Download Trans-HHS Workshop: Diet, DNA Methylation

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Genome evolution wikipedia , lookup

Genomic imprinting wikipedia , lookup

Human genome wikipedia , lookup

Mutation wikipedia , lookup

Comparative genomic hybridization wikipedia , lookup

DNA profiling wikipedia , lookup

Zinc finger nuclease wikipedia , lookup

SNP genotyping wikipedia , lookup

DNA repair wikipedia , lookup

Genetic engineering wikipedia , lookup

Replisome wikipedia , lookup

DNA polymerase wikipedia , lookup

Epigenetics of human development wikipedia , lookup

Epigenetics of neurodegenerative diseases wikipedia , lookup

Gene wikipedia , lookup

Mutagen wikipedia , lookup

Epigenetics of depression wikipedia , lookup

No-SCAR (Scarless Cas9 Assisted Recombineering) Genome Editing wikipedia , lookup

Polycomb Group Proteins and Cancer wikipedia , lookup

Gel electrophoresis of nucleic acids wikipedia , lookup

Genealogical DNA test wikipedia , lookup

Primary transcript wikipedia , lookup

Genomic library wikipedia , lookup

United Kingdom National DNA Database wikipedia , lookup

Nucleic acid analogue wikipedia , lookup

Genomics wikipedia , lookup

Nucleosome wikipedia , lookup

DNA damage theory of aging wikipedia , lookup

Behavioral epigenetics wikipedia , lookup

DNA vaccination wikipedia , lookup

Designer baby wikipedia , lookup

Point mutation wikipedia , lookup

Epigenetic clock wikipedia , lookup

Molecular cloning wikipedia , lookup

Genome editing wikipedia , lookup

Nucleic acid double helix wikipedia , lookup

Oncogenomics wikipedia , lookup

Epigenetics wikipedia , lookup

DNA supercoil wikipedia , lookup

Cre-Lox recombination wikipedia , lookup

Non-coding DNA wikipedia , lookup

Cell-free fetal DNA wikipedia , lookup

Deoxyribozyme wikipedia , lookup

Extrachromosomal DNA wikipedia , lookup

Microevolution wikipedia , lookup

Site-specific recombinase technology wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

History of genetic engineering wikipedia , lookup

DNA methylation wikipedia , lookup

Epigenetics in stem-cell differentiation wikipedia , lookup

Helitron (biology) wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Therapeutic gene modulation wikipedia , lookup

