR target genes, acyl-CoA oxidase and phosphoenolpyruvate carboxykinase, was also increased which supports the suggestion that such altered epigenetic regulation of transcription factors modifies the activities of important metabolic pathways(Reference Lillycrop, Phillips and Jackson83, Reference Lillycrop, Slater-Jefferies and Hanson84). Sequence analysis of the PPARα promoter showed that the methylation status of only a few CpG dinucleotides was altered by the PR diet(Reference Lillycrop, Phillips and Torrens117). This suggests that the process of induced epigenetic change is targeted and that the resulting change in transcription may reflect changes in the interaction of the gene with relatively few transcription factors, thus inducing specific changes in the regulation of gene function and hence response to environmental cues. Methylation of the GR and PPARα promoters was also reduced in the heart and the PPARα promoter was hypomethylated in the whole umbilical cord(Reference Burdge, Phillips and Dunn88). Hypomethylation of the GR promoter was associated with an increase in histone modifications which facilitate transcription while those that suppress gene expression were reduced or unchanged(Reference Lillycrop, Slater-Jefferies and Hanson84). While this may be primarily the result of reduced binding of the MeCP2 to the GR promoter because of the reduced level of DNA methylation, reduced MeCP2 expression may also have contributed to higher levels of histone acetylation.

Induction of vascular dysfunction in the offspring of rats fed PR diet during pregnancy was prevented by supplementation of the PR diet with glycine or folic acid(Reference Jackson, Dunn and Marchand18Reference Torrens, Brawley and Anthony20). Hypomethylation of the hepatic GR and PPARα promoters was also prevented by addition of 5-fold more folic acid to the PR diet(Reference Lillycrop, Phillips and Jackson83). Thus one-carbon metabolism plays a central role in the induction of an altered phenotype by maternal dietary restriction as it does in the agouti mouse(Reference Waterland and Jirtle110).

DNA methyltransferases and one-carbon metabolism

Methylation of CpG dinucleotides de novo is catalysed by DNA methyltransferase (Dnmt) 3a and Dnmt3b, and is maintained through mitosis by gene-specific methylation of hemimethylated DNA by Dnmt1(Reference Bird89) (Fig. 1). Over-expression of Dnmt1 results in hypermethylation of DNA and embryonic lethality(Reference Biniszkiewicz, Gribnau and Ramsahoye118), while transient depletion of xDnmt1 in Xenopus embryos induces DNA hypomethylation producing an altered phenotype and sustained depletion causes apoptosis(Reference Stancheva and Meehan119, Reference Stancheva, Hensey and Meehan120). Thus variations in Dnmt1 expression alter the phenotype of the embryo. Dnmt1 activity is inhibited by homocysteine(Reference James, Melnyk and Pogribna121), and Dnmt1 and Dnmt3a expression is modulated by folic acid intake in adult rats(Reference Ghoshal, Li and Datta122). Furthermore, the Dnmt1 promoter contains a GR response element(Reference Rouleau, Tanigawa and Szyf123) which may induce negative regulation of Dnmt1 expression(Reference Ichinose, Miki and Tatematsu124). This suggests how administration of glucocorticoids during pregnancy may induce stable changes to the expression of gluconeogenic enzymes in the offspring as a result of increasing GR activity(Reference Nyirenda, Lindsay and Kenyon125). Thus Dnmt activity may be altered by increased glucocorticoid exposure or as a result of changes to one-carbon metabolism, and so represent one candidate mechanism for the induction of altered epigenetic regulation of genes in response to the intra-uterine environment. Feeding a PR diet to rats during pregnancy induced a reduction in Dnmt1 expression and in binding of Dnmt1 at the GR promoter(Reference Lillycrop, Slater-Jefferies and Hanson84), but not the expression of Dnmt3a, Dnmt3b or the putative DNA demethylase methyl binding domain-2(Reference Detich, Theberge and Szyf126), and the binding of Dnmt3a at the GR promoter was unaltered. This suggests that hypomethylation of the GR promoter in the liver of the offspring, and probably other genes including PPARα, is induced by the maternal diet as a result of lower capacity to maintain patterns of cytosine methylation during mitosis. Down-regulation of Dnmt1 expression may result from increased exposure to homocysteine(Reference Brawley, Torrens and Anthony19, Reference Petrie, Duthie and Rees127) and/or corticosteroids(Reference Langley-Evans, Gardner and Jackson128, Reference Langley-Evans129) which may act directly or by decreasing folate bioavailability(Reference Terzolo, Allasino and Bosio130). The central role of one-carbon metabolism is highlighted by the prevention of reduced Dnmt1 expression by increasing the folic acid content of the PR diet(Reference Lillycrop, Slater-Jefferies and Hanson84). Since Dnmt1 activity appears to be targeted to a subset of gene promoters(Reference Rhee, Jair and Yen131Reference Robertson, Ait-Si-Ali and Yokochi133), altered Dnmt1 expression provides a mechanism for induction of gene-specific promoter hypomethylation. Since Dnmt1 activity is also required for progression through mitosis(Reference Milutinovic, Zhuang and Niveleau134, Reference Suetake, Shi and Watanabe135), reduced Dnmt1 activity could also account for the reduction in embryo cell mass(Reference Kwong, Wild and Roberts136).

Fig. 1 Gene silencing by DNA methylation. Gene expression is silenced in the early embryo by the activities of DNA methyltransferase (Dnmt) 3a and Dnmt3b which catalyse methylation of CpG dinucleotides de novo. This recruits methyl CpG binding protein-2 (MeCP2) which in turn recruits the histone deacetylase (HDAC)–histone methyltransferase (HMT) complex which induce condensation of chromatin at the promoter. Methylation of CpG dinucleotides, and hence gene silencing, is maintained through mitotic cycles by Dnmt1 activity.

