Español | 日本語

Summary

  • Folate is a generic term referring to both natural folates in food and folic acid, the synthetic form used in supplements and fortified food. Folate is critical in the metabolism of nucleic acid precursors and several amino acids, as well as in methylation reactions. (More information) 
  • Severe deficiency in either folate or vitamin B12 can lead to megaloblastic anemia, which causes fatigue, weakness, and shortness of breath. Improper treatment of vitamin B12-dependent megaloblastic anemia with high-dose supplemental folic acid can potentially delay the diagnosis of vitamin B12 deficiency and thus leave the individual at risk of developing irreversible brain and nervous system damage. (More information)
  • Folate status is influenced by the presence of genetic variations in folate metabolism enzymes, particularly polymorphisms of the 5,10-methylenetetrahydrofolate reductase (MTHFR) gene. (More information) 
  • Inadequate folate status during early pregnancy increases the risk of congenital anomalies. The introduction of mandatory folic acid fortification of refined grain products in the US in 1998 has reduced the prevalence of neural tube defects (NTDs) in newborns. Yet, folate status is considered inadequate in a majority of women of childbearing age worldwide. Moreover, genetic factors might modify the risk of NTDs by increasing the susceptibility to folate deficiency during pregnancy. Several studies have investigated the role of folic acid supplementation in the prevention of congenital anomalies other than NTDs. (More information) 
  • Folate deficiency and elevated concentrations of homocysteine in the blood are associated with increased risk of cardiovascular disease. Although folic acid supplementation has been proven effective to control circulating homocysteine concentrations, homocysteine lowering has not affected the incidence of cardiovascular disease in supplementation trials. Yet, supplementation with folic acid and other B vitamins does appear to lower the risk of stroke. (More information) 
  • Low folate status has been linked to increased cancer risk. However, intervention trials with high doses of folic acid have not generally shown any benefit on cancer incidence. There is some concern that high doses of supplemental folic acid might increase the risk of certain cancers, but further study is needed. (More information) 
  • Prospective cohort studies have reported an inverse association between folate status and colorectal cancer risk, especially among men. However, the relationship between folate status and cancer risk is complex and requires further research. (More information) 
  • Folate is essential for brain development and function. Low folate status and/or high homocysteine concentrations are associated with cognitive dysfunction in aging (from mild impairments to dementia). However, whether supplemental folic acid confers long-term benefits in maintaining cognitive health is not yet known. (More information) 
  • Several autosomal recessive disorders affecting folate transport and metabolism can be treated with high doses of folinic acid, a folate derivative. (More information) 
     

Folate is a water-soluble B-vitamin, which is also known as vitamin B9 or folacin. Naturally occurring folates exist in many chemical forms; folates are found in food, as well as in metabolically active forms in the human body. Folic acid is the major synthetic form found in fortified foods and vitamin supplements. Other synthetic forms include folinic acid (Figure 1) and levomefolic acid. Folic acid has no biological activity unless converted into folates (1). In the following discussion, forms found in food or the body are referred to as "folates," while the form found in supplements or fortified food is referred to as "folic acid."

Folate Figure 1. Chemical Structures. Chemical structures of folic acid (C19H19N7O6), 5-methyltetrahydrofolate (C20H25N7O6), and folinic acid (C20H23N7O7)

Function

One-carbon metabolism

The only function of folate coenzymes in the body appears to be in mediating the transfer of one-carbon units (2). Folate coenzymes act as acceptors and donors of one-carbon units in a variety of reactions critical to the metabolism of nucleic acids and amino acids (Figure 2) (3).

Folate Figure 2. Overview of One-carbon Metabolism. 5,10-methylenetetrahydrofolate is required for the synthesis of nucleic acids, and 5-methyltetrahydrofolate is required for the formation of methionine from homocysteine. Methionine, in the form of methyl donor S-adenosylmethionine (SAM), is essential to many biological methylation reactions, including DNA methylation. Methylenetetrahydrofolate reductase (MTHFR) is a riboflavin (FAD)-dependent enzyme that catalyzes the reduction of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate.