Cancer epigenetics wikipedia , lookup

Epigenetics of diabetes Type 2 wikipedia , lookup

Epigenetics in learning and memory wikipedia , lookup

Bisulfite sequencing wikipedia , lookup

Epigenomics wikipedia , lookup

Nutriepigenomics wikipedia , lookup

Transcript
Trans-HHS Workshop:
Diet, DNA Methylation Processes and Health
Gene-Nutrient Interactions and DNA Methylation1,2
Simonetta Friso3 and Sang-Woon Choi
Vitamin Metabolism Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research
Center on Aging, Tufts University, Boston, MA, 02111
KEY WORDS:
●
MTHFR
●
C677T
●
folate
●
DNA methylation
Nutrition research has recently highlighted the role of
several nutrients in regulating the genome machinery. A number of vitamins and micronutrients are substrates and/or cofactors in the metabolic pathways that regulate DNA synthesis
and/or repair and the expression of genes (1). It has been
documented that the deficiency of such nutrients may result in
the disruption of genomic integrity and alteration of DNA
●
gene expression
methylation, thus linking nutrition with modulation of gene
expression. The discovery of polymorphic enzymes involved in
critical steps of nucleic acids metabolic pathways contributed
to new insights into the interplay of genetics and nutrition for
the phenotypic expression of a defect. The response to a
nutrient status seems in many cases to be specific for each
genotype, and specific nutrient impairment results in different
gene expression, depending on each genotype. The field of
gene-nutrient interactions, therefore, seems to be a fascinating
model to explain the different response to environmental/diet
exposure at the molecular level.
Recently, the interaction between nutrients and DNA
methylation has been emphasized. DNA methylation, a characteristic feature of many eukaryotic genomes, consists in the
addition of a methyl group at the carbon 5⬘ position of cytosine within the cytosine-guanine (CpG)4 dinucleotide (2) in a
complex reaction that probably involves the flipping of the
cytosine base out of the intact double helix (3). Typically,
DNA methylation occurs in CpG-dinucleotide-rich regions,
1
Presented at the “Trans-HHS Workshop: Diet, DNA Methylation Processes
and Health” held August 6 – 8, 2001, in Bethesda, MD. This meeting was sponsored by the National Center for Toxicological Research, Food and Drug Administration; Center for Cancer Research, National Cancer Institute; Division of Cancer Prevention, National Cancer Institute; National Heart, Lung and Blood
Institute; National Institute of Child Health and Human Development; National
Institute of Diabetes and Digestive and Kidney Diseases; National Institute of
Environmental Health Sciences; Division of Nutrition Research Coordination,
National Institutes of Health; Office of Dietary Supplements, National Institutes of
Health; American Society for Nutritional Sciences; and the International Life
Sciences Institute of North America. Workshop proceedings are published as a
supplement to The Journal of Nutrition. Guest editors for the supplement were
Lionel A. Poirier, National Center for Toxicological Research, Food and Drug
Administration, Jefferson, AR and Sharon A. Ross, Nutritional Science Research
Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD.
2
This material is based upon work supported by the U. S. Department of
Agriculture, under agreement No. 581950-9-001. Any opinions, findings, conclusions or recommendations expressed in this publication are those of the authors
and do not necessarily reflect the view of the U. S. Department of Agriculture.
3
To whom correspondence should be addressed.
E-mail: [email protected].
4
Abbreviations used: C677T, cytosine-to-thymine transition at position 677;
CC, cytosine/cytosine; CpG, cytosine guanine dinucleotide; DNase, deoxyribonuclease; Dnmt1, DNA methyltransferase 1; methyl-THF, 5-methyltetrahydrofolate; MTHFR, methylenetetrahydrofolate reductase; RBC, red blood cell; SAM,
S-adenosylmethionine; TT, thymine/thymine.
0022-3166/02 $3.00 © 2002 American Society for Nutritional Sciences.
2382S
Downloaded from jn.nutrition.org by guest on June 15, 2012
ABSTRACT Many micronutrients and vitamins are critical for DNA synthesis/repair and maintenance of DNA
methylation patterns. Folate has been most extensively investigated in this regard because of its unique function
as methyl donor for nucleotide synthesis and biological methylation. Cell culture and animal and human studies
showed that deficiency of folate induces disruption of DNA as well as alterations in DNA methylation status. Animal
models of methyl deficiency demonstrated an even stronger cause-and-effect relationship than did studies using
a folate-deficient diet alone. Such observations imply that the adverse effects of inadequate folate status on DNA
metabolism are mostly due to the impairment of methyl supply. Recently, an interaction was observed between
folate status and a common mutation in the gene encoding for methylenetetrahydrofolate reductase, an essential
enzyme in one-carbon metabolism, in determining genomic DNA methylation. This finding suggests that the
interaction between a nutritional status with a genetic polymorphism can modulate gene expression through DNA
methylation, especially when such polymorphism limits the methyl supply. DNA methylation, both genome-wide
and gene-specific, is of particular interest for the study of cancer, aging and other conditions related to cell-cycle
