1,416
Views
36
CrossRef citations to date
0
Altmetric
Research Paper

Impact of folic acid fortification on global DNA methylation and one-carbon biomarkers in the Women's Health Initiative Observational Study cohort

, , , , , , , , , , , , , , & show all
Pages 396-403 | Received 08 Oct 2013, Accepted 22 Nov 2013, Published online: 03 Dec 2013

Abstract

DNA methylation is an epigenetic mechanism that regulates gene expression and can be modified by one-carbon nutrients. The objective of this study was to investigate the impact of folic acid (FA) fortification of the US food supply on leukocyte global DNA methylation and the relationship between DNA methylation, red blood cell (RBC) folate, and other one-carbon biomarkers among postmenopausal women enrolled in the Women's Health Initiative Observational Study. We selected 408 women from the highest and lowest tertiles of RBC folate distribution matching on age and timing of the baseline blood draw, which spanned the pre- (1994–1995), peri- (1996–1997), or post-fortification (1998) periods. Global DNA methylation was assessed by liquid chromatography-tandem mass spectrometry and expressed as a percentage of total cytosine. We observed an interaction (P = 0.02) between fortification period and RBC folate in relation to DNA methylation. Women with higher (vs. lower) RBC folate had higher mean DNA methylation (5.12 vs. 4.99%; P = 0.05) in the pre-fortification period, but lower (4.95 vs. 5.16%; P = 0.03) DNA methylation in the post-fortification period. We also observed significant correlations between one-carbon biomarkers and DNA methylation in the pre-fortification period, but not in the peri- or post-fortification period. The correlation between plasma homocysteine and DNA methylation was reversed from an inverse relationship during the pre-fortification period to a positive relationship during the post-fortification period. Our data suggest that (1) during FA fortification, higher RBC folate status is associated with a reduction in leukocyte global DNA methylation among postmenopausal women and; (2) the relationship between one-carbon biomarkers and global DNA methylation is dependent on folate availability.

Introduction

DNA methylation is an epigenetic modification of the genome, which influences gene expression and genome integrity.Citation1 DNA methylation can be modified by nutrients involved in one-carbon metabolism (e.g., folate, choline, vitamin B12, and vitamin B6), and disturbances in methylation reactions caused by abnormal status of these nutrients have been implicated in a number of human diseases including cancer.Citation2-Citation6

Folate, in the form of 5-methyltetrahydrofolate (5-methyl-THF), participates in cellular methylation reactions (including DNA methylation) by donating a methyl group for the vitamin B12-dependent re-methylation of homocysteine to methionine (Fig. S1). Folic acid (FA), the synthetic form of folate, can also participate in DNA methylation after its reduction to THF and conversion to 5-methyl-THF.Citation7 Homocysteine re-methylation to methionine can also proceed via a folate and B12-independent route in which betaine (a derivative of choline) serves as the methyl donor.Citation8

In January of 1998, the US Food and Drug Administration mandated FA fortification of enriched cereal-grain products (i.e., addition of 140 µg of FA/100 g of grain) in an effort to reduce the occurrence of neural tube defects, a mandate that some food companies initiated in 1996 and 1997.Citation9 FA fortification of the US food supply has led to significant increases in serum and red blood cell (RBC) folate concentrations as well as decreases in plasma total homocysteine;Citation10,Citation11 however, less is known about the impact of FA fortification on DNA methylation.

In this report, we investigated the association between mandatory FA fortification and leukocyte global DNA methylation, as well as the relationship between global DNA methylation, RBC folate, plasma choline, and other biomarkers of one-carbon metabolism among postmenopausal women enrolled in the Women's Health Initiative Observational Study (WHI-OS).

Results

Characteristics of the study population

The participants of this study were a subset of the control group from a nested case-control study investigating colorectal cancer risk in the WHI-OS.Citation12,Citation13 Baseline demographic and biochemical characteristics of the participants, which corresponded to FA fortification periods [pre (1994–1995), peri (1996–1997), or post (1998)], are shown in . BMI differed among FA fortification periods (P = 0.01) with a higher mean BMI (28.3 kg/m2) in the post-fortification period than in the pre- (26.5 kg/m2; P = 0.02) and peri-fortification (26.4 kg/m2; P = 0.01) periods. The ethnic distribution also differed among fortification periods (P = 0.03).