Nucleic acid metabolism

Folate coenzymes play a vital role in DNA metabolism through two different pathways: (1) The synthesis of DNA from its precursors (thymidine and purines) is dependent on folate coenzymes. (2) A folate coenzyme is required for the synthesis of methionine from homocysteine, and methionine is required for the synthesis of S-adenosylmethionine (SAM). SAM is a methyl group (one-carbon unit) donor used in most biological methylation reactions, including the methylation of a number of sites within DNA, RNA, proteins, and phospholipids. The methylation of DNA plays a role in controlling gene expression and is critical during cell differentiation. Aberrations in DNA methylation have been linked to the development of cancer (see Cancer).

Amino acid metabolism

Folate coenzymes are required for the metabolism of several important amino acids, namely methionine, cysteine, serine, glycine, and histidine. The synthesis of methionine from homocysteine is catalyzed by methionine synthase, an enzyme that requires not only folate (as 5-methyltetrahydrofolate) but also vitamin B12. Thus, folate (and/or vitamin B12) deficiency can result in decreased synthesis of methionine and an accumulation of homocysteine. Elevated blood concentrations of homocysteine have been considered for many years to be a risk factor for some chronic diseases, including cardiovascular disease and dementia (see Disease Prevention).

Nutrient interactions

Vitamin B12 and vitamin B6

The metabolism of homocysteine, an intermediate in the metabolism of sulfur-containing amino acids, provides an example of the interrelationships among nutrients necessary for optimal physiological function and health. Healthy individuals utilize two different pathways to metabolize homocysteine (Figure 3). One pathway (methionine synthase) synthesizes methionine from homocysteine and is dependent on both folate and vitamin B12 as cofactors. The other pathway converts homocysteine to another amino acid, cysteine, and requires two vitamin B6-dependent enzymes. Thus, the concentration of homocysteine in the blood is regulated by three B-vitamins: folate, vitamin B12, and vitamin B6 (4). In some individuals, riboflavin (vitamin B2) is also involved in the regulation of homocysteine concentrations (see the article on Riboflavin). 

Folate Figure 3. Homocysteine Metabolism. Homocysteine is methylated to form the essential amino acid methionine in two pathways. The reaction of homocysteine remethylation catalyzed by the vitamin B<sub>12</sub>-dependent methionine synthase captures a methyl group from the folate-dependent one-carbon pool (5-methyltetrahydrofolate). A second pathway requires betaine (N,N,N-trimethylglycine) as a methyl donor for the methylation of homocysteine catalyzed by betaine homocysteine methyltransferase. The catabolic pathway of homocysteine, known as the transsulfuration pathway, converts homocysteine to the amino acid cysteine via two vitamin B<sub>6</sub> (PLP)-dependent enzymes. Cystathionine beta synthase catalyzes the condensation of homocysteine with serine to form cystathionine, and cystathionine is then converted to cysteine, alpha-ketobutyrate, and ammonia by cystathionine gamma lyase.

Riboflavin

Although less well recognized, folate has an important metabolic interaction with riboflavin. Riboflavin is a precursor of flavin adenine dinucleotide (FAD), a coenzyme required for the activity of the folate-metabolizing enzyme, 5,10-methylenetetrahydrofolate reductase (MTHFR). FAD-dependent MTHFR in turn catalyzes the reaction that generates 5-methyltetrahydrofolate (see Figure 2). This active form of folate is required to form methionine from homocysteine. Along with other B-vitamins, higher riboflavin intakes have been associated with decreased plasma homocysteine concentrations (5). The effects of riboflavin on folate metabolism appear to be greatest in individuals homozygous for the common c.677C>T polymorphism (i.e., TT genotype) in the MTHFR gene (see Genetic variation in folate requirements) (6). These individuals (up to 32% of the population) typically present with low folate status, along with elevated homocysteine concentrations, particularly when folate and/or riboflavin intake is suboptimal. The elevated homocysteine concentration in these individuals, however, is highly responsive to lowering with riboflavin supplementation. In addition, riboflavin supplementation has been shown to reduce blood pressure in hypertensive individuals homozygous for the MTHFR c.677C>T polymorphism (7, 8), confirming the importance of the riboflavin-MTHFR interaction (9, 10).  