regulation and tissue-specific differentiation, because it affects gene expression without permanent alterations in
DNA sequence such as mutations or allele deletions. Understanding the patterns of DNA methylation through the
interaction with nutrients is fundamental, not only to provide pathophysiological explanations for the development
of certain diseases, but also to improve the knowledge of possible prevention strategies by modifying a nutritional
status in at-risk populations. J. Nutr. 132: 2382S–2387S, 2002.
GENE-NUTRIENT INTERACTIONS AND DNA METHYLATION
DISCUSSION
Effect of nutrients on genomic integrity and DNA
methylation
Reduced dietary intake or low tissue/plasma levels of several
nutrients have been associated with higher risk for developing
cancer. Interestingly, many micronutrients and vitamins are
indispensable in DNA metabolic pathways (1,11).
Although most studies have been conducted in vitro and in
animal models and there is no clear evidence for the optimal
dietary ranges able to protect against DNA damage, roles in
maintaining genomic stability have been documented for several nutrients. For example, vitamin C and E deficiencies are
known to cause DNA oxidation and chromosome damage
(12,13). Vitamin D exerts an antioxidant activity, stabilizes
chromosomal structure and prevents DNA double strandbreaks (14). Magnesium is an essential cofactor in DNA
metabolism, and its role has been recognized in maintaining
high fidelity in DNA transcription (15). Iron may cause DNA
breaks (16). A carotenoid-rich diet has been shown to reduce
DNA damage (17). Vitamin B-12 deficiency is associated with
micronuclei formation (1,18), and reduced transcobalamin II
is associated with chromosomal abnormalities (19).
The roles of nutrients in DNA methylation, especially in
genome-wide methylation, have also been described. Zinc
deficiency can reduce the utilization of methyl groups from
S-adenosylmethionine (SAM) in rat liver and results in
genomic DNA hypomethylation as well as histone hypomethylation (20,21). Dietary deficiency in selenium decreased
genomic DNA methylation in Caco-2 cells and in the rat liver
and colon (22,23). Vitamin C deficiency has been associated
with DNA hypermethylation in lung cancer cells (24,25).
Interestingly, niacin, precursor of NAD⫹, is required to maintain the unmethylated state of CpG dinucleotides by inhibiting the enzymatic DNA methylation (26,27), because it is
necessary for the synthesis of poly-ADP–ribose polymerase-1,
which converts histone H1 to poly-ADP–ribosylated forms.
The poly-ADP–ribosylated forms of histone H1 are responsible
for the enzymatic inhibition of DNA methylation (26,27).
Nevertheless, folate and/or methyl group dietary supply
provides the most compelling data for the interaction of nutrients and DNA methylation, because these dietary elements
are directly involved in DNA methylation via one-carbon
metabolism. The sole metabolic function of all coenzymatic
forms of folate is to transfer one-carbon units. Within the
scope of this function is the synthesis of SAM, universal
methyl donor for several biological methylation reactions, and
the de novo deoxynucleoside triphosphate synthesis. Methionine is regenerated from homocysteine by methionine synthase in a reaction in which 5-methyltetrahydrofolate (methyl
THF) serves both as a cofactor and as a substrate. The reduced
availability of methyl-THF, the main circulating form of folate, decreases the biosynthesis of SAM, thus limiting the
availability of methyl groups for methylation reactions. Therefore, not only can dietary folate depletion decrease genomic
DNA methylation in both human (28,29) and animal models
(30), but, as described in the study by Rampersaud and colleagues, a folate replete diet also may restore the DNA methylation status (29).
Gene-nutrient interactions in one-carbon metabolism
Methylenetetrahydrofolate reductase (MTHFR; EC 1.5.1.20)
is considered a key enzyme in the one-carbon metabolism
because it catalyzes the irreversible conversion of 5,10-methylenetetrahydrofolate to methyl-THF (31). In 1988 Kang and
colleagues identified a variant of the MTHFR that causes
enzyme thermolability and reduced activity (32). The mutant
enzyme was associated with elevated plasma homocysteine
levels, which is to be expected because the conversion of
homocysteine to methionine is impaired (31). Not only did
the mild hyperhomocystinemia appear as an indicator of altered one-carbon metabolism, but the higher levels of this
sulfur-containing amino acid also were recognized to be an
independent risk factor for cardiovascular disease (33,34).
The thermolabile variant of the MTHFR is due to a common missense mutation, a cytosine-to-thymine transition at
base pair 677 (C677T) (35) that results in an alanine-tovaline substitution in the MTHFR amino acid sequence. The
prevalence of the valine-valine substitution is rather common,
with a frequency in homozygous persons of up to 20% in
certain populations (35,36,37). By determining plasma total
homocysteine levels, a strong gene-nutrient interaction was
demonstrated in the phenotypic expression of this polymorphism in MTHFR (37,38). Only those affected homozygous
persons with inadequate folate status, as indicated by blood
folate levels, showed elevated plasma homocysteine concentrations (38). An intermediate effect has also been observed in
heterozygous persons (37). These findings contributed to the
opening of a new field of interest for both nutrition and
genetics, especially because the relationship of the MTHFR