A model for the induction of an altered metabolic phenotype in the offspring by prenatal under-nutrition

Based upon current data, we have suggested a mechanism for the induction of an altered phenotype in the offspring by nutrient constraint during pregnancy in which promoter methylation is lost in a gene-specific manner during mitosis due to decreased Dnmt1 expression and activity(Reference Lillycrop, Slater-Jefferies and Hanson84, Reference Burdge, Hanson and Slater-Jefferies85). This is accompanied by reduced binding of the MeCP2–histone deacetylase–histone methyltransferase complex leading to persistence of histone modifications that permit transcription.

Epigenetics and cancer

A change in the epigenetic regulation of genes has been implicated as a causal mechanism in specific cancers including lung, prostate and breast cancer(Reference Liu, Wylie and Andrews137), colon cancer(Reference Zhu138) and haemopoietic cancers(Reference Galm, Herman and Baylin139). Specifically, increased cancer risk is associated with global hypomethylation of the genome with concurrent hypermethylation or hypomethylation of the promoters of specific genes. The mechanism by which global hypomethylation is induced is unclear, but may reflect the global decline in DNA methylation associated with increasing age(Reference Liu, Wylie and Andrews137). The age-related decline in global methylation is related to a reduction in Dnmt1 activity(Reference Lopatina, Haskell and Andrews140) which, in turn, may induce expression of oncogenes such as c-Myc and c-N-ras (Reference Lopatina, Haskell and Andrews140). Thus it appears that modulation of Dnmt1 activity is a key regulatory step in both fetal programming and in the induction of the tumorigenesis. This may be accompanied by methylation de novo of tumour suppressor genes(Reference Lengauer141) by increased Dnmt3a activity leading to aberrant activation of genes involved in cell proliferation and cell differentiation(Reference Strathdee, Appleton and Illand142). Together these changes represent a shift in the regulation of gene control which, in turn, may predispose the genome to further changes in methylation which result ultimately in neoplasia(Reference Issa143). The mechanism leading to gene-specific hypermethylation in cancer is unclear, although it is possible that, as in fetal programming, the balance of nutrients involved in one-carbon metabolism, including folate, vitamin B6 and B12, may modulate the activities of Dnmt(Reference Issa143). Hypermethylation of specific genes in cancer appears to be similar to the effect of increasing the folic acid content of the PR diet fed to pregnant rats on the methylation status of CpG in the liver PPARα promoter in the offspring. While the methylation of CpG which were hypomethylated in the offspring of dams fed a PR diet was normalised by increasing the folic acid content of the maternal PR diet, hypermethylation was also induced in other specific CpG dinucleotides(Reference Lillycrop, Phillips and Torrens117). The observation that nutrition in early life can induce both hypomethylation and hypermethylation of specific CpG dinucleotides suggests a mechanism for induction of different disease endpoints (for example, metabolic disease or cancer) by variation in the same environmental exposure, which is marked by differences in the direction of association between birth weight and disease risk. Prenatal under-nutrition results in hypomethylation of specific CpG dinucleotides in individual gene promoters which would tend to increase binding of regulatory proteins. It is possible therefore that higher nutrient availability, such as increased folic acid intake, may induce hypermethylation of other CpG which would result in different DNA–protein interactions and so induce a shift in regulation. One key example of the role of epigenetics in modulating gene activity by shifting the balance between agonist and suppressor proteins is the induction of tumorigenesis by activation of telomerase in differentiated cells. Telomerase activity is down-regulated in most cells during terminal differentiation in embryogenesis as a result of methylation of the GC-rich promoter region, but is often active in cancer cells. It has been proposed that activation of telomerase in preneoplastic cells is due to a shift in regulation between the activator c-Myc and the suppressor WT1 by changes in the methylation status of specific CpG within the binding domains of these transcription factors in the promoter of the catalytic subunit with confers RT activity (hTERT)(Reference Tollefsbol and Andrews144). One consequence of hTERT activation is to increase Dnmt1 activity(Reference Young, Sedivy and Smith145) leading to copying of aberrant patterns of cytosine methylation. This suggests a synergistic role for hTERT and Dnmt1 in controlling cell proliferation and the methylation status of the genome.

Summary

The findings of epidemiological studies of the relationship between prenatal growth and risk-specific cancers, metabolic disease and CVD suggest that the early life environment is a causal component of the aetiology of these conditions. This is further implied by the common role for altered epigenetic regulation of specific genes and of altered Dnmt activity. Thus, risk of what may generally be considered to be very different disease entities may reflect a continuum of developmental changes which operate via the same enzymes and pathways which induce alterations to the epigenetic regulation of specific genes. Risk of specific diseases may reflect the nature and/or the magnitude of the environmental exposure during early life. It is not known how these environmental cues may be targeted in a manner which induces altered epigenetic regulation of specific genes or of individual CpG dinucleotides and so lead to increased risk of different disease processes. However, such specificity is implied by emerging evidence that the magnitude of the maternal nutritional challenge and the relative amount of specific nutrients in the maternal diet induce directionally opposite changes in the physiology and epigenotype of the offspring(Reference Dunn, Burdge and Jackson21, Reference Lillycrop, Phillips and Jackson83, Reference Gluckman, Lillycrop and Vickers146).

Overall, these findings support the concept that a range of prenatal nutritional environments from constraint to abundance may induce risk of ultimately different pathological processes (Fig. 2). The induced epigenetic changes are likely to be permissive for altered gene expression and hence determine the interaction between an organism and its environment over the life course and, in turn, determine whether increased risk due to the early life environment becomes disease in later life. However, this is an emerging field of research and a substantial number of studies will be required to demonstrate directly a causal association between variations in early life nutrition, induced epigenetic change and differential disease risk, to characterise and understand the underlying mechanisms and to develop prognostic markers in order translate the research findings into clinical tools.