Table 1. Baseline characteristics of the study participants (n = 408) according to folic acid (FA) fortification period1-3

Plasma folate differed among FA fortification periods (P = 0.002) with higher median plasma folate (20 ng/mL) in the post-fortification period than in the pre- (14 ng/mL; P = 0.001) and peri-fortification (16 ng/mL; P = 0.002) periods. Similarly, RBC folate differed among FA fortification periods (P = 0.002) with higher median RBC folate in the peri- (572 ng/mL; P = 0.02) and post-fortification (726 ng/mL; P < 0.001) periods compared with the pre-fortification (424 ng/mL) period.

Effect of FA fortification period on baseline leukocyte global DNA methylation

Leukocyte global DNA methylation did not differ (P = 0.86, unadjusted; P = 0.38, multivariate-adjusted) among FA fortification periods (). However, we observed an interaction (P = 0.02) between fortification period and RBC folate status in relation to DNA methylation. Specifically, the highest (vs. lowest) RBC folate group had higher marginal mean DNA methylation (5.12 vs. 4.99%; P = 0.05) in the pre-fortification period, but lower DNA methylation (4.95 vs. 5.16%; P = 0.03) in the post-fortification period (). In addition, leukocyte global DNA methylation tended to differ (P = 0.08; multivariate-adjusted) among fortification periods (post < peri < pre) within the highest RBC folate group, but not within the lowest RBC folate group (P = 0.20; multivariate-adjusted) ().

Table 2. Baseline leukocyte global DNA methylation levels (%) according to folic acid (FA) fortification period1

Table 3. Baseline leukocyte global DNA methylation levels (%) within the lowest and highest RBC folate groups according to folic acid (FA) fortification period1,2

Correlations between baseline leukocyte global DNA methylation and one-carbon biomarkers according to FA fortification period

The univariate Spearman correlations between one-carbon biomarkers and leukocyte global DNA methylation are shown in . Prior to fortification, there were significant, but modest, positive associations of global DNA methylation with plasma folate (r = 0.20, P = 0.04) and RBC folate (r = 0.24, P = 0.01) as well as a borderline significant positive association with plasma vitamin B12 (r = 0.18, P = 0.06). Global DNA methylation was also inversely correlated with plasma methylmalonic acid (MMA; r = -0.26, P = 0.03), choline (r = -0.31, P = 0.002) and homocysteine (r = -0.26, P = 0.007). In the peri-fortification period, no significant relationships were observed between one-carbon biomarkers and global DNA methylation. Finally, in the post-fortification period, global DNA methylation was positively correlated with plasma homocysteine (r = 0.28, P = 0.02).

Table 4. Spearman rank correlation coefficients (r) between baseline leukocyte global DNA methylation and one-carbon biomarkers according to folic acid (FA) fortification period1

Main predictors of baseline leukocyte global DNA methylation according to FA fortification period

One-carbon biomarkers that predicted global DNA methylation were identified according to FA fortification period, testing them individually in multivariate-adjusted models (). Prior to fortification, RBC folate positively predicted global DNA methylation (β = 0.25, P = 0.02) explaining 5% of the residual variation (partial R2 = 0.05). Plasma vitamin B12 also tended to positively predict DNA methylation (β = 0.20, P = 0.08) explaining 3% of the residual variation (partial R2 = 0.03); however, plasma homocysteine (β = -21.23, P = 0.03), MMA (β = -1.18, P = 0.05), and choline (β = -57.82, P = 0.002) negatively predicted global DNA methylation explaining 4% (partial R2 = 0.04), 6% (partial R2 = 0.06) and 10% (partial R2 = 0.10) of the residual variation, respectively. In the peri-fortification period, no significant predictors of DNA methylation were detected. Finally, in the post-fortification period, plasma homocysteine tended to positively predict global DNA methylation (β = 29.37, P = 0.07) explaining 4% of the residual variation (partial R2 = 0.04). The overall R2 explained by one-carbon biomarkers, tested in an unadjusted linear regression model in which all variables were included simultaneously, was 0.12 in the pre- and peri-fortification periods and 0.19 in the post-fortification period (Table S1) with plasma choline, plasma dimethylglycine (DMG), and plasma homocysteine being the strongest predictors in each period, respectively (Table S2).

Table 5. Predictors of baseline leukocyte global DNA methylation according to folic acid (FA) fortification period1,2

Discussion

The present study investigated the association between mandatory FA fortification and leukocyte global DNA methylation, as well as the relationship between global DNA methylation, RBC folate, and other biomarkers of one-carbon metabolism in postmenopausal women. The following two main findings emerged: (1) FA fortification period and RBC folate status interacted to influence global DNA methylation and; (2) associations between one-carbon biomarkers and global DNA methylation differed between FA fortification periods.