polymorphism with plasma folate levels was implicated as the
likely link between the C677T genetic defect and cardiovascular disease (34) as well as neural tube defects (39).
The MTHFR C677T polymorphism also provides a paradigm of gene-nutrient interaction in carcinogenesis (40,41).
The mutant thymine/thymine (TT) genotype is associated
with a lower risk of developing colorectal cancer; however,
this protective effect is observed only in persons with adequate
folate status. Among those persons with low systemic folate
status, the protection associated with the mutation is eliminated (42) and an even higher risk of developing colorectal
cancer is reported (43).
The biological significance of the MTHFR C677T mutation is predominantly related to the reduced availability of
methyl-THF. Also consistent with this concept is the recent
Downloaded from jn.nutrition.org by guest on June 15, 2012
the so-called “CpG islands” that, in contrast to the overall
genome, are highly represented in gene promoter regions or
initial exons of genes (4). DNA methylation is a fundamental
mechanism for the epigenetic control of gene expression and
the maintenance of genomic integrity (5,6). Therefore, an
evaluation of genomic DNA methylation status is important
for the study of cell growth regulation, tissue-specific differentiation (2,4,7) and carcinogenesis (6).
Most recently, an interaction was described between folate
status and a common mutation in a key enzyme of the onecarbon metabolism that is responsible for the availability of
methyl groups for biological methylation reactions, including
that of DNA (8,9). These findings suggest that modulating
DNA methylation by nutrition may be a fascinating new field
of gene and nutrient interaction. Therefore, it is of considerable interest to identify the factors that determine the patterns
of methylation, not only to provide evidence for the mechanisms of several pathological conditions, but also to identify
at-risk populations in which to conduct appropriate diet-based
interventions (10).
The purpose of this review is to discuss the most recent
knowledge about the effects of nutrients on gene expression
and integrity, with an emphasis on gene-nutrient interactions
in the modulation of DNA methylation.
2383S
2384S
SUPPLEMENT
observation that the distribution of different coenzymatic
forms of folate is altered in MTHFR TT homozygotes (44).
The red blood cells (RBC) of TT homozygous mutants show
variable amounts of formylated tetrahydrofolate polyglutamates at the expense of methylated tetrahydrofolates. In contrast, cells from the cytosine/cytosine (CC) wild-type persons
contain exclusively methylated tetrahydrofolate derivatives
(44).
A common mutation that results in an altered activity of
methionine synthase (5-methyltetrahydrofolate– homocysteine S-methyltransferase; EC 2.1.1.13), another important
enzyme of the homocysteine/methionine metabolic pathway,
has been described as interacting with levels of vitamin B-6
(45). This polymorphism, an adenine-to-guanine transition at
nucleotide position 2,756 which results in substitution of
glycine for aspartic acid at amino acid position 919, has been
observed to cause elevation of plasma total homocysteine
levels in persons with low levels of vitamin B-6 (45). However,
more evidence is needed to demonstrate a clear gene-nutrient
interaction in determining the biochemical expression of this
genotype.
A recent study investigated whether the mutant MTHFR,
in association with folate status, affected methylation of DNA.
(8). It was observed that subjects homozygous for the MTHFR
C677T polymorphism possessed a lower degree of genomic
DNA methylation in peripheral lymphocytes compared with
the CC wild-type persons (8) and also that there was an
inverse correlation between RBC folate and DNA methylation status. Because of the small number of subjects and the
indirect method used to assess genomic DNA methylation, a
large cohort of persons was subsequently studied using a newly
developed quantitative and highly specific liquid chromatography/electrospray ionization-mass spectrometry assay in
which the previous observations were reproduced and extended (9). The results showed that genomic DNA methylation in peripheral blood mononuclear cells directly correlated
with folate status and inversely correlated with plasma homocysteine levels. MTHFR TT genotypes had a diminished level
of DNA methylation compared with those with the CC wildtype. When analyzed according to folate status, however, only
the TT subjects with low levels of folate accounted for the
diminished DNA methylation (as shown in a model in Fig. 1).
Moreover, in TT subjects, DNA methylation status correlated
with the methylated proportion of RBC folate and was inversely related to the formylated proportion of RBC folates
that are known to be solely represented in TT persons (9).
These findings indicate that the MTHFR C677T polymorphism influences DNA methylation status through an interaction with folate status.
Nutrients and gene-specific DNA methylation
Gene-specific DNA methylation at the promoter region.
Approximately one-half of human genes have CpG islands in
their 5⬘-promoter regions or within their first exons (46). CpG
islands usually are unmethylated (47), and the methylation of
these CpG-rich sequences induces inhibition of their expression. The patterns of DNA methylation, therefore, distinguish
at a molecular level the genes to be expressed selectively (48).
