Hostname: page-component-7c8c6479df-ph5wq Total loading time: 0 Render date: 2024-03-26T17:14:29.831Z Has data issue: false hasContentIssue false

Epigenetic modifications and human pathologies: cancer and CVD

Published online by Cambridge University Press:  11 November 2010

Susan J. Duthie*
Affiliation:
Nutrition and Epigenetics Group, Division of Vascular Health, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, UK
*
Correspondence author: Dr Susan J. Duthie, fax +44 1224 716629, email s.duthie@abdn.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Epigenetic changes are inherited alterations in DNA that affect gene expression and function without altering the DNA sequence. DNA methylation is one epigenetic process implicated in human disease that is influenced by diet. DNA methylation involves addition of a 1-C moiety to cytosine groups in DNA. Methylated genes are not transcribed or are transcribed at a reduced rate. Global under-methylation (hypomethylation) and site-specific over-methylation (hypermethylation) are common features of human tumours. DNA hypomethylation, leading to increased expression of specific proto-oncogenes (e.g. genes involved in proliferation or metastasis) can increase the risk of cancer as can hypermethylation and reduced expression of tumour suppressor (TS) genes (e.g. DNA repair genes). DNA methyltransferases (DNMT), together with the methyl donor S-adenosylmethionine (SAM), facilitate DNA methylation. Abnormal DNA methylation is implicated not only in the development of human cancer but also in CVD. Polyphenols, a group of phytochemicals consumed in significant amounts in the human diet, effect risk of cancer. Flavonoids from tea, soft fruits and soya are potent inhibitors of DNMT in vitro, capable of reversing hypermethylation and reactivating TS genes. Folates, a group of water-soluble B vitamins found in high concentration in green leafy vegetables, regulate DNA methylation through their ability to generate SAM. People who habitually consume the lowest level of folate or with the lowest blood folate concentrations have a significantly increased risk of developing several cancers and CVD. This review describes how flavonoids and folates in the human diet alter DNA methylation and may modify the risk of human colon cancer and CVD.

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

Abbreviations:
EGCG

(−)-epigallocatechin 3-gallate

SAH

S-adenosylhomocysteine

SAM

S-adenosylmethionine

DNMT

DNA methyltransferase

MMP

matrix metalloproteinase

SMC

smooth muscle cell

TS

tumour suppressor

Epigenetics and regulation of gene activity

Epigenetics is defined as processes that act to regulate heritable changes in gene activity that are transmitted through meiosis and mitosis but that are not accompanied by changes in the DNA coding sequence. Epigenetic signals (or marks) control gene expression through remodelling of chromatin. Chromatin consists of DNA and DNA-binding proteins including histones, which interact to form repetitive nucleoprotein units called nucleosomes. Epigenetic control of gene expression is critically important in regulating embryonic development, cellular differentiation and organogenesis and for key biological processes such as imprinting, and silencing of large chromosomal domains such as the X-chromosome in females(Reference Jones and Baylin1). Conversely, dysregulation of epigenetic processes may be causal for several human diseases including cancer and heart disease(Reference Jones and Baylin1, Reference Turunen, Aavik and Yla-Herttula2) (Fig. 1).

Fig. 1. Epigenetics, diet and human disease. A simplified scheme illustrating the structure of mammalian chromatin and defining the role of epigenetic modifications both in normal human development and disease.

Fig. 2. Folate deficiency, cancer and CVD: a common mechanism? A simplified scheme describing how folate deficiency may alter normal DNA methylation and how this could impact both on risk of colon cancer and atherosclerosis.

Epigenetic mechanisms include nucleosome remodelling, histone modifications (including acetylation, methylation and ubiquitination), regulatory non-coding (small and micro) RNA and DNA methylation. These are linked processes and all contribute to controlling gene activity by changing chromatin architecture and/or access by transcription factors(Reference Jones and Baylin1).

Dysregulation of any of these processes has severe consequences for the phenotype and function of the cell. During malignant transformation, cancer cells can exhibit global changes in the chromatin structure affecting the whole epigenome, altering the expression of several hundreds of genes and perturbing entire metabolic pathways. Consequently, it has been proposed that epigenetic dysregulation can be as detrimental to the cell as mutations in DNA coding(Reference Jones and Baylin1).

Epigenetics, diet and disease

Although epigenetic marks remain comparatively stable during mitotic transmission, they are influenced by ageing and by the environment. In an elegant study of monozygotic twins aged between 3 and 74 years (n 40 pairs), genome-wide cytosine methylation, gene-specific DNA methylation and histone acetylation (H3 and H4) patterns in lymphocytes were found to correlate strongly in young twins, but these associations weakened both with age and depending on how divergent the twin's health and lifestyles had become(Reference Fraga, Ballestar and Paz3). Approximately one-third of twins harboured epigenetic differences. Global gene expression, measured by microarray analysis, differed more that fourfold in the oldest twin pair compared to gene activity in the youngest(Reference Fraga, Ballestar and Paz3).

Diet can profoundly alter epigenetic patterns in animals. The Agouti mouse has been used extensively to investigate the impact of maternal nutrition on the fetal epigenome and the phenotype of the offspring. In this model, coat colour is linked to the methylation status of the agouti gene, which is highly dependent on maternal diet. Most importantly, altered epigenetic marking is associated with an increased risk of diseases including a diabetes-like condition, obesity and tumorigenesis(Reference Yen, Gill and Frigeri4, Reference Dolinoy5). The influence of diet on epigenetics and fetal programming in relation to health and disease will be covered in greater detail in a subsequent paper (KA Lillycrop, in this issue).

How diet contributes to epigenetic control of gene expression and human disease risk is poorly understood. It is acknowledged that abnormal epigenetic control of gene expression increases disease risk (certainly for cancer) and that diet can also profoundly influence risk. What remains to be established is a causal link between diet and epigenetics in the development of human disease and whether diet is differentially effective at various stages in the human lifespan.

Certain bioactive food components, such as sulforaphanes from broccoli, diallyl disuphides from garlic and resveratrol in wine, have been shown (in vitro and in vivo) to alter epigenetic processes with positive consequences for cell function, including control of proliferation, up-regulated apoptosis and a reduction in inflammation(Reference Ross, Dwyer and Umar6). All of these food components are associated with a reduced risk of human cancer at various sites. Inhibition of histone deacetylase activity increases the transcription factor access to DNA and induces gene reactivation. Isothiocyanates present in cruciferous vegetables inhibit histone deacetylase activity in colon, breast and prostate cancer cells in vitro. Sulforaphane, a powerful inducer of phase 2 detoxification enzymes, increases histone acetylation and inhibits intestinal polyp formation in Apc min mice. Moreover, histone deacetylase activity (H3 and H4) is rapidly inhibited (3–6 h) in blood cells from human volunteers fed a relatively small single bolus (68 g) of broccoli sprouts (reviewed in(Reference Dashwood and Ho7)). Conversely, diallyl disulfide from garlic increases H3 and H4 histone acetylation, tumour suppressor (p21) gene expression and inhibits colon cancer cell growth in culture (reviewed in(Reference Druesne-Pecollo, Chaumontet and Latino-Martel8)). The polyphenols curcumin (from the spice turmeric) and resveratrol (from red wine), which have strong antioxidant, anti-inflammatory and anti-carcinogenic properties, also modify histone acetylation patterns in circulating inflammatory cells and induce histone deacetylase activity(Reference Rahman9).