FA fortification period interacted with RBC folate status to influence global DNA methylation

Previous studies have found that global DNA methylation can be altered by folate depletion or repletion in healthy adults.Citation14-Citation17 In postmenopausal women, global DNA methylation significantly decreased under folate depletionCitation14,Citation15 and increased upon folate repletion.Citation14 Based on these findings and the role of folate as a methyl donor, we anticipated that global DNA methylation would be higher among postmenopausal women with higher (vs. lower) RBC folate status. This expected result was observed in the pre-fortification period, but not in the post-fortification period during which women with higher (vs. lower) RBC folate status had lower DNA methylation.

Excess FA intake through fortified foods and supplements can lead to the accumulation of unmetabolized FA,Citation18 which may interfere with normal folate metabolismCitation19-Citation22 and lower global DNA methylation.Citation23 Although supraphysiologic folate status (i.e., total plasma folate concentrations > 19.8 ng/mL)Citation24 was observed among postmenopausal women with higher RBC folate in the post-fortification period (25.3 ng/mL; Table S3), it was similarly observed among women with higher RBC folate in the pre-fortification period (25.1 ng/mL) and appeared to be mostly attributable to FA supplement use in both periods. Specifically, a higher percentage of FA supplement users was observed among participants with higher (vs. lower) RBC concentrations (76% vs. 20%; Table S4), and higher RBC folate concentrations were observed among FA supplement users (vs. non-users) across fortification periods (Table S5). Nonetheless, a previous study conducted in the US reported the highest concentration of plasma unmetabolized FA in subjects exposed to both FA fortified foods and supplements as compared to those exposed only to FA fortified foods, or only to supplements.Citation25 Thus, it is possible that unmetabolized FA was elevated to a greater extent in the post- (vs. pre-) fortification period among women with higher RBC folate status. Measurements of unmetabolized FA in our cohort are needed to further explore this possibility, and additional studies are required to clarify the health outcomes, if any, of the inverse relationship between leukocyte global DNA methylation and high RBC folate in the era of FA fortification.

Associations between one-carbon biomarkers and global DNA methylation differed among FA fortification periods

Previous human studies have reported conflicting results with positiveCitation14,Citation16,Citation26 or noCitation27,Citation28 relationships between circulating folate (i.e., plasma and RBC folate) and global DNA methylation.Citation29 In the present study, plasma and RBC folate were positively correlated with DNA methylation in the pre-fortification period, but not in the peri- or post-fortification period. These findings suggest that the relationship between folate status and global DNA methylation is nonlinear and that folate status is likely to be a stronger predictor of global DNA methylation when folate availability is lower (i.e., prior to FA fortification). However, as alluded to above, it is possible that the differences in the relationship between circulating folate and DNA methylation across fortification periods arose from differences in the amounts of metabolized and unmetabolized folate. For example, metabolized folate present in the pre-fortification period may positively associate with global DNA methylation, while unmetabolized FA more likely to be present in the peri- and post-fortification periodsCitation24 may attenuate the positive relationship between folate status and global DNA methylation. Taken together, when total folate status (metabolized plus unmetabolized) is considered across the full spectrum from deficiency to very high, the overall association between folate and global DNA methylation may approximate a reverse U-shaped curve rather than a linear relationship.

Folate intake/status may also modify the relationship between DNA methylation and other nutrients involved in one-carbon metabolism. Indeed, biomarkers of vitamin B12 status (i.e., plasma vitamin B12 and MMA) were associated with leukocyte global DNA methylation in the pre-fortification period, but not in the peri- or post-fortification period. Both folate and vitamin B12 are required for the provision of methyl groups through the methionine synthase reaction (Fig. S1). However, folate is suggested to be a stronger determinant of biomarkers of the methylation cycle (e.g., plasma homocysteine) than vitamin B12,Citation30,Citation31 which may explain the lack of association between vitamin B12 status and global DNA methylation in the peri- and post-fortification periods.

The relationship between plasma choline and global DNA methylation was also modified by FA exposure with an inverse relationship observed in the pre-fortification period, but not in the peri- or post-fortification period. The inverse relationship between choline (a methyl donor) and DNA methylation in the pre-fortification period is unexpected and requires confirmation in other studies. However, when folate is less abundant (i.e., prior to FA fortification), supply of S-adenosylmethionine (SAM) for methylation reactions may be reduced thereby creating a competition among the various methyltransferases. As the affinity of DNA methyltransferase for SAM is ~18 times higher than phosphatidylethanolamine N-methyltransferase,Citation32 the enzyme that produces choline endogenously, SAM may be preferentially partitioned toward DNA methylation thus reducing endogenous choline production. In turn, this could lead to the inverse relationship observed in the pre-fortification period between DNA methylation and plasma choline.