Fig. 2 A model for induction of increased risk of CVD/metabolic disease or cancer by different nutritional exposures acting on the same genome during development. Optimal nutrition during development facilitates establishment of an epigenotype which is expressed as a healthy phenotype. Nutritional constraint induces altered epigenetic regulation in genes associated with increased risk of CVD/metabolic disease. Conversely, nutrient abundance during development induces epigenetic changes in genes associated with increased risk of cancer. However, for both altered epigenotypes, the disease phenotype is only manifest when the organism is exposed to appropriate environmental signals, such as poor diet, during the life course. If these later environmental cues are avoided, possibly by lifestyle choice, then a healthy phenotype is maintained.

Acknowledgements

G. C. B. receives salary support from the British Heart Foundation. G. C. B. wrote the manuscript with substantial contributions from K. A. L. and A. A. J. All authors declare no conflict of interest.

References

1World Cancer Research Fund/American Institute for Cancer Research (2007) Food, Nutrition, Physical Activity and the Prevention of Cancer: A Global Perspective. Washington, DC: American Institute for Cancer Research.Google Scholar
2Vucic, EA, Brown, CJ & Lam, WL (2008) Epigenetics of cancer progression. Pharmacogenomics 9, 215234.CrossRefGoogle ScholarPubMed
3Jackson, AA (2005) Integrating the ideas of life course across cellular, individual, and population levels in cancer causation. J Nutr 135, Suppl. 12, 2927S2933S.CrossRefGoogle ScholarPubMed
4Barker, DJP (1998) Mothers, Babies and Health in Later Life, 2nd ed.Edinburgh: Churchill Livingstone.Google Scholar
5Godfrey, KM & Barker, DJ (2001) Fetal programming and adult health. Public Health Nutr 4, 611624.CrossRefGoogle ScholarPubMed
6Gluckman, PD & Hanson, MA (editors) (2006) Developmental Origins of Disease. Cambridge: Cambridge University Press.CrossRefGoogle Scholar
7Roseboom, TJ, van der Meulen, JH, Ravelli, AC, et al. (2001) Effects of prenatal exposure to the Dutch famine on adult disease in later life: an overview. Mol Cell Endocrinol 185, 9398.CrossRefGoogle Scholar
8Gluckman, PD & Hanson, MA (2004) Living with the past: evolution, development, and patterns of disease. Science 305, 17331736.CrossRefGoogle ScholarPubMed
9Jasienska, G, Thune, I & Ellison, PT (2006) Fatness at birth predicts adult susceptibility to ovarian suppression: an empirical test of the Predictive Adaptive Response hypothesis. Proc Natl Acad Sci U S A 103, 1275912762.CrossRefGoogle ScholarPubMed
10Gluckman, PD, Hanson, MA & Beedle, AS (2007) Non-genomic transgenerational inheritance of disease risk. Bioessays 29, 145154.CrossRefGoogle ScholarPubMed
11Kaati, G, Bygren, LO & Edvinsson, S (2002) Cardiovascular and diabetes mortality determined by nutrition during parents' and grandparents' slow growth period. Eur J Hum Genet 10, 682688.CrossRefGoogle ScholarPubMed
12Stein, AD & Lumey, LH (2000) The relationship between maternal and offspring birth weights after maternal prenatal famine exposure: the Dutch Famine Birth Cohort Study. Hum Biol 72, 641654.Google ScholarPubMed
13Armitage, JA, Khan, IY, Taylor, PD, et al. (2004) Developmental programming of the metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental models in mammals? J Physiol 561, 355377.CrossRefGoogle ScholarPubMed
14Bertram, CE & Hanson, MA (2001) Animal models and programming of the metabolic syndrome. Br Med Bull 60, 103121.CrossRefGoogle ScholarPubMed
15Langley, SC & Jackson, AA (1994) Increased systolic blood pressure in adult rats induced by fetal exposure to maternal low protein diets. Clin Sci (Lond) 86, 217222.CrossRefGoogle ScholarPubMed
16Vickers, MH, Gluckman, PD, Coveny, AH, et al. (2005) Neonatal leptin treatment reverses developmental programming. Endocrinologt 146, 42114216.CrossRefGoogle ScholarPubMed
17Khan, IY, Dekou, V, Douglas, G, et al. (2005) A high-fat diet during rat pregnancy or suckling induces cardiovascular dysfunction in adult offspring. Am J Physiol Regul Integr Comp Physiol 288, R127R133.CrossRefGoogle ScholarPubMed
18Jackson, AA, Dunn, RL, Marchand, MC, et al. (2002) Increased systolic blood pressure in rats induced by a maternal low-protein diet is reversed by dietary supplementation with glycine. Clin Sci (Lond) 103, 633639.CrossRefGoogle ScholarPubMed
19Brawley, L, Torrens, C, Anthony, FW, et al. (2004) Glycine rectifies vascular dysfunction induced by dietary protein imbalance during pregnancy. J Physiol 554, 497504.CrossRefGoogle ScholarPubMed
20Torrens, C, Brawley, L, Anthony, FW, et al. (2006) Folate supplementation during pregnancy improves offspring cardiovascular dysfunction induced by protein restriction. Hypertension 47, 982987.CrossRefGoogle ScholarPubMed
21Dunn, RL, Burdge, GC & Jackson, AA (2003) Folic acid reduces blood pressure in rat offspring from maternal low protein diet but increases blood pressure in offspring of the maternal control diet. Ped Res 53, Suppl., 2A.Google Scholar
22Burdge, GC, Lillycrop, KA, Jackson, AA, et al. (2008) The nature of the growth pattern and of the metabolic response to fasting in the rat are dependent upon the dietary protein and folic acid intakes of their pregnant dams and post-weaning fat consumption. Br J Nutr 99, 540549.CrossRefGoogle ScholarPubMed
23Waterland, RA, Travisano, M, Tahiliani, KG, et al. (2008) Methyl donor supplementation prevents transgenerational amplification of obesity. Int J Obes (Lond), 32, 13731379.CrossRefGoogle ScholarPubMed
24Greenwald, P, Barlow, JJ, Nasca, PC, et al. (1971) Vaginal cancer after maternal treatment with synthetic estrogens. N Engl J Med 285, 390392.CrossRefGoogle ScholarPubMed
25Kato, H, Yoshimoto, Y & Schull, WJ (1989) Risk of cancer among children exposed to atomic bomb radiation in utero: a review. IARC Sci Publ 96, 365374.Google Scholar