Bioavailability

Dietary folates exist predominantly in the polyglutamyl form (containing several glutamate residues), whereas folic acid — the synthetic vitamin form — is a monoglutamate, containing just one glutamate moiety. In addition, natural folates are reduced molecules, whereas folic acid is fully oxidized. These chemical differences have major implications for the bioavailability of the vitamin such that folic acid is considerably more bioavailable than naturally occurring food folates at equivalent intake levels. 

The intestinal absorption of dietary folates is a two-step process that involves the hydrolysis of folate polyglutamates to the corresponding monoglutamyl derivatives, followed by their transport into intestinal cells (11). There, folic acid is converted into a naturally occurring folate, namely 5-methyltetrahydrofolate, which is the major circulating form of folate in the human body (see Figure 1).

The bioavailability of naturally occurring folates is inherently limited and variable. There is much variability in the ease with which folates are released from different food matrices, and the polyglutamyl "tail" is removed (de-conjugation) before uptake by intestinal cells. Also, other dietary constituents can contribute to instability of labile folates during the processes of digestion. As a result, naturally occurring folates show incomplete bioavailability compared with folic acid. The bioavailability of folic acid, in contrast, is assumed to be 100% when ingested as a supplement, while folic acid in fortified food is estimated to have about 85% the bioavailability of supplemental folic acid.

Of note, folate recommendations in the US and certain other countries are now expressed as Dietary Folate Equivalents (DFEs), a calculation that was devised to take into account the greater bioavailability of folic acid compared to naturally occurring dietary folates (see The Recommended Dietary Allowance).

Transport

Folate and its coenzymes require transporters to cross cell membranes. Folate transporters include the reduced folate carrier (RFC), the proton-coupled folate transporter (PCFT), and the folate receptor proteins, FRα and FRβ. Folate homeostasis is supported by the ubiquitous distribution of folate transporters, although abundance and importance vary among tissues (12). PCFT plays a major role in folate intestinal transport since mutations affecting the gene encoding PCFT cause hereditary folate malabsorption. Defective PCFT also leads to impaired folate transport into the brain (see Disease Treatment). FRα and RFC are also critical for folate transport across the blood-brain barrier when extracellular folate is either low or high, respectively. Folate is essential for the proper development of the embryo and the fetus. The placenta is known to concentrate folate to the fetal circulation, leading to higher folate concentrations in the fetus compared to those found in the pregnant woman. All three types of receptors have been associated with folate transport across the placenta during pregnancy (13).

Deficiency

Causes

Although folate deficiency is uncommon in the United States, it is estimated to be more prevalent worldwide (11). Most often caused by a dietary insufficiency, folate deficiency can also occur in a number of other situations. Chronic, heavy alcohol consumption is associated with diminished absorption of folate (in addition to low dietary intake), which can lead to folate deficiency (14). Smoking is also associated with low folate status. In one study, folate concentrations in blood were about 15% lower in smokers compared to nonsmokers (15). Additionally, impaired folate transport to the fetus has been described in pregnant women who either smoked or abused alcohol during their pregnancy (16, 17).

Pregnancy is a time when the folate requirement is greatly increased to sustain the demand for rapid cell replication and growth of fetal, placental, and maternal tissue. Conditions such as cancer or inflammation can also result in increased rates of cell division and metabolism, causing an increase in the body's demand for folate (18). Moreover, folate deficiency can result from some malabsorptive conditions, including inflammatory bowel diseases (Crohn’s disease and ulcerative colitis) and celiac disease (11, 19). Several medications can contribute to folate deficiency (see Drug interactions). Finally, a number of genetic diseases affecting folate absorption, transport, or metabolism can cause folate deficiency or impede its metabolic functions (see Disease Treatment).