In contrast, the impact of diet on recently identified epigenetic processes, for instance, endogenous non-coding or regulatory (small and micro) RNA that suppress gene activity at the transcriptional level and where dysregulation may increase the risk of cancer remain unexplored(Reference Guil and Esteller10).

Most studies investigating how diet alters the epigenome have explored how dietary components alter DNA methylation.

Aberrant DNA methylation and abnormal gene expression

Post-replicative DNA methylation in mammals involves addition of a 1-C methyl group at the fifth carbon position of cytosine residues within CpG dinucleotides. S-adenosylmethionine (SAM) acts as methyl donor in this reaction, which is catalysed by DNA methyltransferase (DNMT) enzymes. DNMT 3 (3a and 3b) regulates de novo methylation during development, whereas DNMT 1 maintains DNA methylation patterns during cell replication. Approximately 4% of cytosines in DNA are modified to 5-methylcytosine, with most occurring in the sequence, CpG. The genome methylation pattern is precisely inherited during mitosis and is highly tissue specific and species specific. Cytosine methylation changes the structure of the major groove in the DNA molecule and disrupts the attachment of DNA-binding proteins and transcription factors. In general, genes methylated at specific sites (for example upstream of a promoter region) are either not transcribed into mRNA or are transcribed at a reduced rate, and translation into the protein for which the gene encodes is reduced. Epigenetic DNA methylation therefore contributes to the control of gene and ultimately protein expression(Reference Costello and Plass11).

Most information on how dysregulated DNA methylation affects human disease risk comes from research into cancer. Two types of aberrant DNA methylation are universally present in tumours. DNA, both from benign and malignant carcinomas, is substantially undermethylated (hypomethylated) compared to DNA from adjacent normal tissue. DNA hypomethylation is associated with increased transcription and expression of proto-oncogenes that stimulate malignant cell growth. Genome-wide DNA hypomethylation occurs early in carcinogenesis and precedes mutation and deletion events. Conversely, increased DNA methylation (hypermethylation) is associated with gene silencing. Hypermethylation of cytosines in the promoter region of tumour suppressor (TS) genes (for example DNA repair genes and genes that down-regulate cell proliferation) is linked with cancer progression. Gene-specific DNA hypermethylation occurs both in early and advanced stages of malignant transformation. Aberrant DNA methylation and gene expression patterns may be causal in tumorigenesis(Reference Gaudet, Hodgson and Eden12).

Many bioactive food compounds including folate, selenium, flavonoids, alcohol and fatty acids are associated with an altered risk both of human colon cancer and heart disease. All of these dietary components modify cytosine methylation. The impact of several of these phytochemicals and micronutrients on DNA methylation in colon carcinogenesis are reviewed comprehensively elsewhere(Reference Johnson and Belshaw13, Reference Arasaradnam, Commane and Bradburn14).

The remainder of this review describes how specific dietary polyphenols reactivate TS genes by reversing DNA hypermethylation and the consequence of this for risk of cancer and how the vitamin B folate modifies cytosine methylation in relation to both heart disease and colon carcinogenesis.

Polyphenols, DNA methyltransferase activity and tumour suppressor gene re-expression

Hypermethylation of CpG islands in the gene promoter contributes to gene silencing. TS gene hypermethylation is a universal event in early cancer, with more genes becoming hypermethylated as the disease progresses. Drugs such as 5-aza-2′-deoxycytidine (decitabine) that reverse DNA hypermethylation and re-establish TS gene expression are used in cancer therapy. However, they are genotoxic to normal cells. Maintenance of normal DNA methylation and gene expression patterns by dietary phytochemicals and reversal of methylation-induced inactivation of TS genes may be an alternative approach for the prevention and treatment of cancer.

Polyphenols, a group of plant phytochemicals consumed in significant amounts in the human diet modulate genomic stability and risk of cancer in vitro and in vivo (Reference Duthie, Duthie and Kyle15Reference Duthie17). There is strong evidence that polyphenols from tea, soft fruits and berries, vegetables, apples, and even from wine, are potent anti-carcinogenic agents in vitro and in animal models that prevent DNA instability at several sites in the carcinogenic pathway(Reference Duthie, Duthie and Kyle15Reference Duthie17). Genoprotective mechanisms include modulation of carcinogen metabolism (either by inhibiting the activation of cytochromes P450 or by inducing detoxification by glutathione transferase and glucuronyltransferase), decreased binding of the carcinogen to DNA, inhibition of oxidative DNA damage, alteration in cell signalling and gene expression, reduced inflammation, increased apoptosis and inhibition of malignant transformation, cell invasiveness, angiogenesis and metastasis(Reference Duthie, Duthie and Kyle15). Moreover, several polyphenols are potent inhibitors of DNMT activity in vitro, capable of reversing DNA hypermethylation and reactivating TS gene activity. Polyphenols inhibit DNMT activity and DNA methylation in two ways. First, by direct insertion into the binding pocket of DNMT (competitive inhibition) and second, indirectly by decreasing intracellular SAM concentrations and non-competitively inhibiting DNMT activity(Reference Fang, Chen and Yang18). Tea and soya are two of the most widely consumed plant products worldwide. Tea components (especially green and black tea) and specific soya isoflavones inhibit DNMT activity in human cancer cells. Genistein from soyabean and (−)-epigallocatechin 3-gallate (EGCG) from green tea are the most potent DNMT inhibitors. EGCG (and other green tea metabolites) dose-dependently inhibit DNMT activity in human oesophageal cancer cells(Reference Fang, Chen and Yang18). Molecular modelling reveals that the gallate moiety on the D ring of EGCG interacts strongly with the cytosine-active site on the DNMT enzyme. Additionally, hydrogen bonds formed between hydroxyl groups of the A and B rings and Sr1229 and Cys 1225 on the protein contribute to the high-affinity binding of EGCG and inhibition of activity(Reference Fang, Chen and Yang18). Genistein (from soya) was also found to interact strongly with DNMT, dose-dependently inhibiting enzyme activity(Reference Fang, Chen and Yang18). Certain polyphenols are substrates for catechol O-methyltransferase, which is structurally analogous to DNMT. SAM is consumed in the reaction and intracellular S-adenosylhomocysteine (SAH), a potent inhibitor of DNMT is elevated. Polyphenols including EGCG, quercetin, myricetin and fisetin may decrease DNA methylation by altering the cellular ratio SAM:SAH and indirectly inhibiting DNMT activity. There is currently little evidence in support of this mechanism. Feeding mice green tea extract containing EGCG (65% (v/v) for 7 d) or transgenic Apc min mice (0·16% (v/v) EGCG for 9 weeks) did not alter intestinal or liver SAH(Reference Fang, Chen and Yang18).