26Ekbom, A (2006) The developmental environment and the early origins of cancer. InDevelopmental Origins of Disease, pp. 415425 [Gluckman, PD and Hanson, MA, editors]. Cambridge: Cambridge University Press.CrossRefGoogle Scholar
27Michels, KB & Xue, F (2006) Role of birthweight in the etiology of breast cancer. Int J Cancer 119, 20072025.CrossRefGoogle ScholarPubMed
28Le Marchand, L, Kolonel, LN, Myers, BC, et al. (1988) Birth characteristics of premenopausal women with breast cancer. Br J Cancer 57, 437439.CrossRefGoogle ScholarPubMed
29Ekbom, A, Trichopoulos, D, Adami, HO, et al. (1992) Evidence of prenatal influences on breast cancer risk. Lancet 340, 10151018.CrossRefGoogle ScholarPubMed
30Michels, KB, Trichopoulos, D, Robins, JM, et al. (1996) Birthweight as a risk factor for breast cancer. Lancet 348, 15421546.CrossRefGoogle ScholarPubMed
31Sanderson, M, Williams, MA, Malone, KE, et al. (1996) Perinatal factors and risk of breast cancer. Epidemiology 7, 3437.CrossRefGoogle ScholarPubMed
32Ekbom, A, Hsieh, CC, Lipworth, L, et al. (1997) Intrauterine environment and breast cancer risk in women: a population-based study. J Natl Cancer Inst 89, 7176.CrossRefGoogle ScholarPubMed
33Mogren, I, Damber, L, Tavelin, B, et al. (1999) Characteristics of pregnancy and birth and malignancy in the offspring (Sweden). Cancer Causes Control 10, 8594.CrossRefGoogle ScholarPubMed
34Innes, K, Byers, T & Schymura, M (2000) Birth characteristics and subsequent risk for breast cancer in very young women. Am J Epidemiol 152, 11211128.CrossRefGoogle ScholarPubMed
35De Stavola, BL, Hardy, R, Kuh, D, et al. (2000) Birthweight, childhood growth and risk of breast cancer in a British cohort. Br J Cancer 83, 964968.CrossRefGoogle Scholar
36Kaijser, M, Lichtenstein, P, Granath, F, et al. (2001) In utero exposures and breast cancer: a study of opposite-sexed twins. J Natl Cancer Inst 93, 6062.CrossRefGoogle ScholarPubMed
37Andersson, SW, Bengtsson, C, Hallberg, L, et al. (2001) Cancer risk in Swedish women: the relation to size at birth. Br J Cancer 84, 11931198.CrossRefGoogle ScholarPubMed
38Hilakivi-Clarke, L, Forsen, T, Eriksson, JG, et al. (2001) Tallness and overweight during childhood have opposing effects on breast cancer risk. Br J Cancer 85, 16801684.CrossRefGoogle ScholarPubMed
39Sanderson, M, Shu, XO, Jin, F, et al. (2002) Weight at birth and adolescence and premenopausal breast cancer risk in a low-risk population. Br J Cancer 86, 8488.CrossRefGoogle Scholar
40Titus-Ernstoff, L, Egan, KM, Newcomb, PA, et al. (2002) Early life factors in relation to breast cancer risk in postmenopausal women. Cancer Epidemiol Biomarkers Prev 11, 207210.Google ScholarPubMed
41Vatten, LJ, Maehle, BO, Lund Nilsen, TI, et al. (2002) Birth weight as a predictor of breast cancer: a case–control study in Norway. Br J Cancer 86, 8991.CrossRefGoogle ScholarPubMed
42Ahlgren, M, Sorensen, T, Wohlfahrt, J, et al. (2003) Birth weight and risk of breast cancer in a cohort of 106,504 women. Int J Cancer 107, 9971000.CrossRefGoogle Scholar
43McCormack, VA, dos Santos Silva, I, De Stavola, BL, et al. (2003) Fetal growth and subsequent risk of breast cancer: results from long term follow up of Swedish cohort. Br Med J 326, 248.CrossRefGoogle ScholarPubMed
44Kaijser, M, Akre, O, Cnattingius, S, et al. (2003) Preterm birth, birth weight, and subsequent risk of female breast cancer. Br J Cancer 89, 16641666.CrossRefGoogle ScholarPubMed
45Mellemkjaer, L, Olsen, ML, Sorensen, HT, et al. (2003) Birth weight and risk of early-onset breast cancer (Denmark). Cancer Causes Control 14, 6164.CrossRefGoogle ScholarPubMed
46dos Santos Silva, I, De Stavola, BL, Hardy, RJ, et al. (2004) Is the association of birth weight with premenopausal breast cancer risk mediated through childhood growth? Br J Cancer 91, 519524.CrossRefGoogle ScholarPubMed
47Ahlgren, M, Melbye, M, Wohlfahrt, J, et al. (2004) Growth patterns and the risk of breast cancer in women. N Engl J Med 351, 16191626.CrossRefGoogle ScholarPubMed
48Hodgson, ME, Newman, B & Millikan, RC (2004) Birth weight, parental age, birth order and breast cancer risk in African-American and white women: a population-based case–control study. Breast Cancer Res 6, R656R667.CrossRefGoogle ScholarPubMed
49Lahmann, PH, Gullberg, B, Olsson, H, et al. (2004) Birth weight is associated with postmenopausal breast cancer risk in Swedish women. Br J Cancer 91, 666668.CrossRefGoogle ScholarPubMed
50McCormack, VA, dos Santos Silva, I, Koupil, I, et al. (2005) Birth characteristics and adult cancer incidence: Swedish cohort of over 11 000 men and women. Int J Cancer 115, 611617.CrossRefGoogle Scholar
51Vatten, LJ, Nilsen, TI, Tretli, S, et al. (2005) Size at birth and risk of breast cancer: prospective population-based study. Int J Cancer 114, 461464.CrossRefGoogle ScholarPubMed
52Michels, KB, Xue, F, Terry, KL, et al. (2006) Longitudinal study of birthweight and the incidence of breast cancer in adulthood. Carcinogenesis 27, 24642468.CrossRefGoogle ScholarPubMed
53Okasha, M, McCarron, P, Gunnell, D, et al. (2003) Exposures in childhood, adolescence and early adulthood and breast cancer risk: a systematic review of the literature. Breast Cancer Res Treat 78, 223276.CrossRefGoogle Scholar
54Xue, F & Michels, KB (2007) Intrauterine factors and risk of breast cancer: a systematic review and meta-analysis of current evidence. Lancet Oncol 8, 10881100.CrossRefGoogle ScholarPubMed
55de Assis, S, Khan, G & Hilakivi-Clarke, L (2006) High birth weight increases mammary tumorigenesis in rats. Int J Cancer 119, 15371546.CrossRefGoogle ScholarPubMed
56Fernandez-Twinn, DS, Ekizoglou, S, Gusterson, BA, et al. (2007) Compensatory mammary growth following protein restriction during pregnancy and lactation increases early-onset mammary tumor incidence in rats. Carcinogenesis 28, 545552.CrossRefGoogle ScholarPubMed