Prior to FA fortification, we observed an inverse relationship between plasma homocysteine and global DNA methylation, which is consistent with previous reports.Citation26,Citation33 Interestingly, however, plasma homocysteine was positively correlated with DNA methylation in the post-fortification period. The divergent relationships between homocysteine and DNA methylation across fortification periods may arise from the fact that homocysteine is both a precursor and product of cellular methylation reactions. These data collectively suggest that the relationship between homocysteine and DNA methylation is dynamic and likely to be dependent on folate availability.

Strengths and limitations

The present study had several strengths including: (1) a unique opportunity to investigate the impact of mandatory FA fortification on global DNA methylation by stratifying into three fortification periods (pre-, peri-, and post-) and; (2) examination of a wide range of biomarkers involved in one-carbon metabolism as potential predictors of global DNA methylation according to FA fortification period. Several limitations should also be noted: (1) relatively small sample size; (2) potential for residual confounding by factors that were either not collected in the WHI-OS or not measured with sufficient precision and; (3) single measures of one-carbon biomarkers and global DNA methylation within each FA fortification period, which may not fully reflect the true complexity of DNA methylation reactions.

Conclusion

These data suggest that during FA fortification, higher RBC folate status is associated with a reduction in leukocyte global DNA methylation among postmenopausal women. If reductions in leukocyte global DNA methylation are shown to have adverse health outcomes in future studies, FA supplement use may not be advisable among postmenopausal women residing in the US or other countries with mandated FA fortification programs. The present study also suggests that FA intake via fortification modifies the relationship between one-carbon biomarkers and global DNA methylation, but potential biologic mechanisms need discerning.

Materials and Methods

Subjects and study design

The WHI-OS is a prospective cohort study that was established to investigate the predictors and causes of morbidity and mortality in postmenopausal women.Citation34,Citation35 The study enrolled 93 676 postmenopausal women, aged 50–79 y, at 40 clinical centers throughout the US between 1993 and 1998. These years of enrollment spanned the pre- (prior to January 1, 1996), peri- (1996–1997), and post- (after January 1, 1998) FA fortification periods in the US.Citation9 Women were excluded from the study if they had medical conditions with a predicted survival of less than 3 y; if they had adherence/retention issues (alcoholism, drug dependency, mental illness, or dementia); or if they were participating in another clinical trial. The study was approved by the human subject review boards at the Fred Hutchinson Cancer Research Center where the WHI Clinical Coordinating Center is located and at all 40 clinical centers. Written informed consent was obtained from all participants.Citation34,Citation35

In the present study, participants were a subset of those from a nested case-control study investigating colorectal cancer risk in the WHI-OS.Citation12,Citation13 From the controls of the study, we selected 408 women from the lowest (n = 202) and highest (n = 206) tertiles of baseline RBC folate concentrations, matching on age and timing of the baseline blood draw, which spanned the following FA fortification periods: pre-fortification (1994–1995; n = 71 low tertile, 51 high tertile), peri-fortification (1996–1997; n = 102 low tertile, 102 high tertile), and post-fortification (1998; n = 29 low tertile, 53 high tertile). The proportions in the fortification periods correspond approximately to the recruitment of the WHI-OS. However, because the nested case-control study from which we were sampling did not contain at least 50 participants in the post-fortification period, low RBC folate group, we selected additional participants from the pre-fortification, low RBC folate group in order to maintain approximately the same number of participants in each of the low and high RBC folate tertiles.

Data collection

Baseline demographic and health-related characteristics (i.e., age, race/ethnicity, education, smoking status, and physical activity) were collected using standardized questionnaires.Citation34 Height and weight were measured using a standardized protocol, and BMI was calculated as weight (kg)/height (m2). Dietary intake of folate, vitamin B6 and vitamin B12 was based on data derived from the WHI food-frequency questionnaire as previously described.Citation36 Supplemental vitamin intakes of B2, B6 and B12 were assessed by an inventory in which nutrients were recorded based on participants’ current dietary supplement bottles, which they brought to the clinic visits. To account for differences in bioavailability between synthetic FA and natural food folate, dietary folate equivalent (DFE) was used as the unit for total folate intake.Citation37