DNMT 1 inhibition demethylates CpG islands in the promoter regions of silenced TS genes including p16ink4a, retinoic acid receptor β, methylguanine methyltransferase, mMLH1 and glutathione S-transferase π in cancer cells. Reactivation of TS genes is associated with a corresponding increase in mRNA and protein expression. EGCG (20–50 μm for up to 6 d) reverses DNA hypermethylation and increases expression of methylguanine methyltransferase, p16 and hMLH1 in cultured KYSE 510 oesophageal cancer cells. Changes in cytosine methylation and mRNA expression were detected after only 48 h exposure to the green tea polyphenol and were progressive with time. Moreover, reversal of DNA hypermethylation was associated with increased protein expression (determined by Western blotting). EGCG can also reactivate retinoic acid receptor β in prostate and breast cancer cells, p16 in colon cancer cells and glutathione S-transferase π in prostate cells(Reference Fang, Wang and Hou19Reference Shukla, Trokhan and Resnik21). Genistein (at the same concentrations) partially reversed DNA hypermethylation and reactivated p16, retinoic acid receptor β and methylguanine methyltransferase gene expression(Reference Fang, Chen and Sun22). The isoflavones biochanin A and daidzein and the flavonoids, myricetin, quercetin, hesperitin, naringenein, apigenin and luteolin were active, but less effective at altering DNA methylation and reactivating TS gene expression(Reference Fang, Chen and Sun22). Reactivation of TS gene expression by polyphenols is associated with reduced cancer cell progression. Malignant transformation involves phenotypic changes in initiated cells that promote invasion into extracellular tissue matrices. The TS matrix metalloproteinase (MMP) inhibitor RECK inhibits angiogenesis, invasion and metastasis. RECK activity is down-regulated in several human cancers and is associated with promoter hypermethylation in the gene. In an elegant study, Kato et al.(Reference Kato, Long and Makita23) investigated how EGCG altered RECK DNA methylation and expression in four human oral squamous cell carcinoma cell lines. Critically, they investigated whether reversing hypermethylation and increasing RECK gene expression could inhibit oral squamous cell carcinoma migration and invasiveness. EGCG (20–50 μm for up to 6 d) dose- and time-dependently decreased RECK DNA methylation and increased mRNA in two of the four cell lines. Significantly, this was associated with a subsequent decrease in MMP-2 and MMP-9 expression and in the ability of the cancer cells to invade a three-dimensional collagen model. Cell proliferation, depth of migration and frequency and size of invasive foci were all reduced by EGCG(Reference Kato, Long and Makita23). However, the ability of EGCG to decrease DNA hypermethylation and reactivate TS gene expression and activity is inconsistent, with some studies reporting no effect of EGCG on DNA methylation in several human cancer cell lines(Reference Chuang, Yoo and Kwan24, Reference Stressman, Brueckner and Musch25).

Evidence for a modulating effect of dietary polyphenols on DNA methylation and re-expression of TS genes in vivo is less convincing. Consumption of genistein by mice appears to correlate positively with changes in prostate DNA methylation at CpG islands(Reference Day, Bauer and DesBordes26). Conversely, feeding a composite of green tea polyphenols (0·1, 0·3 and 0·6% (v/v) in drinking water for 12 or 24 weeks) to wild-type or TRAMP (transgenic adenocarcinoma of mouse prostate) mice did not change normal or cancer-specific DNA methylation patterns either in target prostate tissue or in gut and liver tissue(Reference Morey-Kinney, Zhang and Pascual27). In this model, genome-wide DNA hypomethylation occurs at an early stage in cancer progression with loci-specific hypermethylation detected in late-stage disease. No significant dose- or time-dependent effect of GTP on either global- or gene-specific DNA methylation (measured by several different biomarkers) was observed(Reference Morey-Kinney, Zhang and Pascual27).

In general, the concentrations of polyphenols employed to inhibit DNMT activity both in vitro and in vivo are considerably higher than can be achieved nutritionally. The levels of EGCG shown to be effective in reversing DNA hypermethylation and TS gene silencing (generally 10–50 μm) are 50-fold higher than blood and tissue concentrations measured after tea drinking(Reference Fang, Chen and Yang18). The highest dose of GTP studied in the TRAMP mouse model (0·6% (v/v)) is equivalent to a human drinking eighteen cups of green tea per day. Similarly, the concentration of genistein needed to alter cytosine methylation and TS gene activity (5–20 μm) are 3–10-fold higher than can be achieved by eating soya products(Reference Fang, Chen and Yang18). Dietary polyphenols are rapidly metabolized intracellularly through several biochemical pathways, including glucuronidation, sulfation and methylation. The lack of effect of polyphenols on DNMT activity, DNA methylation and gene expression in vivo is probably due to limited functional bioavailability post-metabolism. Whether dietary polyphenols can modify DNA methylation and TS gene expression in human subjects is unknown, but it is doubtful whether these compounds, consumed at nutritional levels, will have a major impact on DNA methylation and gene reactivation.

Folate deficiency and human diseases

Folates, a family of water-soluble B vitamins, play a crucial role in the development of human diseases such as cancer and heart disease in adults, cognitive dysfunction and dementia in the elderly and congenital defects in babies(Reference Czeizel28Reference Duthie, Whalley and Collins30). Suboptimal folate status is widespread with 40% of 15–18 year olds in the UK exhibiting marginal folate status and overt folate deficiency common in people over 65 years of age, especially in the institutionalized elderly.

Low-folate status has been implicated in cancer development, notably of the cervix, lung, breast, brain, colorectum and pancreas. The evidence linking folate deficiency and human cancer is strongest for the colon. Data from the majority of human studies (retrospective, case–control and prospective) suggest that individuals with the highest habitual folate intake or with the highest circulating folate concentrations have a reduced risk of developing colon polyps or tumours(Reference Giovannucci31Reference Kim33). Folate acts to maintain genomic stability by regulating DNA biosynthesis, DNA repair and DNA methylation. Folate deficiency both initiates and accelerates cancer by disrupting each of these processes.