57McKay, JA, Williams, EA & Mathers, JC (2008) Gender-specific modulation of tumorigenesis by folic acid supply in the Apc mouse during early neonatal life. Br J Nutr 99, 550558.CrossRefGoogle ScholarPubMed
58MacMahon, B & Newill, VA (1962) Birth characteristics of children dying of malignant neoplasms. J Natl Cancer Inst 28, 231244.Google ScholarPubMed
59Fasal, E, Jackson, EW & Klauber, MR (1971) Birth characteristics and leukemia in childhood. J Natl Cancer Inst 47, 501509.Google ScholarPubMed
60Wertelecki, W & Mantel, N (1973) Increased birth weight in leukemia. Pediatr Res 7, 132138.CrossRefGoogle ScholarPubMed
61Daling, JR, Starzyk, P, Olshan, AF, et al. (1984) Birth weight and the incidence of childhood cancer. J Natl Cancer Inst 72, 10391041.Google ScholarPubMed
62Shaw, G, Lavey, R, Jackson, R, et al. (1984) Association of childhood leukemia with maternal age, birth order, and paternal occupation. A case–control study. Am J Epidemiol 119, 788795.CrossRefGoogle ScholarPubMed
63Eisenberg, DE & Sorahan, T (1987) Birth weight and childhood cancer deaths. J Natl Cancer Inst 78, 10951100.Google ScholarPubMed
64Shu, XO, Gao, YT, Brinton, LA, et al. (1988) A population-based case–control study of childhood leukemia in Shanghai. Cancer 62, 635644.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
65Kaye, SA, Robison, LL, Smithson, WA, et al. (1991) Maternal reproductive history and birth characteristics in childhood acute lymphoblastic leukemia. Cancer 68, 13511355.3.0.CO;2-J>CrossRefGoogle ScholarPubMed
66Savitz, DA & Ananth, CV (1994) Birth characteristics of childhood cancer cases, controls, and their siblings. Pediatr Hematol Oncol 11, 587599.CrossRefGoogle ScholarPubMed
67Cnattingius, S, Zack, MM, Ekbom, A, et al. (1995) Prenatal and neonatal risk factors for childhood lymphatic leukemia. J Natl Cancer Inst 87, 908914.CrossRefGoogle ScholarPubMed
68Cnattingius, S, Zack, M, Ekbom, A, et al. (1995) Prenatal and neonatal risk factors for childhood myeloid leukemia. Cancer Epidemiol Biomarkers Prev 4, 441445.Google ScholarPubMed
69Ross, JA, Potter, JD, Shu, XO, et al. (1997) Evaluating the relationships among maternal reproductive history, birth characteristics, and infant leukemia: a report from the Children's Cancer Group. Ann Epidemiol 7, 172179.CrossRefGoogle ScholarPubMed
70Yeazel, MW, Ross, JA, Buckley, JD, et al. (1997) High birth weight and risk of specific childhood cancers: a report from the Children's Cancer Group. J Pediatr 131, 671677.CrossRefGoogle ScholarPubMed
71Westergaard, T, Andersen, PK, Pedersen, JB, et al. (1997) Birth characteristics, sibling patterns, and acute leukemia risk in childhood: a population-based cohort study. J Natl Cancer Inst 89, 939947.CrossRefGoogle ScholarPubMed
72Okcu, MF, Goodman, KJ, Carozza, SE, et al. (2002) Birth weight, ethnicity, and occurrence of cancer in children: a population-based, incident case–control study in the State of Texas, USA. Cancer Causes Control 13, 595602.CrossRefGoogle ScholarPubMed
73Paltiel, O, Harlap, S, Deutsch, L, et al. (2004) Birth weight and other risk factors for acute leukemia in the Jerusalem Perinatal Study cohort. Cancer Epidemiol Biomarkers Prev 13, 10571064.CrossRefGoogle ScholarPubMed
74Hjalgrim, LL, Rostgaard, K, Hjalgrim, H, et al. (2004) Birth weight and risk for childhood leukemia in Denmark, Sweden, Norway, and Iceland. J Natl Cancer Inst 96, 15491556.CrossRefGoogle ScholarPubMed
75Slovis, TL & Roebuck, DJ (2006) Hepatoblastoma: why so many low-birth-weight infants? Pediatr Radiol 36, 173174.CrossRefGoogle ScholarPubMed
76Reynolds, P, Urayama, KY, Von Behren, J, et al. (2004) Birth characteristics and hepatoblastoma risk in young children. Cancer 100, 10701076.CrossRefGoogle ScholarPubMed
77Feusner, J, Buckley, J, Robison, L, et al. (1998) Prematurity and hepatoblastoma: more than just an association? J Pediatr 133, 585586.CrossRefGoogle ScholarPubMed
78Tanimura, M, Matsui, I, Abe, J, et al. (1998) Increased risk of hepatoblastoma among immature children with a lower birth weight. Cancer Res 58, 30323035.Google ScholarPubMed
79Feusner, J & Plaschkes, J (2002) Hepatoblastoma and low birth weight: a trend or chance observation? Med Pediatr Oncol 39, 508509.CrossRefGoogle ScholarPubMed
80McCormack, VA, dos Santos Silva, I, Koupil, I, et al. (2005) Birth characteristics and adult cancer incidence: Swedish cohort of over 11 000 men and women. Int J Cancer 115, 611617.CrossRefGoogle Scholar
81Troisi, R, Masters, MN, Joshipura, K, et al. (2006) Perinatal factors, growth and development, and osteosarcoma risk. Br J Cancer 95, 16031607.CrossRefGoogle ScholarPubMed
82Bertram, C, Trowern, AR, Copin, N, et al. (2001) The maternal diet during pregnancy programs altered expression of the glucocorticoid receptor and type 2 11β-hydroxysteroid dehydrogenase: potential molecular mechanisms underlying the programming of hypertension in utero. Endocrinology 142, 28412853.CrossRefGoogle ScholarPubMed
83Lillycrop, KA, Phillips, ES, Jackson, AA, et al. (2005) Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J Nutr 135, 13821386.CrossRefGoogle ScholarPubMed
84Lillycrop, KA, Slater-Jefferies, JL, Hanson, MA, et al. (2007) Induction of altered epigenetic regulation of the hepatic glucocorticoid receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests that reduced DNA methyltransferase-1 expression is involved in impaired DNA methylation and changes in histone modifications. Br J Nutr 97, 10641073.CrossRefGoogle ScholarPubMed
85Burdge, GC, Hanson, MA, Slater-Jefferies, JL, et al. (2007) Epigenetic regulation of transcription: a mechanism for inducing variations in phenotype (fetal programming) by differences in nutrition during early life? Br J Nutr 97, 10361046.CrossRefGoogle ScholarPubMed