Analytic measurements

Blood samples were drawn at baseline after at least 12 h of fasting. Samples were kept at 4 °C for up to 1 h prior to centrifugation. Plasma and serum were collected and stored at −70 °C until analysis.Citation13 Leukocyte global DNA methylation was measured in de-identified samples using liquid chromatography-tandem mass spectrometry (LC-MS/MS) as described by Song et al.Citation38 with modifications based on our instrumentation.Citation39 A total of 11 batches (40 samples per batch) were run in duplicate. For each batch, all samples from both comparisons (i.e., high and low RBC folate) were randomly ordered and equally represented and matched on period of blood draw by pre-, peri- and post-fortification and on age. Both internal laboratory controls and 10% blind duplicate samples were used to determine assay precision and monitor assay performance. Internal laboratory controls included: (1) unmethylated lambda DNA (Promega); (2) methylated lambda DNA (~30% of DNA methylated); (3) four in-house human biological control samples and; (4) a negative control (water). All quality control samples were prepared in duplicate and interspersed among the samples. DNA methylation is expressed as a percentage of total cytosine: [methylated cytosine / (methylated + unmethylated cytosine)] × 100%.

Plasma concentrations of choline and its metabolites (i.e., betaine, DMG, trimethylamine N-oxide [TMAO]) were measured using stable isotope dilution LC-MS/MS methodology.Citation40 Plasma total homocysteine and cysteine were determined by high-pressure liquid chromatography (HPLC) with post-column fluorescence detectionCitation41; plasma and RBC folate as well as plasma vitamin B12 were measured by radioassay (SimulTRAC; MP Biomedicals); plasma pyridoxal-5′-phosphate (PLP) was analyzed by HPLC with fluorescence detectionCitation42; plasma MMA was measured by LC-MS/MSCitation43; plasma creatinine was quantified by the Jaffe rate reaction method (DxC Instrument; Beckman Coulter); and methylenetetrahydrofolate reductase (MTHFR) C677T genotype (rs 1801133) was determined by the Illumina 384-plex BeadXpress genotyping platform (Illumina Inc.).

Inter-assay coefficients of variance of the blind duplicate control samples for each of the assays were as follows: global DNA methylation, 5.5%; choline, 5.6%; betaine, 4.6%; DMG, 11.9%; TMAO, 5.8%; homocysteine, 6.5%; cysteine, 7.1%; RBC folate, 10.2%; plasma folate, 4.8%; vitamin B12, 6.2%; PLP, 5.9%; MMA, 15.0%; and creatinine, 4.1%.

Statistical analysis

Differences in baseline characteristics of the study population between FA fortification periods were analyzed by: (1) one-way analysis of variance (ANOVA) for normally distributed continuous variables; (2) non-parametric Kruskal-Wallis tests for non-normally distributed continuous variables or; (3) chi-square tests for categorical variables. Linear regression models were used to: (1) examine the influence of FA fortification and RBC folate (and their interaction term) on baseline leukcoyte global DNA methylation and; (2) identify the one-carbon biomarkers that predicted leukocyte global DNA methylation. The partial R2 for each predictor variable was determined to estimate the contribution of each predictor to the total variability in DNA methylation. Spearman rank correlation coefficients (r) were also computed to examine associations between leukocyte global DNA methylation and one-carbon biomarkers. In the multivariate-adjusted analyses, we controlled for age and BMI along with plasma creatinine, ethnicity (white/not white) and MTHFR C677T genotype as these three variables were shown to be influential in a univariate model assessing possible confounders on DNA methylation. MTHFR C677T genotype was treated as an additive variable (i.e., minor allele count) in our statistical models because of reduced variation; parameter estimates were not changed substantially when MTHFR C677T genotype was treated as a categorical variable. Because approximately 25% of plasma creatinine values were missing among the sample due to insufficient sample availability, simple mean imputation was used for the missing creatinine values. Model results using multiple imputation and simple mean imputation were similar. Significance was defined as P < 0.05, and all statistical tests were 2-sided. The data were analyzed by SAS version 9.3 (SAS Institute Inc.).

Abbreviations:
ANOVA =

Analysis of variance

BMI =

body mass index

DFE =

dietary folate equivalent

DMG =

dimethylglycine

FA =

folic acid

HPLC =

high-pressure liquid chromatography

LC-MS/MS =

liquid chromatography-tandem mass spectrometry

MMA =

methylmalonic acid

MTHFR =

methylenetetrahydrofolate reductase

PLP =

pyridoxal-5′-phosphate

RBC =

red blood cell

TMAO =

trimethylamine N-oxide

WHI-OS =

Women's Health Initiative Observational Study

5-methyl-THF =

5-methyltetrahydrofolate

Supplemental material

Additional material

Download Zip (236.4 KB)