Poor folate status is also associated with an increased risk of CVD. Historically, this has been attributed to the metabolic relationship between low folate and high homocysteine (hyperhomocysteinemia). Adverse effects of homocysteine include impairment of endothelial and smooth muscle cell (SMC) function, changes to vasodilation, altered extracellular matrix biochemistry, endoplasmic reticulum stress and increased coagulation and impaired fibrinolysis(Reference McNulty, Pentieva and Hoey34). Most human observational studies support a positive relationship between homocysteine and CVD(35Reference Wierzbicki37). However, several recent large intervention trials have failed to find any benefit of homocysteine-lowering therapies against death from cardiovascular causes in patients with pre-existing vascular disease or diabetics(38). Consequently, there is considerable debate whether homocysteine is causal for vascular dysfunction(Reference Splaver, Lamas and Hennekens36). In two recent prospective studies of men without prior CVD, high-serum folate was independently and significantly associated with a reduced risk of vascular events after adjusting for relevant confounders(Reference Voutilainen, Lakka and Porkkala-Sarataho39, Reference Voutilainen, Virtanen and Rissanen40). Blood homocysteine levels did not correlate with risk(Reference Voutilainen, Virtanen and Rissanen40). Several alternative theories of how low folate may independently increase CVD have been proposed. Folate can directly influence vascular endothelial cell function. Nitric oxide production and vasodilation is impaired in CVD. Folates are critical in maintaining nitric oxide production and preventing oxygen radical formation by physically stabilizing tetrahydrobiopterin in the conversion of l-arginine to l-citrulline and preventing endothelial nitric oxide synthase uncoupling. Folic acid supplementation improves endothelial cell function in hyperhomocysteinemic patients with coronary artery disease, in diabetics, in subjects with hypercholesterolemia and in healthy subjects fed a high methionine load(Reference Doshi, McDowell and Moat41, Reference Usui, Matsuoka and Miyazaki42). However, in addition to deregulating vascular endothelial cells, low folate may perturb vascular SMC gene expression, growth and function by disrupting DNA methylation(Reference Turunen, Aavik and Yla-Herttula2).

Folate deficiency, cancer and CVD: a common mechanism

Within the methionine cycle, 5-methyltetrahydrofolate remethylates homocysteine to methionine, which is subsequently metabolized to SAM. As described earlier, SAM serves as 1-C donor in the methylation of DNA. Under conditions of low-dietary folate, SAM concentrations are depleted causing hypomethylation of newly synthesised DNA and increased proto-oncogene expression (see aberrant DNA methylation and abnormal gene expression above). While abnormal DNA methylation and gene expression is a consistent event in tumorigenesis, aberrant cytosine methylation and proto-oncogene activation may also be a common mechanism linking malignant transformation in cancer and vascular dysfunction in heart disease (Fig. 2). The atherosclerotic plaque and the tumour are both monoclonal in origin with unregulated cell proliferation providing a growth advantage for selected clones. During atherogenesis, SMC (which make up approximately 90% of the aorta cell population) transform from quiescent, contractile cells to synthetic cells that migrate to the intima and rapidly multiply. Overexpression of growth factors and pro-proliferative oncogenes such as c-myc and p53 (which are commonly dysregulated in cancer) also drive SMC invasiveness and plaque formation. Moreover, dysregulated expression of genes involved in lipid accumulation, connective tissue formation, calcification and inflammation further accelerate plaque progression. Hence, changes in the phenotype and invasiveness of SMC in the vascular plaque are similar to changes in monoclonality and cell proliferation described for tumour formation(Reference Turunen, Aavik and Yla-Herttula2).

Low-folate status is associated both with alterations in cytosine methylation in experimental and human studies (see later) and an increased risk of malignant transformation. Whether folate status can similarly influence DNA methylation and risk of vascular disease remains to be established.

The remainder of this review will describe evidence that abnormal DNA methylation may be causal in the development of atherosclerosis and that folate status influences genome-wide DNA methylation and the incidence of both colon cancer and vascular disease. How gene-specific DNA hypermethylation influences disease risk was covered in a paper also presented at this symposium (E Lund, unpublished results).

Folate, abnormal genome-wide DNA methylation and colon cancer

While extreme methyl donor depletion alters genome-wide cytosine methylation (primarily in the liver) and induces hepatocarcinogenesis in animal models(Reference Wainfan and Poirier43, Reference Pogribny, James and Jernigan44), the effect of folate deficiency alone on global DNA methylation in vitro and in vivo is strongly influenced by the treatment regime, rodent species, tissue and genes examined (reviewed in(Reference Kim33)). Folate depletion decreases global DNA methylation in some human and animal cell lines in vitro, but not in others. Mouse NIH/3T3 fibroblast and CHO-K1 DNA became hypomethylated after 12 d in folate-depleted medium, but not in human colon adenocarcinoma HCT116 and Caco-2 cells(Reference Stempak, Sohn and Chiang45). Conversely, global and p53 TS gene DNA was hypomethylated in human colon adenoma cells grown in folate-depleted medium but was restored by folic acid repletion(Reference Wasson, McGlynn and McNulty46). We have shown that global cytosine methylation is decreased in SV40-immortalised human colonocytes grown in folate-free medium for 14 d(Reference Duthie, Narayanan and Blum47) but not in NCM460 non-malignantly transformed human colon cells under the same conditions(Reference Duthie, Mavrommatis and Rucklidge48). Severe and prolonged folate depletion in rodents induces global DNA hypomethylation in the liver and certain regions of the colon(Reference Balaghi and Wagner49Reference James, Pogribny and Pogribna51). Moderate folate deficiency does not consistently induce global DNA hypomethylation in blood, liver and colon, despite a significant depletion in blood and tissue folate and liver SAM(Reference Sohn, Stempack and Reid52Reference Duthie54).

Lymphocyte DNA is hypomethylated in women made experimentally folate deficient over several weeks(Reference Jacob, Gretz and Taylor55, Reference Rampersaud, Kauwell and Hutson56) and low-dietary folate intake (<200 μg/d) correlates with hypomethylation of long-interspersed nucleotide element repeats (LINE-1; used as an indicator of global DNA methylation) in human colon tumours(Reference Schernhammer, Giovannucci and Kawasaki57). Conversely, LINE-1 methylation was not associated either with intake or blood folate in colon biopsy tissue collected from the Aspirin/Folate Polyp Prevention study(Reference Figueiredo, Grau and Wallace58). Little association between folate and genome-wide DNA methylation is apparent in healthy individuals with sufficient blood folate(Reference Kim33, Reference Fenech, Aitken and Rinaldi59, Reference Ingrosso, Cimmino and Perna60). The effect of supplementation with synthetic folic acid on DNA methylation is highly inconsistent and dependent on initial folate status, the level and duration of intervention, the genes and tissue reported and the health status of the subject(Reference Duthie54). DNA hypomethylation is reversed in lymphocytes from folate-deficient volunteers repleted with synthetic folic acid(Reference Jacob, Gretz and Taylor55, Reference Rampersaud, Kauwell and Hutson56) and in leucocytes from colorectal adenoma patients given folic acid (400 μg/d for 10 weeks(Reference Pufulete, Al-Ghnaniem and Khushal61)). However, DNA methylation remained unchanged in the colonic mucosa of these patients(Reference Pufulete, Al-Ghnaniem and Khushal61). Pharmacological (rather than nutritional) concentrations of folic acid (5–10 mg/d for 3–6 months) increased DNA methylation in the colorectum(Reference Cravo, Fidalgo and Pereira62, Reference Cravo, Pinto and Chaves63), but intervention with a more moderate dose (1 mg folic acid/d for 3 years) did not(Reference Figueiredo, Grau and Wallace58). Supplementing healthy volunteers (with initial circulating folate concentrations within the normal range) has little impact on blood cell DNA methylation despite significantly elevated whole blood, plasma and lymphocyte folate(Reference Fenech, Aitken and Rinaldi59, Reference Basten, Duthie and Pirie64).