86Bogdarina, I, Murphy, HC, Burns, SP, et al. (2004) Investigation of the role of epigenetic modification of the rat glucokinase gene in fetal programming. Life Sci 74, 14071415.CrossRefGoogle ScholarPubMed
87Maloney, CA, Gosby, AK, Phuyal, JL, et al. (2003) Site-specific changes in the expression of fat-partitioning genes in weanling rats exposed to a low-protein diet in utero. Obes Res 11, 461468.CrossRefGoogle ScholarPubMed
88Burdge, GC, Phillips, ES, Dunn, RL, et al. (2004) Effect of reduced maternal protein consumption during pregnancy in the rat on plasma lipid concentrations and expression of peroxisomal proliferator-activated receptors in the liver and adipose tissue of the offspring. Nutr Res 24, 639646.CrossRefGoogle Scholar
89Bird, A (2001) DNA methylation patterns and epigenetic memory. Genes Dev 16, 621.CrossRefGoogle Scholar
90Reik, W, Dean, W & Walter, J (2001) Epigenetic reprogramming in mammalian development. Science 293, 10891093.CrossRefGoogle ScholarPubMed
91Yoder, JA, Soman, NS, Verdine, GL, et al. (1997) DNA (cytosine-5)-methyltransferases in mouse cells and tissues. Studies with a mechanism-based probe. J Mol Biol 270, 385395.CrossRefGoogle ScholarPubMed
92Li, E, Beard, C & Jaenisch, R (1993) Role for DNA methylation in genomic imprinting. Nature 366, 362365.CrossRefGoogle ScholarPubMed
93Walsh, CP, Chaillet, JR & Bestor, TH (1998) Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nature Genet 20, 116117.CrossRefGoogle ScholarPubMed
94Waterland, RA & Jirtle, RL (2003) Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol 23, 52935300.CrossRefGoogle ScholarPubMed
95Gidekel, S & Bergman, Y (2002) A unique developmental pattern of Oct-3/4 DNA methylation is controlled by a cis-demodification element. J Biol Chem 277, 3452134530.CrossRefGoogle ScholarPubMed
96Hershko, AY, Kafri, T, Fainsod, A, et al. (2003) Methylation of HoxA5 and HoxB5 and its relevance to expression during mouse development. Gene 302, 6572.CrossRefGoogle ScholarPubMed
97Grainger, RM, Hazard-Leonards, RM, Samaha, F, et al. (1983) Is hypomethylation linked to activation of delta-crystallin genes during lens development? Nature 306, 8891.CrossRefGoogle ScholarPubMed
98Benvenisty, N, Mencher, D, Meyuhas, O, et al. (1985) Sequential changes in DNA methylation patterns of the rat phosphoenolpyruvate carboxykinase gene during development. Proc Natl Acad Sci U S A 82, 267271.CrossRefGoogle ScholarPubMed
99Fuks, F, Hurd, PJ, Wolf, D, et al. (2003) The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J Biol Chem 278, 40354040.CrossRefGoogle ScholarPubMed
100Turner, BM (2000) Histone acetylation and an epigenetic code. Bioessays 22, 836845.3.0.CO;2-X>CrossRefGoogle Scholar
101Strahl, BD, Ohba, R, Cook, RG, et al. (1999) Methylation of histone H3 at lysine 4 is highly conserved and correlates with transcriptionally active nuclei in Tetrahymena. Proc Natl Acad Sci U S A 96, 1496714972.CrossRefGoogle ScholarPubMed
102Lachner, M, O'Carroll, D, Rea, S, et al. (2001) Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116120.CrossRefGoogle ScholarPubMed
103Zegerman, P, Canas, B, Pappin, D, et al. (2002) Histone H3 lysine 4 methylation disrupts binding of nucleosome remodeling and deacetylase (NuRD) repressor complex. J Biol Chem 277, 1162111624.CrossRefGoogle ScholarPubMed
104Litt, MD, Simpson, M, Gaszner, M, et al. (2001) Correlation between histone lysine methylation and developmental changes at the chicken beta-globin locus. Science 293, 24532455.CrossRefGoogle ScholarPubMed
105Nakayama, J, Rice, JC, Strahl, BD, et al. (2001) Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly. Science 292, 110113.CrossRefGoogle ScholarPubMed
106Weaver, IC, Cervoni, N, Champagne, FA, et al. (2004) Epigenetic programming by maternal behavior. Nature Neurosci 7, 847854.CrossRefGoogle ScholarPubMed
107Weaver, IC, Champagne, FA, Brown, SE, et al. (2005) Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking later in life. J Neurosci 25, 1104511054.CrossRefGoogle ScholarPubMed
108Weaver, IC, Meaney, MJ & Szyf, M (2006) Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc Natl Acad Sci U S A 103, 34803485.CrossRefGoogle ScholarPubMed
109Pham, TD, MacLennan, NK, Chiu, CT, et al. (2003) Uteroplacental insufficiency increases apoptosis and alters p53 gene methylation in the full-term IUGR rat kidney. Am J Physiol Regul Integr Comp Physiol 285, R962R970.CrossRefGoogle ScholarPubMed
110Waterland, RA & Jirtle, RL (2004) Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition 20, 6368.CrossRefGoogle ScholarPubMed
111Doherty, AS, Mann, MR, Tremblay, KD, et al. (2000) Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo. Biol Reprod 62, 15261535.CrossRefGoogle ScholarPubMed
112Khosla, S, Dean, W, Brown, D, et al. (2001) Culture of preimplantation mouse embryos affects fetal development and the expression of imprinted genes. Biol Reprod 64, 918926.CrossRefGoogle ScholarPubMed
113Cox, GF, Burger, J, Lip, V, et al. (2002) Intracytoplasmic sperm injection may increase the risk of imprinting defects. Am J Hum Genet 71, 162164.CrossRefGoogle ScholarPubMed
114Orstavik, KH, Eiklid, K, van der Hagen, CB, et al. (2003) Another case of imprinting defect in a girl with Angelman syndrome who was conceived by intracytoplasmic semen injection. Am J Hum Genet 72, 218219.CrossRefGoogle Scholar
115DeBaun, MR, Niemitz, EL & Feinberg, AP (2003) Association of in vitro fertilization with Beckwith–Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet 72, 156160.CrossRefGoogle ScholarPubMed
116Wolff, GL, Kodell, RL, Moore, SR, et al. (1998) Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. FASEB J 12, 949957.CrossRefGoogle ScholarPubMed
117Lillycrop, KA, Phillips, ES, Torrens, C, et al. (2008) Feeding pregnant rats a protein-restricted diet persistently alters the methylation of specific cytosines in the hepatic PPARα promoter of the offspring. Br J Nutr 100, 278282.CrossRefGoogle ScholarPubMed