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

The authors thank the study participants for making the program possible, and the WHI investigators and staff for their dedication. A full listing of WHI investigators can be found at: https://cleo.whi.org/researchers/Documents%20%20Write%20a%20Paper/WHI%20Investigator%20Short%20List.pdf. In addition, we would like to thank the research assistants and postdocs who have supported the WOMIn Study over the years, including Rachel Galbraith, Liz Poole, Clare Abbenhardt and Nina Habermann. The WHI program is funded by the National Heart, Lung, and Blood Institute, National Institutes of Health, US. Department of Health and Human Services through contracts HHSN268201100046C, HHSN268201100001C, HHSN268201100002C, HHSN268201100003C, HHSN268201100004C, and HHSN271201100004C.

Financial Support

Grants NIH R01 CA120523, N01 WH22110.

10.4161/epi.27323

References

  • McCabe DC, Caudill MA. DNA methylation, genomic silencing, and links to nutrition and cancer. Nutr Rev 2005; 63:183 - 95; http://dx.doi.org/10.1111/j.1753-4887.2005.tb00136.x; PMID: 16028562
  • Newberne PM, Rogers AE. Labile methyl groups and the promotion of cancer. Annu Rev Nutr 1986; 6:407 - 32; http://dx.doi.org/10.1146/annurev.nu.06.070186.002203; PMID: 2425831
  • Wainfan E, Dizik M, Stender M, Christman JK. Rapid appearance of hypomethylated DNA in livers of rats fed cancer-promoting, methyl-deficient diets. Cancer Res 1989; 49:4094 - 7; PMID: 2743304
  • Fang JY, Xiao SD, Zhu SS, Yuan JM, Qiu DK, Jiang SJ. Relationship of plasma folic acid and status of DNA methylation in human gastric cancer. J Gastroenterol 1997; 32:171 - 5; http://dx.doi.org/10.1007/BF02936363; PMID: 9085163
  • Davis CD, Uthus EO. DNA methylation, cancer susceptibility, and nutrient interactions. Exp Biol Med (Maywood) 2004; 229:988 - 95; PMID: 15522834
  • Kim YI. Folate and DNA methylation: a mechanistic link between folate deficiency and colorectal cancer?. Cancer Epidemiol Biomarkers Prev 2004; 13:511 - 9; PMID: 15066913
  • Wagner C. Biochemical role of folate in cellular metabolism. In: Bailey LB, editor. Folate in health and disease. New York: Marcel Dekker Inc.; 1995. p. 23–42.
  • Caudill MA. Folate and choline interrelationships: metabolic and potential health implications. In: Bailey LB, editor. Folate in health and disease. Boca Raton (FL): CRC Press; 2009. p. 449–65.
  • US Food and Drug Administration. Food standards: amendment of standards of identity for enriched grain products to require addition of folic acid. Final Rule. 21 CFR Parts 136, 137, and 139. Fed Regist 1996; 61:8781 - 97
  • Jacques PF, Selhub J, Bostom AG, Wilson PW, Rosenberg IH. The effect of folic acid fortification on plasma folate and total homocysteine concentrations. N Engl J Med 1999; 340:1449 - 54; http://dx.doi.org/10.1056/NEJM199905133401901; PMID: 10320382
  • Pfeiffer CM, Caudill SP, Gunter EW, Osterloh J, Sampson EJ. Biochemical indicators of B vitamin status in the US population after folic acid fortification: results from the National Health and Nutrition Examination Survey 1999-2000. Am J Clin Nutr 2005; 82:442 - 50; PMID: 16087991
  • Toriola AT, Cheng TY, Neuhouser ML, Wener MH, Zheng Y, Brown E, Miller JW, Song X, Beresford SA, Gunter MJ, et al. Biomarkers of inflammation are associated with colorectal cancer risk in women but are not suitable as early detection markers. Int J Cancer 2013; 132:2648 - 58; http://dx.doi.org/10.1002/ijc.27942; PMID: 23161620
  • Miller JW, Beresford SA, Neuhouser ML, Cheng TY, Song X, Brown EC, Zheng Y, Rodriguez B, Green R, Ulrich CM. Homocysteine, cysteine, and risk of incident colorectal cancer in the Women’s Health Initiative observational cohort. Am J Clin Nutr 2013; 97:827 - 34; http://dx.doi.org/10.3945/ajcn.112.049932; PMID: 23426034