Folate, DNA methylation and vascular disease

While there is good evidence that abnormal DNA methylation is causal for human carcinogenesis, decreased genomic methylation is also observed in cultured vascular cells and diseased vascular tissue. 5-methylcytosine levels drop rapidly (about 65%) in healthy rabbit aorta SMC cultured from aortic explants. Moreover, these cells change from a contractile to an invasive phenotype(Reference Hiltunen, Turunen and Hakkinen65). Hypomethylated DNA is also detectable in atherosclerotic plaques. Global genomic DNA is under methylated in advanced atherosclerotic lesions isolated from ApoE knockout mice and denuded New Zealand White rabbit aorta(Reference Hiltunen, Turunen and Hakkinen65). Most importantly, changes in DNA methylation patterns in mononuclear cells and in the aorta of ApoE mice occur early (4 weeks of age) and prior to appearance of vascular lesions, supporting a pathogenic role for abnormal DNA methylation in atherosclerosis(Reference Lund, Andersson and Lauria66). Cytosine methylation is reduced (9%) in advanced human atherosclerotic lesions compared to normal arteries or arteries with fatty streaks and at a level similar to that detected in human tumours(Reference Hiltunen, Turunen and Hakkinen65). Both global genomic DNA and gene-specific DNA is hypomethylated in blood cells from uraemia patients (with hyperhomocysteinemia) compared to healthy subjects(Reference Ingrosso, Cimmino and Perna60). Gene-specific hypomethylation has also been measured in human atherosclerotic advanced lesions and is associated with expression of the SMC proto-oncogene platelet-derived growth factor(Reference Hiltunen, Turunen and Hakkinen65) which may increase SMC proliferation in the atherosclerotic lesion. In addition to increasing SMC proliferation and invasiveness, DNA hypomethylation may also accelerate atherogenicity by up-regulating genes involved in lipid deposition and inflammation and increasing plaque instability. Several CpG islands are hypomethylated in the 15-lipoxygenase gene promoter and mRNA expression is correspondingly increased in human plaques(Reference Hiltunen, Turunen and Hakkinen65). As in cancer, gene-specific DNA hypermethylation is also present in atherosclerosis. Methylation of the estrogen receptor-α gene (which restricts cell proliferation) is increased (about two-fold) in arteries from patients with severe atherosclerosis compared to healthy subjects(Reference Post, Goldschmidt-Clermont and Wilhide67). In addition, estrogen receptor-α hypermethylation is associated with a switch from a contractile to synthetic phenotype in human SMC explants(Reference Ying, Hassanain and Roos68). Aberrant expression of other genes implicated in atherosclerotic plaque progression include interferon-γ, MMP-2, MMP-7, tissue inhibitor of metalloproteinases (TIMP)-3, p53 and EC-SOD (extracellular superoxide dismutase)(Reference Hiltunen and Yla-Herttuala69).

Cancer and atherosclerosis are diseases of old age. Moreover, normal ageing is associated with altered DNA methylation patterns, making it difficult to clearly establish the effects of diet alone on DNA methylation and disease risk. Similarly, determining whether low folate induces plaque development through abnormal DNA methylation independently of homocysteine is complicated by the intimate metabolic relationship between these two metabolites. While there is evidence that homocysteine is associated with altered DNA methylation and vascular disease, currently there is only very limited evidence that folate alone alters cytosine methylation in atherosclerosis.

In heterozygous and homozygous variant methylenetetrahydrofolate reductase mice, folate concentrations are low, homocysteine is elevated and brain and ovary tissue DNA is hypomethylated. Lipid deposition is increased in the aorta of aged methylenetetrahydrofolate reductase variant mice(Reference Chen, Karaplis and Ackerman70). Similarly, in mice heterozygous for the cystathionine-β-synthase gene, DNA methylation patterns in the imprinted H19 gene are changed in response to hyperhomocysteinemia(Reference Devlin, Bottiglieri and Domann71). However, here, alterations in methylation profiles are highly tissue specific and do not correlate consistently with H19 gene expression(Reference Devlin, Bottiglieri and Domann71). Plasma homocysteine and DNA methylation in peripheral mononuclear cells are negatively associated in healthy human subjects(Reference Yi, Melnyk and Pogribna72), while supraphysiological folate supplementation (15 mg/d for 8 weeks) reverses genome-wide DNA hypomethylation and abnormal gene expression (H19, IGF2) in hyperhomocysteinemic patients(Reference Ingrosso, Cimmino and Perna60).

In an attempt to establish the independent effects of folate and hyperhomocysteinemia on vascular function, Brown et al. (Reference Brown, Huang and Lu73) developed an in vitro model of prolonged folate deficiency in cultured human endothelial (EA.hy926) cells(Reference Brown, Huang and Lu73). Several intracellular folate vitamers were significantly depleted (up to 99%) but homocysteine, SAM and SAH were unchanged relative to cells grown in adequate folic acid. MCP-1, a cytokine up-regulated early in atherogenesis, was increased providing limited evidence that folate deficiency (independently of homocysteine) is associated with an atherogenic phenotype. However, global DNA methylation was unaltered and gene-specific methylation was not measured(Reference Brown, Huang and Lu73).

The effect of hyperhomocysteinemia and/or vitamin B deficiency on aortic plaque formation has been investigated extensively in rodent models. Generally, hyperhomocysteinemia is associated with a 2-fold increase in aortic plaque area(Reference Hoffman, Lalla and Lu74Reference Zhou, Moller and Ritskes-Hoitinga76) but vitamin B deficiency is not(Reference Hoffman, Lalla and Lu74, Reference Troen, Lutgens and Smith77). Conversely, vitamin B supplementation (up to three times control levels in the diet) is reported to decrease endogenous and homocysteine-induced plaque formation in ApoE null mice(Reference Hoffman, Lalla and Lu74, Reference Zhou, Moller and Ritskes-Hoitinga76). However, the data are inconsistent and profoundly dependent on the dietary regimen(Reference Hoffman, Lalla and Lu74, Reference Zhou, Moller and Ritskes-Hoitinga76). DNA methylation was not measured in any of these studies. We have recently developed an ApoE mouse model of folate and combined folate, vitamins B6 and B12 deficiency that demonstrates either mild (2-fold increase in homocysteine) or moderate hyperhomocysteinemia (7-fold increase in homocysteine). Folate (but not combined vitamin B deficiency) significantly increased atherosclerotic plaque formation (approximately 17%) in the aorta of mice fed a high fat Westernised diet for 16 weeks. Moreover plaque area was not associated with plasma homocysteine. However, global DNA methylation in the mouse heart, aorta and liver was similar across all groups indicating that disease progression is probably not related to altered cytosine methylation in response either to vitamin B or hyperhomocysteinemia (SJ Duthie, unpublished results).