118Biniszkiewicz, D, Gribnau, J, Ramsahoye, B, et al. (2002) Dnmt1 overexpression causes genomic hypermethylation, loss of imprinting, and embryonic lethality. Mol Cell Biol 22, 21242135.CrossRefGoogle ScholarPubMed
119Stancheva, I & Meehan, RR (2000) Transient depletion of xDnmt1 leads to premature gene activation in Xenopus embryos. Genes Dev 14, 313327.CrossRefGoogle ScholarPubMed
120Stancheva, I, Hensey, C & Meehan, RR (2001) Loss of the maintenance methyltransferase, xDnmt1, induces apoptosis in Xenopus embryos. EMBO J 20, 19631973.CrossRefGoogle ScholarPubMed
121James, SJ, Melnyk, S, Pogribna, M, et al. (2002) Elevation in S-adenosylhomocysteine and DNA hypomethylation: potential epigenetic mechanism for homocysteine-related pathology. J Nutr 132, Suppl., 2361S2366S.CrossRefGoogle ScholarPubMed
122Ghoshal, K, Li, X, Datta, J, et al. (2006) A folate- and methyl-deficient diet alters the expression of DNA methyltransferases and methyl CpG binding proteins involved in epigenetic gene silencing in livers of F344 rats. J Nutr 136, 15221527.CrossRefGoogle ScholarPubMed
123Rouleau, J, Tanigawa, G & Szyf, M (1992) The mouse DNA methyltransferase 5′-region. A unique housekeeping gene promoter. J Biol Chem 267, 73687377.CrossRefGoogle Scholar
124Ichinose, M, Miki, K, Tatematsu, M, et al. (1990) Hydrocortisone-induced enhancement of expression and changes in methylation of pepsinogen genes in stomach mucosa of the developing rat. Biochem Biophys Res Commun 172, 10861093.CrossRefGoogle ScholarPubMed
125Nyirenda, MJ, Lindsay, RS, Kenyon, CJ, et al. (1998) Glucocorticoid exposure in late gestation permanently programs rat hepatic phosphoenolpyruvate carboxykinase and glucocorticoid receptor expression and causes glucose intolerance in adult offspring. J Clin Invest 101, 21742181.CrossRefGoogle ScholarPubMed
126Detich, N, Theberge, J & Szyf, M (2002) Promoter-specific activation and demethylation by MBD2/demethylase. J Biol Chem 277, 3579135794.CrossRefGoogle ScholarPubMed
127Petrie, L, Duthie, SJ, Rees, WD, et al. (2002) Serum concentrations of homocysteine are elevated during early pregnancy in rodent models of fetal programming. Br J Nutr 88, 471477.CrossRefGoogle ScholarPubMed
128Langley-Evans, SC, Gardner, DS & Jackson, AA (1996) Maternal protein restriction influences the programming of the rat hypothalamic-pituitary-adrenal axis. J Nutr 126, 15781585.CrossRefGoogle ScholarPubMed
129Langley-Evans, SC (1997) Hypertension induced by foetal exposure to a maternal low-protein diet, in the rat, is prevented by pharmacological blockade of maternal glucocorticoid synthesis. J Hypertens 15, 537544.CrossRefGoogle ScholarPubMed
130Terzolo, M, Allasino, B, Bosio, S, et al. (2004) Hyperhomocysteinemia in patients with Cushing's syndrome. J Clin Endocrinol Metab 89, 37453751.CrossRefGoogle ScholarPubMed
131Rhee, I, Jair, KW, Yen, RW, et al. (2000) CpG methylation is maintained in human cancer cells lacking DNMT1. Nature 404, 10031007.CrossRefGoogle ScholarPubMed
132Jackson-Grusby, L, Beard, C, Possemato, R, et al. (2001) Loss of genomic methylation causes p53-dependent apoptosis and epigenetic deregulation. Nature Genet 27, 3139.CrossRefGoogle ScholarPubMed
133Robertson, KD, Ait-Si-Ali, S, Yokochi, T, et al. (2000) Dnmt1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nature Genet 25, 338342.CrossRefGoogle ScholarPubMed
134Milutinovic, S, Zhuang, Q, Niveleau, A, et al. (2003) Epigenomic stress response. Knockdown of DNA methyltransferase 1 triggers an intra-S-phase arrest of DNA replication and induction of stress response genes. J Biol Chem 278, 1498514995.CrossRefGoogle ScholarPubMed
135Suetake, L, Shi, L, Watanabe, D, et al. (2001) Proliferation stage-dependent expression of DNA methyltransferase (Dnmt1) in mouse small intestine. Cell Struct Funct 26, 7986.CrossRefGoogle ScholarPubMed
136Kwong, WY, Wild, AE, Roberts, P, et al. (2000) Maternal undernutrition during the preimplantation period of rat development causes blastocyst abnormalities and programming of postnatal hypertension. Development 127, 41954202.CrossRefGoogle ScholarPubMed
137Liu, L, Wylie, RC, Andrews, LG, et al. (2004) Aging, cancer and nutrition: the DNA methylation connection. Mech Ageing Dev 124, 989998.CrossRefGoogle Scholar
138Zhu, J (2006) DNA methylation and hepatocellular carcinoma. J Hepatobiliary Pancreat Surg 13, 265273.Google ScholarPubMed
139Galm, O, Herman, JG & Baylin, SB (2006) The fundamental role of epigenetics in hematopoietic malignancies. Blood Rev 20, 113.CrossRefGoogle ScholarPubMed
140Lopatina, N, Haskell, JF, Andrews, LG, et al. (2002) Differential maintenance and de novo methylating activity by three DNA methyltransferases in aging and immortalized fibroblasts. J Cell Biochem 84, 324334.CrossRefGoogle ScholarPubMed
141Lengauer, C (2003) Cancer. An unstable liaison. Science 300, 442443.CrossRefGoogle ScholarPubMed
142Strathdee, G, Appleton, K, Illand, M, et al. (2001) Primary ovarian carcinomas display multiple methylator phenotypes involving known tumor suppressor genes. Am J Pathol 158, 11211127.CrossRefGoogle ScholarPubMed
143Issa, JP (1999) Aging, DNA methylation and cancer. Crit Rev Oncol Hematol 32, 3143.CrossRefGoogle ScholarPubMed
144Tollefsbol, TO & Andrews, LG (2001) Mechanisms for telomerase gene control in aging cells and tumorigenesis. Med Hypotheses 56, 630637.CrossRefGoogle ScholarPubMed
145Young, JI, Sedivy, JM & Smith, JR (2003) Telomerase expression in normal human fibroblasts stabilizes DNA 5-methylcytosine transferase I. J Biol Chem 278, 1990419908.CrossRefGoogle ScholarPubMed
146Gluckman, PD, Lillycrop, KA, Vickers, MH, et al. (2007) Metabolic plasticity during mammalian development is directionally dependent on early nutritional status. Proc Natl Acad Sci U S A 104, 1279612800.CrossRefGoogle ScholarPubMed
Figure 0