Alterations in DNA methylation have been described as regulating a differentiation path in a particular tissue. DNA in
germ line cells usually is fully methylated, and demethylation
usually is observed in a tissue-specific fashion, except that most
of the housekeeping genes usually are maintained in a completely unmethylated state in both the germ line and in
tissue-specific sites (49).
Although the exact molecular mechanism by which DNA
methylation represses the transcription is not yet clear, data
demonstrating an active role of promoter methylation in gene
silencing are quite convincing. In vitro methylation of promoter-reporter constructs inhibits their subsequent expression
in transfected cells (50). Demethylation by 5-azadeoxycytidine, a DNA methyltransferase inhibitor, leads to re-expression of previously methylated genes (51). Homozygous embryos with a germline deletion of the DNA methyltransferase
1 (Dnmt1; EC 2.1.1.37) gene, on which the prototypical
mammalian cytosine DNA methyltransferase is encoded, reexpress a number of genes, including the normally silent alleles
of several imprinted genes and the abundant but normally
repressed endogenous retroviral sequences that are methylated
and silent in heterozygous littermates (52).
The mechanism for the CpG island-associated gene silencing seems to involve the link of specific methylated DNA
binding proteins, followed by the recruitment of a silencing
complex that includes histone deacetylases (53,54). The de
novo methylation, by itself, has a minimal effect on gene
expression. However, methylated DNA recruits methyl-binding proteins, which also attracts a protein complex that con-
Downloaded from jn.nutrition.org by guest on June 15, 2012
Effect of folate and MTHFR gene interaction in genomic
DNA methylation
FIGURE 1
Model illustrating the gene-nutrient interaction between the MTHFR C677T variant and folate levels for genomic DNA
methylation status. The enzyme MTHFR is responsible for the irreversible conversion of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate (CH3-THF). The MTHFR C677T variant encodes for a thermolabile enzyme with lower activity. As shown in this scheme, under folate
deficiency conditions, subjects carrying in homozygosity the mutant
allele for the thermolabile MTHFR (TT) (right) have a decreased formation of CH3-THF that results in lower production of S-adenosylmethionine and consequent lower availability of methyl groups (CH3) for the
methylation reactions including DNA methylation. The reduced availability of CH3-THF also is reflected by diminished homocysteine remethylation with consequent higher total plasma homocysteine levels
(tHcy). On the contrary, subjects wild-type for the C677T genotype (CC)
are not affected by folate deficiency (left), because the synthesis of
CH3-THF for methylation reactions and for the conversion of homocysteine to methionine is preserved. The MTHFR genotype does not alter
the availability of CH3-THF when folate status is adequate. The size of
the arrows indicates the different entity in enzyme activity and the flux
through the folate pools. The size of the font for the metabolic products
indicates approximately the relative change in the amount of metabolites among the MTHFR genotypes.
GENE-NUTRIENT INTERACTIONS AND DNA METHYLATION
2385S
FIGURE 2 Molecular effects of nutrients on gene
expression and integrity through modulation of DNA
methylation.
c-fos, c-Ha-ras and c-myc were correlated with hypomethylation at specific sites within these genes (61,62). These observations suggest that nutrients may affect gene transcription by
exon-specific DNA methylation.
Hypomethylation of the coding regions of critical genes can
lead to instability either because this region becomes more
susceptible to endogenous nucleases (63) or because the site of
hypomethylation is likely to undergo enzymatic deamination
to uracil (64,65). The latter situation is particularly prone to
occur in conditions where intracellular S-adenosylmethionine
levels are low, such as in folate depletion (65). In a folatedeficient rat model, Kim and colleagues observed that hypomethylation of hypermutable sites (exon 5 through 8) on the
p53 gene was related to increased DNA strand breaks in the
same region (66). Furthermore, hypomethylation within the
exon 8 of the colonic p53 gene was shown to be induced in an
animal model of chemical carcinogenesis (67), and hypomethylation of this site in peripheral mononuclear cell DNA
was highly associated with the development of lung cancer in
a nested case-control study (68). Cell culture studies indicate
that these foci of aberrant methylation may serve as initiators
of mutations (64) and may induce susceptibility to breakage of
the DNA backbone (69) by increased uracil insertion and
strand breaks (70). Collectively, these studies suggest that
folate deficiency might induce DNA strand breaks and subsequent mutations through exon site hypomethylation. On the
other hand, increasing levels of dietary folate effectively overrode the induction of hypomethylation in a dose-responsive
manner in an animal model of chemical carcinogenesis (67),
which suggests that folate supplementation can reduce gene
disruption by reversing the site-specific DNA hypomethylation.
This review discusses recent data in the field of genenutrient interactions and DNA methylation, a fundamental
epigenetic feature of DNA that affects gene expression and
genomic integrity (Fig. 2). Several nutrients are involved in
the maintenance of DNA metabolism, however most convincing data indicate a critical role for folate, an essential vitamin