Symptoms

Clinical folate deficiency leads to megaloblastic anemia, which is reversible with folic acid treatment. Rapidly dividing cells like those derived from bone marrow are most vulnerable to the effects of folate deficiency since DNA synthesis and cell division are dependent on folate coenzymes. When folate supply to the rapidly dividing cells of the bone marrow is inadequate, blood cell division is reduced, resulting in fewer but larger red blood cells. This type of anemia is called megaloblastic or macrocytic anemia, referring to the enlarged, immature red blood cells. Neutrophils, a type of white blood cell, become hypersegmented, a change that can be found by examining a blood sample microscopically. Because normal red blood cells have a lifetime in the circulation of approximately four months, it can take months for folate-deficient individuals to develop the characteristic megaloblastic anemia. Progression of such an anemia leads to a decreased oxygen carrying capacity of the blood and may ultimately result in symptoms of fatigue, weakness, and shortness of breath (1). It is important to point out that megaloblastic anemia resulting from folate deficiency is identical to the megaloblastic anemia resulting from vitamin B12 deficiency, and further clinical testing is required to diagnose the true cause of megaloblastic anemia (see Toxicity).

Individuals in the early stages of folate deficiency may not show obvious symptoms, but blood concentrations of homocysteine may increase (see Disease Prevention). Yet, the concentration of circulating homocysteine is not a specific indicator of folate status, as elevated homocysteine can be the result of vitamin B12 and other B-vitamin deficiencies, lifestyle factors, and renal insufficiency. Subclinical deficiency is typically detected by measurement of folate concentrations in serum/plasma or in red blood cells.

The Recommended Dietary Allowance (RDA)

Determination of the RDA

Traditionally, the dietary folate requirement was defined as the amount needed to prevent a deficiency severe enough to cause symptoms like anemia. The most recent RDA (1998; Table 1) was based primarily on the adequacy of red blood cell folate concentrations at different levels of folate intake, as judged by the absence of abnormal hematological indicators. Red cell folate has been shown to correlate with liver folate stores and is used as an indicator of long-term folate status. Plasma or serum folate reflects recent folate intake and is not a reliable biomarker for folate status, especially when used as a one-time assessment (11). Maintenance of normal blood homocysteine concentrations, an indicator of one-carbon metabolism, was considered only as an ancillary indicator of adequate folate intake.

Because pregnancy is associated with a significant increase in cell division and other metabolic processes that require folate coenzymes, the RDA for pregnant women is considerably higher than for women who are not pregnant (3). However, the prevention of neural tube defects (NTDs) was not considered when setting the RDA for pregnant women. Rather, reducing the risk of NTDs was considered in a separate recommendation for women capable of becoming pregnant (see Disease Prevention), because the crucial events in the development of the neural tube occur before many women are aware that they are pregnant (20).

Dietary Folate Equivalents (DFEs)

When the Food and Nutrition Board of the US Institute of Medicine (now the National Academy of Medicine) last revised the dietary recommendations for folate, they introduced a new unit, the Dietary Folate Equivalent (DFE) (1). Use of the DFE reflects the higher bioavailability of synthetic folic acid found in supplements and fortified food compared to that of naturally occurring food folates (20).

  • 1 microgram (μg) of food folate provides 1 μg of DFEs
  • 1 μg of folic acid taken with meals or as fortified food provides 1.7 μg of DFEs
  • 1 μg of folic acid (supplement) taken on an empty stomach provides 2 μg of DFEs

For example, a serving of food containing 60 μg of folate would provide 60 μg of DFEs, while a serving of pasta fortified with 60 μg of folic acid would provide 1.7 x 60 = 102 μg of DFEs due to the higher bioavailability of folic acid. A folic acid supplement of 400 μg taken on an empty stomach would provide 800 μg of DFEs. It should be noted that DFEs were determined in studies with adults and whether folic acid in infant formula is more bioavailable than folates in mother's milk has not been studied. Use of DFEs to determine a folate requirement for the infant would not be desirable.