  • Jacob RA, Gretz DM, Taylor PC, James SJ, Pogribny IP, Miller BJ, Henning SM, Swendseid ME. Moderate folate depletion increases plasma homocysteine and decreases lymphocyte DNA methylation in postmenopausal women. J Nutr 1998; 128:1204 - 12; PMID: 9649607
  • Rampersaud GC, Kauwell GP, Hutson AD, Cerda JJ, Bailey LB. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr 2000; 72:998 - 1003; PMID: 11010943
  • Shelnutt KP, Kauwell GP, Gregory JF 3rd, Maneval DR, Quinlivan EP, Theriaque DW, Henderson GN, Bailey LB. Methylenetetrahydrofolate reductase 677C-->T polymorphism affects DNA methylation in response to controlled folate intake in young women. J Nutr Biochem 2004; 15:554 - 60; http://dx.doi.org/10.1016/j.jnutbio.2004.04.003; PMID: 15350988
  • Axume J, Smith SS, Pogribny IP, Moriarty DJ, Caudill MA. The MTHFR 677TT genotype and folate intake interact to lower global leukocyte DNA methylation in young Mexican American women. Nutr Res 2007; 27:1365 - 1317; http://dx.doi.org/10.1016/j.nutres.2006.12.006; PMID: 18167510
  • Kelly P, McPartlin J, Goggins M, Weir DG, Scott JM. Unmetabolized folic acid in serum: acute studies in subjects consuming fortified food and supplements. Am J Clin Nutr 1997; 65:1790 - 5; PMID: 9174474
  • Troen AM, Mitchell B, Sorensen B, Wener MH, Johnston A, Wood B, Selhub J, McTiernan A, Yasui Y, Oral E, et al. Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr 2006; 136:189 - 94; PMID: 16365081
  • Ashokkumar B, Mohammed ZM, Vaziri ND, Said HM. Effect of folate oversupplementation on folate uptake by human intestinal and renal epithelial cells. Am J Clin Nutr 2007; 86:159 - 66; PMID: 17616776
  • Bailey SW, Ayling JE. The extremely slow and variable activity of dihydrofolate reductase in human liver and its implications for high folic acid intake. Proc Natl Acad Sci U S A 2009; 106:15424 - 9; http://dx.doi.org/10.1073/pnas.0902072106; PMID: 19706381
  • Sauer J, Mason JB, Choi SW. Too much folate: a risk factor for cancer and cardiovascular disease?. Curr Opin Clin Nutr Metab Care 2009; 12:30 - 6; http://dx.doi.org/10.1097/MCO.0b013e32831cec62; PMID: 19057184
  • Charles MA, Johnson IT, Belshaw NJ. Supra-physiological folic acid concentrations induce aberrant DNA methylation in normal human cells in vitro. Epigenetics 2012; 7:689 - 94; http://dx.doi.org/10.4161/epi.20461; PMID: 22617627
  • Pfeiffer CM, Johnson CL, Jain RB, Yetley EA, Picciano MF, Rader JI, Fisher KD, Mulinare J, Osterloh JD. Trends in blood folate and vitamin B-12 concentrations in the United States, 1988 2004. Am J Clin Nutr 2007; 86:718 - 27; PMID: 17823438
  • Kalmbach RD, Choumenkovitch SF, Troen AM, D’Agostino R, Jacques PF, Selhub J. Circulating folic acid in plasma: relation to folic acid fortification. Am J Clin Nutr 2008; 88:763 - 8; PMID: 18779294
  • Friso S, Choi SW, Girelli D, Mason JB, Dolnikowski GG, Bagley PJ, Olivieri O, Jacques PF, Rosenberg IH, Corrocher R, et al. A common mutation in the 5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation through an interaction with folate status. Proc Natl Acad Sci U S A 2002; 99:5606 - 11; http://dx.doi.org/10.1073/pnas.062066299; PMID: 11929966
  • Fenech M, Aitken C, Rinaldi J. Folate, vitamin B12, homocysteine status and DNA damage in young Australian adults. Carcinogenesis 1998; 19:1163 - 71; http://dx.doi.org/10.1093/carcin/19.7.1163; PMID: 9683174
  • Kok RM, Smith DE, Barto R, Spijkerman AM, Teerlink T, Gellekink HJ, Jakobs C, Smulders YM. Global DNA methylation measured by liquid chromatography-tandem mass spectrometry: analytical technique, reference values and determinants in healthy subjects. Clin Chem Lab Med 2007; 45:903 - 11; http://dx.doi.org/10.1515/CCLM.2007.137; PMID: 17617036
  • Crider KS, Yang TP, Berry RJ, Bailey LB. Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role. Adv Nutr 2012; 3:21 - 38; http://dx.doi.org/10.3945/an.111.000992; PMID: 22332098