for DNA metabolism because it is involved in both DNA
synthesis/repair and DNA methylation. The observation of an
interaction between a common mutation in MTHFR, a key
enzyme of the one-carbon metabolic pathway, and DNA
methylation provides the basis for research on the potential
role of nutrients in modulating an epigenetic feature of DNA
as well as in possible future prevention strategies.
LITERATURE CITED
1. Fenech, M. & Ferguson L. R. (2001) Vitamins/minerals and genomic
stability in humans. Mutat. Res. 475: 1– 6.
Downloaded from jn.nutrition.org by guest on June 15, 2012
tains histone deacetylases. Through the action of methylbinding proteins and histone deacetylases, the DNA structure
changes to a compact, condensed chromatin configuration
that results in permanent inhibition of messenger RNA and
protein production (55). This conformation makes the DNA
refractory to nuclease or restriction endonuclease digestion
and leads also to the loss of deoxyribonuclease (DNase)-Ihypersensitive sites. On the other hand, unmethylated CpG
islands possess a nuclease-sensitive chromatin structure that
differs from the bulk of the methylated genome (56). In
carcinogenesis, hypermethylation of CpG islands in promoter
region clearly is associated with transcriptional silencing of
gene expression, which has an important role as an alternative
mechanism by which tumor suppressor genes are inactivated
without mutation or allele deletion. On the other hand, hypomethylation of CpG islands is associated with the gene
activation, which also is an important mechanism by which
protooconcogenes are activated.
There are a few studies that suggest that certain nutrients
can affect gene expression by altering methylation of promoter
regions. In a rat model of hepatocellular carcinoma, a cholinedeficient diet induced hypomethylation of CpG sites of the
c-myc gene as well as overexpression of this gene (57). Jhaveri
and colleagues reported that the H-cadherin gene showed
hypermethylation of 5⬘sequences and downregulation of this
gene in response to folate depletion in human nasopharyngeal
carcinoma KB cells (58).
Gene-specific DNA methylation at the coding region. Because local cytosine methylation of a particular sequence can
directly interfere with the binding of certain transcription
factors (59), hypermethylation of the coding region can decrease the gene transcription. Conversely, hypomethylation of
the coding region also can increase the gene transcription by
enhancing the binding of transcription factors. Pogribny and
colleagues evaluated the gene-specific alterations of DNA
methylation during hepatocarcinogenesis with chronic dietary
methyl deficiency (60). The authors reported the progressive
loss of methyl groups at most CpG sites on both coding and
noncoding strands in hepatic DNA during the early phase of
folate/methyl deficiency. After tumor formation, the majority
of cytosines became remethylated. In the preneoplastic nodules, the level of p53 mRNA was increased and associated with
hypomethylation in the coding region, whereas in tumor tissue, p53 mRNA was decreased and was associated with relative hypermethylation. This observation suggests that a folate/
methyl-deficient diet induces liver cancer by affecting the
methylation status of the p53 gene coding region and by
consequent alteration of p53 gene transcription. In other
methyl-deficient animal studies, increased levels of mRNA for
2386S
SUPPLEMENT
R. (1995) A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat. Genet. 10: 111–113.
36. Ma, J., Stampfer, M. J., Hennekens, C. H., Frosst, P., Selhub, J., Horsford, J., Malinow, M. R., Willett, W. C. & Rozen, R. (1996) Methylenetetrahydrofolate reductase polymorphism, plasma folate, homocysteine, and risk of myocardial infarction in U. S. physicians. Circulation 94: 2410 –2416.
37. Girelli, D., Friso, S., Trabetti, E., Olivieri, O., Russo, C., Pessotto, R.,
Faccini, G., Pignatti, P. F., Mazzucco, A. & Corrocher, R. (1998) Methylenetetrahydrofolate reductase C677T mutation, plasma homocysteine, and folate in
subjects from northern Italy with or without angiographically documented severe
coronary atherosclerotic disease: evidence for an important genetic-environmental interaction. Blood 91: 4158 – 4163.
38. Jacques, P. F., Bostom, A. G., Williams, R. R., Ellison, R. C., Eckfeldt,
J. H., Rosenberg, I. H., Selhub, J. & Rozen, R. (1996) Relation between folate
status, a common mutation in methylenetetrahydrofolate reductase, and plasma
homocysteine concentrations. Circulation 93: 7–9.
39. van der Put, N. M., Steegers-Theunissen, R. P., Frosst, P., Trijbels, F. J.,
Eskes, T. K., van den Heuvel, L. P., Mariman, E. C., den Heyer, M., Rozen, R. &
Blom, H. J. (1995) Mutated methylenetetrahydrofolate reductase as a risk factor
for spina bifida. Lancet 346: 1070 –1071.
40. Kim, Y. I. (2000) Methylenetetrahydrofolate reductase polymorphisms,
folate, and cancer risk: a paradigm of gene-nutrient interactions in carcinogenesis. Nutr. Rev. 58: 205–209.
41. Choi, S. W. & Mason, J. B. (2000) Folate and carcinogenesis: an integrated scheme. J. Nutr. 130: 129 –132.
42. Ma, J., Stampfer, M. J., Giovannucci, E., Artigas, C., Hunter, D. J., Fuchs,
C., Willett, W. C., Selhub, J., Hennekens, C. H. & Rozen,
R. (1997) Methylenetetrahydrofolate reductase polymorphism, dietary interactions, and risk of colorectal cancer. Cancer Res. 57: 1098 –1102.
43. Ulrich, C. M., Kampman, E., Bigler, J., Schwartz, S. M., Chen, C., Bostick,