Conclusion

Diet strongly influences the risk of developing cancer and heart disease. Epigenetic changes are present in malignantly transformed tumour cells and in atherosclerotic lesions. Abnormal DNA methylation in target (epithelial or smooth muscle) cells is common to both diseases. Moreover, diet can modulate epigenetic marking and gene expression. There is some evidence that specific nutrients in the human diet that are strongly associated with risk of vascular disease and cancer can modulate DNA methylation.

Certain polyphenols, notably from green tea and soya, inhibit DNMT activity in cancer cells in vitro and reactivate tumour suppressor genes silenced by DNA hypermethylation. The data are weaker in animal studies. Polyphenols undergo extensive metabolism in vivo and limited bioavailability of the active compound (or metabolites) may explain lack of effect at nutritional concentrations.

High-dietary-folate and high-blood-folate status are generally associated with a decreased risk of colorectal cancer and heart disease. Folates have a critical role in maintaining DNA stability by donating 1-C moieties and maintaining DNA methylation. Cell, animal and human studies demonstrate that folate deficiency induces epigenetic changes by attenuating remethylation of SAH to SAM in the methionine cycle, leading to hyperhomocysteinemia, cytosine demethylation, global DNA hypomethylation and proto-oncogene activation. Dysregulated DNA methylation has been proposed to be a common mechanism linking cancer and heart disease by up-regulation of proto-oncogene expression and induction of epithelial (cancer) and SMC (vascular disease) proliferation and migration. However, the effect of folate status on DNA methylation is profoundly dependent not only on the severity and duration of the folate depletion, but also on the gene, tissue and stage of malignant transformation. Similarly, while genome-wide DNA hypomethylation is detected in atherosclerotic plaques from experimental animals and human tissue, the impact of vitamin B and homocysteine on DNA methylation and disease risk remains to be established. On balance, the evidence available currently does not strongly support the hypothesis that altered genome-wide DNA methylation, as a direct consequence of low-folate or vitamin B status, increases human heart disease risk.

Acknowledgements

Funding was provided by the Scottish Government Rural and Environment Research and Analysis Directorate. The author declares no conflict of interest.