Table 1 Associations between breast cancer and birth weight

Figure 1

Table 2 Associations between childhood leukaemia and birth weight

Figure 2

Table 3 The effects of maternal dietary protein restriction during pregnancy, or pregnancy and lactation in the rat on the expression of genes associated with energy balance in the adult offspring

Figure 3

Fig. 1 Gene silencing by DNA methylation. Gene expression is silenced in the early embryo by the activities of DNA methyltransferase (Dnmt) 3a and Dnmt3b which catalyse methylation of CpG dinucleotides de novo. This recruits methyl CpG binding protein-2 (MeCP2) which in turn recruits the histone deacetylase (HDAC)–histone methyltransferase (HMT) complex which induce condensation of chromatin at the promoter. Methylation of CpG dinucleotides, and hence gene silencing, is maintained through mitotic cycles by Dnmt1 activity.

Figure 4

Fig. 2 A model for induction of increased risk of CVD/metabolic disease or cancer by different nutritional exposures acting on the same genome during development. Optimal nutrition during development facilitates establishment of an epigenotype which is expressed as a healthy phenotype. Nutritional constraint induces altered epigenetic regulation in genes associated with increased risk of CVD/metabolic disease. Conversely, nutrient abundance during development induces epigenetic changes in genes associated with increased risk of cancer. However, for both altered epigenotypes, the disease phenotype is only manifest when the organism is exposed to appropriate environmental signals, such as poor diet, during the life course. If these later environmental cues are avoided, possibly by lifestyle choice, then a healthy phenotype is maintained.