R., Fosdick, L., Beresford, S. A., Yasui, Y. & Potter, J. D. (2000) Lack of
association between the C677T MTHFR polymorphism and colorectal hyperplastic polyps. Cancer Epidemiol. Biomark. Prev. 9: 427– 433.
44. Bagley, P. J. & Selhub, J. (1998) A common mutation in the methylenetetrahydrofolate reductase gene is associated with an accumulation of formylated tetrahydrofolates in red blood cells. Proc. Natl. Acad. Sci. USA 95: 13217–
13220.
45. Harmon, D. L., Shields, D. C., Woodside, J. V., McMaster, D., Yarnell,
J. W., Young, I. S., Peng, K., Shane, B., Evans, A. E. & Whitehead,
A. S. (1999) Methionine synthase D919G polymorphism is a significant but
modest determinant of circulating homocysteine concentrations. Genet. Epidemiol. 17: 298 –309.
46. Bird, A. (1992) The essentials of DNA methylation. Cell 70: 5– 8.
47. Antequera, F. & Bird, A. (1993) Number of CpG islands and genes in
human and mouse. Proc. Natl. Acad. Sci. USA 90: 11995–11999.
48. Cedar, H. (1988) DNA methylation and gene activity. Cell 53: 3– 4.
49. Bird, A., Taggart, M., Frommer, M., Miller, O. J. & Macleod, D. (1985) A
fraction of the mouse genome that is derived from islands of nonmethylated,
CpG-rich DNA. Cell 40: 91–99.
50. Stein, R., Razin, A. & Cedar, H. (1982) In vitro methylation of the hamster adenine phosphoribosyltransferase gene inhibits its expression in mouse L
cells. Proc. Natl. Acad. Sci. USA 79: 3418 –3422.
51. Chen, Z. J. & Pikaard, C. S. (1997) Epigenetic silencing of RNA polymerase I transcription: a role for DNA methylation and histone modification in
nucleolar dominance. Genes Dev. 11: 2124 –2136.
52. Li, E., Bestor, T. H. & Jaenisch, R. (1992) Targeted mutation of the DNA
methyltransferase gene results in embryonic lethality. Cell 69: 915–926.
53. Jones, P. L., Veenstra, G. J., Wade, P. A., Vermaak,,D., Kass, S. U.,
Landsberger, N., Strouboulis, J. & Wolffe, A. P. (1998) Methylated DNA and
MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 19:
187–191.
54. Nan, X., Ng, H. H., Johnson, C. A., Laherty, C. D., Turner, B. M., Eisenman, R. N. & Bird, A. (1998) Transcriptional repression by the methyl-CpGbinding protein MeCP2 involves a histone deacetylase complex. Nature (Lond.)
393: 386 –389.
55. Santini, V., Kantarjian, H. M. & Issa, J. P. (2001) Changes in DNA
methylation in neoplasia: pathophysiology and therapeutic implications. Ann.
Intern. Med. 134: 573–586.
56. Tazi, J. & Bird, A. (1990) Alternative chromatin structure at CpG islands.
Cell 60: 909 –920.
57. Tsujiuchi, T., Tsutsumi, M., Sasaki, Y., Takahama, M. & Konishi,
Y. (1999) Hypomethylation of CpG sites and c-myc gene overexpression in
hepatocellular carcinomas, but not hyperplastic nodules, induced by a cholinedeficient L-amino acid-defined diet in rats. Jpn. J. Cancer Res. 90: 909 –913.
58. Jhaveri, M. S., Wagner, C. & Trepel, J. B. (2001) Impact of extracellular
folate levels on global gene expression. Mol. Pharmacol. 60: 1288 –1295.
59. Tate, P. H. & Bird, A. P. (1993) Effects of DNA methylation on DNAbinding proteins and gene expression. Curr. Opin. Genet. Dev. 3: 226 –231.
60. Pogribny, I. P., Basnakian, A. G., Miller, B. J., Lopatina, N. G., Poirier, L. A.
& James, S. J. (1995) Breaks in genomic DNA and within the p53 gene are
associated with hypomethylation in livers of folate/methyl-deficient rats. Cancer
Res. 55: 1894 –1901.
61. Zapisek, W. F., Cronin, G. M., Lyn-Cook, B. D. & Poirier,
L. A. (1992) The onset of oncogene hypomethylation in the livers of rats fed
methyl-deficient, amino acid-defined diets. Carcinogenesis 13: 1869 –1872.
62. Dizik, M., Christman, J. K. & Wainfan, E. (1991) Alterations in expres-
Downloaded from jn.nutrition.org by guest on June 15, 2012
2. Razin, A. & Riggs, A. D. (1980) DNA methylation and gene function.
Science (Wash., DC) 210: 604 – 610.
3. Klimasauskas, S., Kumar, S., Roberts, R. J. & Cheng, X. (1994) HhaI
methyltransferase flips its target base out of the DNA helix. Cell 76: 357–369.
4. Robertson, K. D. & Wolffe, A. P. (2000) DNA methylation in health and
disease. Nat. Rev. Genet. 1: 11–19.
5. Wolffe, A. P. & Matzke, M. A. (1999) Epigenetics: regulation through
repression. Science (Wash., DC) 286: 481– 486.
6. Jones, P. A. & Laird, P. W. (1999) Cancer epigenetics comes of age.
Nat. Genet. 21: 163–167.
7. Feinberg, A. P. (2001) Methylation meets genomics. Nat. Genet. 27:
9 –10.
8. Stern, L. L., Mason, J. B., Selhub, J. & Choi, S. W. (2000) Genomic DNA
hypomethylation, a characteristic of most cancers, is present in peripheral leukocytes of individuals who are homozygous for the C677T polymorphism in the
methylenetetrahydrofolate reductase gene. Cancer Epidemiol. Biomark. Prev. 9:
849 – 853.
9. Friso, S., Choi, S. W., Girelli, D., Mason, J. B., Dolnikowski, G. G., Bagley,
P. J., Olivieri, O., Jacques, P. F., Rosenberg, I. H., Corrocher, R. & Selhub,
J. (2002) A common mutation in the 5,10-methylenetetrahydrofolate reductase
gene affects genomic DNA methylation through an interaction with folate status.
Proc. Natl. Acad. Sci. USA 99: 5606 –5611.
10. Go, V. L., Wong, D. A. & Butrum, R. (2001) Diet, nutrition and cancer
prevention: where are we going from here? J. Nutr. 131: 3121S–3126S.
11. Ames, B. N. (2001) DNA damage from micronutrient deficiencies is
likely to be a major cause of cancer. Mutat. Res. 475: 7–20.
12. Halliwell, B. (2001) Vitamin C and genomic stability. Mutat. Res. 475:
29 –35.
13. Claycombe, K. J. & Meydani, S. N. (2001) Vitamin E and genome stability. Mutat. Res. 475: 37– 44.