References

1.Jones, PA & Baylin, SB (2007) The epigenomics of cancer. Cell 128, 683692.CrossRefGoogle ScholarPubMed
2.Turunen, MP, Aavik, E & Yla-Herttula, S (2009) Epigenetics and atherosclerosis. Biochim Biophys Acta 1790, 886891.CrossRefGoogle ScholarPubMed
3.Fraga, MF, Ballestar, E, Paz, MF et al. (2005) Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA 102, 1060410609.CrossRefGoogle ScholarPubMed
4.Yen, TT, Gill, AM, Frigeri, LG et al. (1994) Obesity, diabetes, and neoplasia in yellow Avy/− mice: ectopic expression of the agouti gene. FASEB J 8, 479488.CrossRefGoogle ScholarPubMed
5.Dolinoy, DC (2008) The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr Rev 66 (Suppl. 1), S7–S11.CrossRefGoogle Scholar
6.Ross, SA, Dwyer, J, Umar, A et al. (2008) Diet, epigenetic events and cancer prevention. Nutr Rev 66 (Suppl. 1), S1S6.CrossRefGoogle ScholarPubMed
7.Dashwood, RH & Ho, E (2008) Dietary agents as histone deacetylase inhibitors: sulforaphane and structurally related isothiocyanates. Nutr Rev 66 (Suppl. 1), S36S38.CrossRefGoogle ScholarPubMed
8.Druesne-Pecollo, N, Chaumontet, C & Latino-Martel, P (2008) Diallyl disulfide increases histone acetylation in colon cells in vitro and in vivo. Nutr Rev 66 (Suppl. 1), S39S41.CrossRefGoogle ScholarPubMed
9.Rahman, I (2008) Dietary polyphenols mediated regulation of oxidative stress and chromatin remodeling in inflammation. Nutr Rev 66 (Suppl. 1), S42S45.CrossRefGoogle ScholarPubMed
10.Guil, S & Esteller, M (2009) DNA methylomes, histone codes and miRNAs: tying it all together. Int J Biochem Cell Biol 41, 8795.CrossRefGoogle ScholarPubMed
11.Costello, JF & Plass, C (2001) Methylation matters. J Med Genet 38, 285303.CrossRefGoogle ScholarPubMed
12.Gaudet, F, Hodgson, JG, Eden, A et al. (2003) Induction of tumors in mice by genomic hypomethylation. Science 300, 489492.CrossRefGoogle ScholarPubMed
13.Johnson, IT & Belshaw, NJ (2008) Environment, diet and CpG island methylation: epigenetic signals in gastrointestinal neoplasia. Food Chem Toxicol 46, 13461359.CrossRefGoogle ScholarPubMed
14.Arasaradnam, RP, Commane, DM, Bradburn, D et al. (2008) A review of dietary factors and its influence on DNA methylation in colorectal carcinogenesis. Epigenetics 3, 193198.CrossRefGoogle ScholarPubMed
15.Duthie, GG, Duthie, SJ & Kyle, JAM (2000) Plant polyphenols in cancer and heart disease: implications as nutritional antioxidants. Nutr Res Rev 13, 79106.CrossRefGoogle ScholarPubMed
16.Ferguson, LR (2001) Role of plant polyphenols in genomic stability. Mutat Res 475, 80111.CrossRefGoogle ScholarPubMed
17.Duthie, SJ (2007) Berry phytochemicals, genomic stability and cancer: evidence for chemoprotection at several stages in the carcinogenic process. Mol Nutr Food Res 51, 665674.CrossRefGoogle ScholarPubMed
18.Fang, M, Chen, D & Yang, CS (2007) Dietary polyphenols may affect DNA methylation. J Nutr 137, 223S228S.CrossRefGoogle ScholarPubMed
19.Fang, MZ, Wang, Y, Hou, Z et al. (2003) Tea polyphenol (−)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res 63, 75637570.Google ScholarPubMed
20.Lee, WJ, Shim, JY & Zhu, BT (2005) Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Mol Pharmacol 68, 10181030.CrossRefGoogle ScholarPubMed
21.Shukla, S, Trokhan, S, Resnik, MI et al. (2005) Epigallocatcehin-3-gallate causes demethylation and activation of GSTP1 gene expression in human prostate cancer LNCaP cells. Proc Am Assoc Cancer Res 46, 1572.Google Scholar
22.Fang, MZ, Chen, D, Sun, Y et al. (2005) Reversal of hypermethylation and reactivation of p16INKa, RAR β and MGMT genes by genistein and other isoflavones from soy. Clin Cancer Res 11, 70337041.CrossRefGoogle Scholar
23.Kato, K, Long, NK, Makita, H et al. (2008) Effects of green tea polyphenol on methylation status of RECK gene and cancer cell invasion in oral squamous cell carcinoma cells. Br J Cancer 99, 647654.CrossRefGoogle ScholarPubMed
24.Chuang, JC, Yoo, CB, Kwan, JM et al. (2005) Comparison of biological effects of non-nucleoside DNA methylation inhibitors versus 5-aza-2′-deoxycytidine. Mol Cancer Ther 4, 15151520.CrossRefGoogle Scholar
25.Stressman, C, Brueckner, B, Musch, et al. (2006) Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 66, 27942800.CrossRefGoogle Scholar
26.Day, JK, Bauer, AM, DesBordes, C et al. (2002) Genistein alters methylation patterns in mice. J Nutr 132, 2419S2423S.CrossRefGoogle ScholarPubMed
27.Morey-Kinney, SR, Zhang, W, Pascual, M et al. (2009) Lack of evidence for green tea polyphenols as DNA methylation inhibitors in murine prostate. Cancer Prev Res 2, 10651075.CrossRefGoogle ScholarPubMed
28.Czeizel, AE (1993) Prevention of congenital abnormalities by periconceptional multivitamin supplementation. Br Med J 306, 16451648.CrossRefGoogle ScholarPubMed
29.Boushey, CJ, Beresford, SAA, Omenn, GS et al. (1995) A quantitative assessment of plasma homocysteine as a risk factor for vascular disease: probable benefits of increasing folic acid intakes. JAMA 274, 10491057.CrossRefGoogle ScholarPubMed
30.Duthie, SJ, Whalley, LJ, Collins, AR et al. (2002) Homocysteine, B vitamin status and cognitive function in the elderly. Am J Clin Nutr 75, 908913.CrossRefGoogle ScholarPubMed
31.Giovannucci, E (2002) Epidemiologic studies of folate and colorectal neoplasia: a review. J Nutr 132, 2350S2355S.CrossRefGoogle ScholarPubMed
32.Sanjoaquin, MA, Allen, N, Couto, E et al. (2005) Folate intake and colorectal cancer risk: a meta-analytical approach. Int J Cancer 113, 825828.CrossRefGoogle Scholar
33.Kim, YI (2007) Folate and colorectal cancer: an evidence-based critical review. Mol Nutr Food Res 51, 267292.CrossRefGoogle ScholarPubMed
34.McNulty, H, Pentieva, K, Hoey, L et al. (2008) Homocysteine, B vitamins and CVD. Proc Nutr Soc 67, 232237.CrossRefGoogle ScholarPubMed
35.The Homocysteine Studies Collaboration (2002) Homocysteine and risk of ischemic heart disease and stroke: a meta-analysis. JAMA 288, 20152022.CrossRefGoogle Scholar
36.Splaver, A, Lamas, GA, Hennekens, CH et al. (2004) Homocysteine and cardiovascular disease: biological mechanisms, observational epidemiology and the need for randomised trials. Am Heart J 148, 3440.CrossRefGoogle Scholar
37.Wierzbicki, AS (2007) Homocysteine and cardiovascular disease: a review of the evidence. Diabetes Vasc Dis Res 4, 143149.CrossRefGoogle ScholarPubMed
38.The Heart Outcomes Prevention Evaluation (HOPE)2 Investigators (2006) Homocysteine lowering with folic acid and B vitamins in vascular disease. NEJM 354, 15671577.CrossRefGoogle Scholar
39.Voutilainen, S, Lakka, TA, Porkkala-Sarataho, E et al. (2000) Low serum folate concentrations are associated with an excess incidence of acute coronary events: the Kuopio Ischaemic Heart Disease Risk Factor Study. Eur J Clin Nutr 54, 424428.CrossRefGoogle ScholarPubMed
40.Voutilainen, S, Virtanen, JK, Rissanen, TH et al. (2004) Serum folate and homocysteine and the incidence of acute coronary events: the Kuopio Ischaemic Heart Disease Risk Factor Study. Am J Clin Nutr 80, 317323.CrossRefGoogle ScholarPubMed
41.Doshi, SN, McDowell, IFW, Moat, SJ et al. (2002) Folic acid improves endothelial function in coronary artery disease via mechanisms largely independent of homocysteine lowering. Circulation 105, 2226.CrossRefGoogle ScholarPubMed
42.Usui, M, Matsuoka, H, Miyazaki, H et al. (1999) Endothelial dysfunction by acute hyperhomocysteinemia: restoration by folic acid. Clin Sci 96, 235239.CrossRefGoogle ScholarPubMed
43.Wainfan, E & Poirier, LA (1992) Methyl groups in carcinogenesis: effects on DNA methylation and gene expression. Cancer Res 52, 2071s2077s.Google ScholarPubMed