  • Jacques PF, Bostom AG, Wilson PW, Rich S, Rosenberg IH, Selhub J. Determinants of plasma total homocysteine concentration in the Framingham Offspring cohort. Am J Clin Nutr 2001; 73:613 - 21; PMID: 11237940
  • Homocysteine Lowering Trialists’ Collaboration. Dose-dependent effects of folic acid on blood concentrations of homocysteine: a meta-analysis of the randomized trials. Am J Clin Nutr 2005; 82:806 - 12; PMID: 16210710
  • Clarke S, Banfield K. Can elevated plasma homocysteine levels result in the inhibition of intracellular methyltransferases? In: Milstien S, Kapatos G, Levine RA, Sharne B, editors. Chemistry and Biology of Pteridines and Folates. Dordrecht (Netherlands): Kluwer Academic Press; 2002. p. 557–62.
  • Yi P, Melnyk S, Pogribna M, Pogribny IP, Hine RJ, James SJ. Increase in plasma homocysteine associated with parallel increases in plasma S-adenosylhomocysteine and lymphocyte DNA hypomethylation. J Biol Chem 2000; 275:29318 - 23; http://dx.doi.org/10.1074/jbc.M002725200; PMID: 10884384
  • The Women’s Health Initiative Study Group. Design of the Women’s Health Initiative clinical trial and observational study. Control Clin Trials 1998; 19:61 - 109; http://dx.doi.org/10.1016/S0197-2456(97)00078-0; PMID: 9492970
  • Langer RD, White E, Lewis CE, Kotchen JM, Hendrix SL, Trevisan M. The Women’s Health Initiative Observational Study: baseline characteristics of participants and reliability of baseline measures. Ann Epidemiol 2003; 13:Suppl S107 - 21; http://dx.doi.org/10.1016/S1047-2797(03)00047-4; PMID: 14575943
  • Zschäbitz S, Cheng TY, Neuhouser ML, Zheng Y, Ray RM, Miller JW, Song X, Maneval DR, Beresford SA, Lane D, et al. B vitamin intakes and incidence of colorectal cancer: results from the Women’s Health Initiative Observational Study cohort. Am J Clin Nutr 2013; 97:332 - 43; http://dx.doi.org/10.3945/ajcn.112.034736; PMID: 23255571
  • Institute of Medicine. Dietary Reference Intakes: thiamin, riboflavin, niacin, vitamin B6, folate, vitamin B12, pantothenic Acid, biotin, and choline. Washington, DC: National Academy Press, 1998.
  • Song L, James SR, Kazim L, Karpf AR. Specific method for the determination of genomic DNA methylation by liquid chromatography-electrospray ionization tandem mass spectrometry. Anal Chem 2005; 77:504 - 10; http://dx.doi.org/10.1021/ac0489420; PMID: 15649046
  • Shin W, Yan J, Abratte CM, Vermeylen F, Caudill MA. Choline intake exceeding current dietary recommendations preserves markers of cellular methylation in a genetic subgroup of folate-compromised men. J Nutr 2010; 140:975 - 80; http://dx.doi.org/10.3945/jn.110.121186; PMID: 20220206
  • Yan J, Jiang X, West AA, Perry CA, Malysheva OV, Devapatla S, Pressman E, Vermeylen F, Stabler SP, Allen RH, et al. Maternal choline intake modulates maternal and fetal biomarkers of choline metabolism in humans. Am J Clin Nutr 2012; 95:1060 - 71; http://dx.doi.org/10.3945/ajcn.111.022772; PMID: 22418088
  • Gilfix BM, Blank DW, Rosenblatt DS. Novel reductant for determination of total plasma homocysteine. Clin Chem 1997; 43:687 - 8; PMID: 9105275
  • Talwar D, Quasim T, McMillan DC, Kinsella J, Williamson C, O’Reilly DS. Optimisation and validation of a sensitive high-performance liquid chromatography assay for routine measurement of pyridoxal 5-phosphate in human plasma and red cells using pre-column semicarbazide derivatisation. J Chromatogr B Analyt Technol Biomed Life Sci 2003; 792:333 - 43; http://dx.doi.org/10.1016/S1570-0232(03)00320-9; PMID: 12860041
  • Pedersen TL, Keyes WR, Shahab-Ferdows S, Allen LH, Newman JW. Methylmalonic acid quantification in low serum volumes by UPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879:1502 - 6; http://dx.doi.org/10.1016/j.jchromb.2011.03.039; PMID: 21497144

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.