14. Chatterjee, M. (2001) Vitamin D and genomic stability. Mutat. Res. 475:
69 – 87.
15. Hartwig, A. (2001) Role of magnesium in genomic stability. Mutat. Res.
475: 113–121.
16. De Freitas, J. M. & Meneghini, R. (2001) Iron and its sensitive balance
in the cell. Mutat. Res. 475: 153–159.
17. Collins, A. R. (2001) Carotenoids and genomic stability. Mutat. Res.
475: 21–28.
18. Fenech, M., Aitken, C. & Rinaldi, J. (1998) Folate, vitamin B12, homocysteine status and DNA damage in young Australian adults. Carcinogenesis 19:
1163–1171.
19. Rana, S. R., Colman, N., Goh, K. O., Herbert, V. & Klemperer,
M. R. (1983) Transcobalamin II deficiency associated with unusual bone marrow findings and chromosomal abnormalities. Am. J. Hematol. 14: 89 –96.
20. Wallwork, J. C. & Duerre, J. A. (1985) Effect of zinc deficiency on
methionine metabolism, methylation reactions and protein synthesis in isolated
perfused rat liver. J. Nutr. 115: 252–262.
21. Dreosti, I. E. (2001) Zinc and the gene. Mutat. Res. 475: 161–167.
22. Davis, C. D., Uthus, E. O. & Finley, J. W. (2000) Dietary selenium and
arsenic affect DNA methylation in vitro in Caco-2 cells and in vivo in rat liver and
colon. J. Nutr. 130: 2903–2909.
23. El-Bayoumy, K. (2001) The protective role of selenium on genetic damage and on cancer. Mutat. Res. 475: 123–139.
24. Piyathilake, C. J., Bell, W. C., Johanning, G. L., Cornwell, P. E., Heimburger, D. C. & Grizzle, W. E. (2000) The accumulation of ascorbic acid by
squamous cell carcinomas of the lung and larynx is associated with global
methylation of DNA. Cancer 89: 171–176.
25. Halliwell, B. (2001) Vitamin C and genomic stability. Mutat. Res. 475:
29 –35.
26. Zardo, G. & Caiafa, P. (1998) The unmethylated state of CpG islands in
mouse fibroblasts depends on the poly(ADP-ribosyl)ation process. J. Biol. Chem.
273: 16517–16520.
27. Hageman, G. J. & Stierum,R. H. (2001) Niacin, poly(ADP-ribose) polymerase-1 and genomic stability. Mutat. Res. 475: 45–56.
28. Jacob, R. A., Gretz, D. M., Taylor, P. C., James, S. J., Pogribny, I. P.,
Miller, B. J., Henning, S. M. & Swendseid, M. E. (1998) Moderate folate depletion increases plasma homocysteine and decreases lymphocyte DNA methylation
in postmenopausal women. J. Nutr. 128: 1204 –1212.
29. Rampersaud, G. C., Kauwell, G. P., Hutson, A. D., Cerda, J. J. & Bailey,
L. B. (2000) Genomic DNA methylation decreases in response to moderate
folate depletion in elderly women. Am. J. Clin. Nutr. 72: 998 –1003.
30. Balaghi, M. & Wagner, C. (1993) DNA methylation in folate deficiency:
use of CpG methylase. Biochem. Biophys. Res. Commun. 193: 1184 –1190.
31. Selhub, J. (1999) Homocysteine metabolism. Annu. Rev. Nutr. 19: 217–
246.
32. Kang, S. S., Zhou, J., Wong, P. W., Kowalisyn. J. & Strokosch,
G. (1988) Intermediate homocysteinemia: a thermolabile variant of methylenetetrahydrofolate reductase. Am. J. Hum. Genet. 43: 414 – 421.
33. Welch, G. N. & Loscalzo, J. (1998) Homocysteine and atherothrombosis. N. Engl. J. Med. 338: 1042–1050.
34. Ueland, P. M., Refsum, H., Beresford, S. A. & Vollset, S. E. (2000) The
controversy over homocysteine and cardiovascular risk. Am. J. Clin. Nutr. 72:
324 –332.
35. Frosst, P., Blom, H. J., Milos, R., Goyette, P., Sheppard, C. A., Matthews,
R. G., Boers, G. J., denHeijer, M., Kluijtmans, L. A., van den Heuvel, L. P. & Rozen,
GENE-NUTRIENT INTERACTIONS AND DNA METHYLATION
sion and methylation of specific genes in livers of rats fed a cancer promoting
methyl-deficient diet. Carcinogenesis 12: 1307–1312.
63. Hansen, R. S., Ellis, N. A. & Gartler, S. M. (1988) Demethylation of
specific sites in the 5⬘ region of the inactive X-linked human phosphoglycerate
kinase gene correlates with the appearance of nuclease sensitivity and gene
expression. Mol. Cell. Biol. 8: 4692– 4699.
64. Shen, J. C., Rideout, W. M., 3rd. & Jones, P. A. (1992) High frequency
mutagenesis by a DNA methyltransferase. Cell 71: 1073–1080.
65. Yang, A. S., Shen, J. C., Zingg, J. M., Mi, S. & Jones, P. A. (1995) HhaI
and HpaII DNA methyltransferases bind DNA mismatches, methylate uracil and
block DNA repair. Nucleic Acids Res. 23: 1380 –1387.
66. Kim, Y. I., Pogribny, I. P., Basnekiau, A. G., Miller, J. W., Sellers, J.,
James, S. J. & Mason, J. B. (1997) Folate deficiency in rats induces DNA strand
breaks and hypomethylation within the p53 tumor suppressor gene. Am. J. Clin.
Nutr. 65: 46 –52.
67. Kim, Y. I., Pogribny, I. P., Salomon, R. N., Choi, S. W., Smith, D. E.,
2387S
James, S. J. & Mason, J. B. (1996) Exon-specific DNA hypomethylation of the
p53 gene of rat colon induced by dimethylhydrazine. Modulation by dietary folate.
Am. J. Pathol. 149: 1129 –1137.
68. Woodson, K., Mason, J., Choi, S. W., Hartman, T., Tangrea, J., Virtamo,
J., Taylor, P. R. & Albanes, D. (2001) Hypomethylation of p53 in peripheral
blood DNA is associated with the development of lung cancer. Cancer Epidemiol.
Biomark. Prev. 10: 69 –74.
69. Pogribny, I. P., Basnakian, A. G., Miller, B. J., Lopatina, N. G., Poirier, L. A.
& James, S. J. (1995) Breaks in genomic DNA and within the p53 gene are
associated with hypomethylation in livers of folate/methyl-deficient rats. Cancer
Res. 55: 1894 –1901.
70. Blount, B. C., Mack, M. M., Wehr, C. M., MacGregor, J. T., Hiatt, R. A.,
Wang, G., Wickramasinghe, S. N., Everson, R. B. & Ames, B. N. (1997) Folate
deficiency causes uracil misincorporation into human DNA and chromosome
breakage: implications for cancer and neuronal damage. Proc. Natl. Acad. Sci.
USA 94: 3290 –3295.
Downloaded from jn.nutrition.org by guest on June 15, 2012