44.Pogribny, IP, James, SJ, Jernigan, S et al. (2004) Genomic hypomethylation is specific for preneoplastic liver in folate/methyl deficient rats and does not occur in non-target tissues. Mutat Res 548, 5359.CrossRefGoogle Scholar
45.Stempak, JM, Sohn, K-Y, Chiang, E-P et al. (2005) Cell and stage of transformation-specific effects of folate deficiency on methionine cycle intermediates and DNA methylation in an in vitro model. Carcinogenesis 26, 981990.CrossRefGoogle ScholarPubMed
46.Wasson, GR, McGlynn, AP, McNulty, H et al. (2006) Global DNA and p53 region-specific hypomethylation in human colonic cells is induced by folate depletion and reversed by folate supplementation. J Nutr 136, 27482753.CrossRefGoogle ScholarPubMed
47.Duthie, SJ, Narayanan, S, Blum, S et al. (2000) Folate deficiency in vitro induces uracil misincorporation, DNA hypomethylation and inhibits DNA excision repair in immortalised normal human colon epithelial cells. Nutr Cancer 37, 127133.CrossRefGoogle Scholar
48.Duthie, SJ, Mavrommatis, Y, Rucklidge, G et al. (2008) The response of human colonocytes to folate deficiency in vitro: functional and proteomic analysis. J Proteome Res 7, 32543266.CrossRefGoogle Scholar
49.Balaghi, M & Wagner, C (1993) DNA methylation in folate deficiency: use of CpG methylase. Biochem Biophys Res Commun 193, 11841190.CrossRefGoogle ScholarPubMed
50.Kim, Y-I, Pogribny, IP, Salomon, RN et al. (1996) Exon-specific DNA hypomethylation of the p53 gene of rat colon induced by dimethylhydrazine: modulation by dietary folate. Am J Clin Pathol 149, 11291137.Google ScholarPubMed
51.James, SJ, Pogribny, IP, Pogribna, M et al. (2003) Mechanisms of DNA damage, DNA hypomethylation and tumour progression in the folate/methyl deficient rat model. J Nutr 133, 3740 S3747 S.CrossRefGoogle ScholarPubMed
52.Sohn, Y-J, Stempack, JM, Reid, S et al. (2003) The effect of dietary folate on genomic and p53-specific DNA methylation in rat colon. Carcinogenesis 24, 8190.CrossRefGoogle ScholarPubMed
53.Duthie, SJ, Narayanan, S, Brand, GM et al. (2000) DNA stability and genomic methylation status in colonocytes isolated from methyl-donor-deficient rats. Eur J Nutr 39, 106111.CrossRefGoogle ScholarPubMed
54.Duthie, SJ (2010) Folate and cancer: how DNA damage, repair and methylation impact on colon carcinogenesis. J Inherit Metab Dis (Epublication ahead of print; DOI 10.1007/s10545-010-9128-0).Google ScholarPubMed
55.Jacob, RA, Gretz, DM, Taylor, PC et al. (1998) Moderate folate depletion increases plasma homocysteine and decreases lymphocyte DNA methylation in postmenopausal women. J Nutr 128, 12041212.CrossRefGoogle ScholarPubMed
56.Rampersaud, GC, Kauwell, GPA, Hutson, AD et al. (2000) Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr 72, 998–1003.CrossRefGoogle ScholarPubMed
57.Schernhammer, ES, Giovannucci, E, Kawasaki, T et al. . (2009) Dietary folate, alcohol and B vitamins in relation to LINE-1 hypomethylation in colon cancer. Gut(Epublication ahead of print, 14 October 2009).Google Scholar
58.Figueiredo, JC, Grau, MV, Wallace, K et al. (2009) Global DNA hypomethylation (LINE-1) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer Epidemiol Biomarkers Prev 18, 10411049.CrossRefGoogle ScholarPubMed
59.Fenech, M, Aitken, C & Rinaldi, J (1998) Folate, vitamin B12, homocysteine status and DNA damage in young Australian adults. Carcinogenesis 19, 11631171.CrossRefGoogle ScholarPubMed
60.Ingrosso, D, Cimmino, A, Perna, AF et al. (2003) Folate treatment and unbalanced methylation and changes of allelic expression induced by hyperhomocysteinemia in patients with uremia. Blood 361, 16931699.Google Scholar
61.Pufulete, M, Al-Ghnaniem, R, Khushal, A et al. (2005) Effect of folic acid supplementation on genomic DNA methylation in patients with colorectal adenoma. Gut 54, 648653.CrossRefGoogle ScholarPubMed
62.Cravo, M, Fidalgo, P, Pereira, AD, et al. . (1994) DNA methylation as an intermediate biomarker in colorectal cancer: modulation by folic acid supplementation. Eur J Cancer Prev 3, 473479.CrossRefGoogle ScholarPubMed
63.Cravo, ML, Pinto, AG, Chaves, P et al. . (1998) Effect of folate supplementation on DNA methylation of rectal mucosa in patients with colonic adenomas: correlation with nutrient intake. Clin Nutr 17, 4549.CrossRefGoogle ScholarPubMed
64.Basten, GP, Duthie, SJ, Pirie, LP et al. (2006) Sensitivity of markers of DNA stability and DNA repair activity to folate supplementation in healthy volunteers. Br J Cancer 94, 19421947.CrossRefGoogle ScholarPubMed
65.Hiltunen, MO, Turunen, MP, Hakkinen, TP et al. (2002) DNA hypomethylation and methyltransferase expression in atherosclerotic lesions. Vasc Med 7, 5–11.CrossRefGoogle ScholarPubMed
66.Lund, G, Andersson, L, Lauria, M et al. (2004) DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking Apolipoprotein E. J Biol Chem 279, 2914729154.CrossRefGoogle ScholarPubMed
67.Post, WS, Goldschmidt-Clermont, PJ, Wilhide, CC et al. (1999) Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res 43, 985991.CrossRefGoogle ScholarPubMed
68.Ying, AK, Hassanain, HH, Roos, CM et al. (2000) Methylation of the estrogen receptor-α gene promoter is selectively increased in proliferating human aoric smooth muscle cells. Cardiovasc Res 46, 172179.CrossRefGoogle Scholar
69.Hiltunen, MO & Yla-Herttuala, S (2003) DNA methylation, smooth muscle cells and atherogenesis. Arterioscler Thromb Vasc Biol 23, 17501753.CrossRefGoogle ScholarPubMed
70.Chen, Z, Karaplis, AC, Ackerman, SL et al. (2005) Mice deficient in methylenetetrahydrofolate reductase exhibit hyperhomocysteinemia and decreased methylation capacity within neuropathology and aortic lipid deposition. Hum Mol Genet 10, 433443.CrossRefGoogle Scholar
71.Devlin, AM, Bottiglieri, T, Domann, FE et al. (2005) Tissue-specific changes in H19 methylation and expression in mice with hyperhomocysteinemia. J Biol Chem 27, 2550625511.CrossRefGoogle Scholar
72.Yi, P, Melnyk, S, Pogribna, M et al. (2000) Increase in plasma homocysteine associated with parallel increase in plasma S-adenosyl homocysteine and lymphocyte DNA hypomethylation. J Biol Chem 275, 2931829323.CrossRefGoogle ScholarPubMed
73.Brown, KS, Huang, Y, Lu, Z-Y et al. (2006) Mild folate deficiency induces a proatherosclerotic phenotype in endothelial cells. Atherosclerosis 189, 133141.CrossRefGoogle ScholarPubMed
74.Hoffman, MA, Lalla, E, Lu, Y et al. (2001) Hyperhomocysteinemia enhances vascular inflammation and accelerates atherosclerosis in a murine model. J Clin Invest 107, 675683.CrossRefGoogle Scholar
75.Zhou, J, Moller, J, Danielsen, CC et al. (2001) Dietary supplementation with methionine and homocysteine promotes early atherosclerosis but not plaque rupture in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 21, 14701476.CrossRefGoogle Scholar
76.Zhou, J, Moller, J, Ritskes-Hoitinga, M et al. (2003) Effects of vitamin supplementation and hyperhomocysteinemia on atherosclerosis in ApoE-deficient mice. Atherosclerosis 168, 255262.CrossRefGoogle ScholarPubMed
77.Troen, AM, Lutgens, E, Smith, DE et al. (2003) The atherogenic effect of excess methionine intake. Proc Natl Acad Sci USA 100, 1508915094.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Epigenetics, diet and human disease. A simplified scheme illustrating the structure of mammalian chromatin and defining the role of epigenetic modifications both in normal human development and disease.

Figure 1

Fig. 2. Folate deficiency, cancer and CVD: a common mechanism? A simplified scheme describing how folate deficiency may alter normal DNA methylation and how this could impact both on risk of colon cancer and atherosclerosis.