Table 1. Recommended Dietary Allowance for Folate in Dietary Folate Equivalents (DFEs)
Life Stage Age Males (μg/day) Females (μg/day)
Infants 0-6 months 65 (AI) 65 (AI)
Infants 7-12 months 80 (AI) 80 (AI)
Children 1-3 years 150 150
Children 4-8 years 200 200
Children 9-13 years 300 300
Adolescents 14-18 years 400 400
Adults 19 years and older 400 400
Pregnancy all ages - 600
Breast-feeding all ages - 500

Genetic variation in folate requirements

A common polymorphism or variation in the sequence of the gene for the enzyme, 5, 10-methylenetetrahydrofolate reductase (MTHFR), known as the MTHFR c.677C>T polymorphism, results in a thermolabile enzyme (21). The substitution of a cytosine (C) by a thymine (T) at nucleotide 677 in the exon 4 of MTHFR gene leads to an alanine-to-valine transition in the catalytic domain of the enzyme. Depending on the population, 20% to 53% of individuals may have inherited one T copy (677C/T genotype), and 3% to 32% of individuals may have inherited two T copies (677T/T genotype) for the MTHFR gene (22). MTHFR catalyzes the reduction of 5,10-methylenetetrahydrofolate (5,10-methylene THF) into 5-methyl tetrahydrofolate (5-MeTHF). The latter is the folate coenzyme required to form methionine from homocysteine (see Figure 2). MTHFR activity is greatly diminished in heterozygous 677C/T (-30%) and homozygous 677T/T (-65%) individuals compared to those with the 677C/C genotype (23). Homozygosity for the mutation (677T/T) is linked to lower concentrations of folate in red blood cells and higher blood concentrations of homocysteine (24, 25). Improving folate nutritional status in elderly women with the T allele reduced plasma homocysteine concentration (26). An important unanswered question about folate is whether the present RDA is enough to compensate for the reduced MTHFR enzyme activity in individuals with at least one T allele, or whether those individuals have a higher folate requirement than the RDA (27).

Disease Prevention

Adverse pregnancy outcomes

Neural tube defects

Fetal growth and development are characterized by widespread cell division. Adequate folate is critical for DNA and RNA synthesis. Neural tube defects (NTDs) arise from failure of embryonic neural tube closure between the 21st and 28th days after conception, a time when many women may not even realize they are pregnant (28). NTDs include various malformations, such as lesions of the brain (e.g., anencephaly, encephalocele) or lesions of the spine (spina bifida), which are devastating and life-threatening (29). The worldwide prevalence of NTDs is estimated to be 2 per 1,000 births, which translates to 214,000-322,000 cases annually (30).

Results of randomized trials have demonstrated 60% to 100% reductions in NTD cases when women consumed folic acid supplements in addition to a varied diet during the periconceptional period (about one month before and at least one month after conception) (31, 32). To decrease the incidence of NTDs in the US (about 1 NTD case per 1,000 pregnancies; 1), the Food and Drug Administration implemented legislation in 1998 requiring the fortification of all enriched grain products with 1.4 mg of folic acid per kg of grain (see Sources). The required level of folic acid fortification in the US was initially estimated to provide 100 μg of additional folic acid in the average person's diet, though it probably provides even more due to overuse of folic acid by food manufacturers (27, 33). As a result of this fortification mandate, a 30% decrease in the prevalence of NTDs was noted compared to the pre-fortification period; the post-fortification prevalence of NTDs was found to be 0.69 cases per 1,000 live births and fetal deaths (34). Recent estimates indicate an NTD prevalence less than 0.5 cases per 1,000 live births and that fortification strategies in the US prevent 1,326 NTDs each year (35). To date, 58 nations worldwide have established mandatory programs of folic acid fortification of staple grains (30, 36), and an estimated 22% of NTDs have been prevented through such programs (36). Many countries in Europe, Asia, and Africa, however, have not mandated folic acid fortification of staple foods (37).

The US Public Health Service recommends that all persons capable of becoming pregnant consume 400 μg of folic acid daily to prevent NTDs. Those with a previously affected pregnancy were also advised to receive 4,000 μg (4 mg) of folic acid daily in order to reduce NTD recurrence (38). These recommendations were made to all persons of childbearing age because adequate folate must be available very early in pregnancy, and because many pregnancies in the US are unplanned (39). Despite the effectiveness of folic acid supplementation in improving folate status, it appears that globally only 30% of women who become pregnant correctly follow the recommendation, and there is some concern that young women from minority ethnic groups and lower socioeconomic backgrounds are the least likely to follow the recommendation (40-43).

Also, a genetic component in NTD etiology is evidenced by the increased risk in women with a family history of an NTD and also by variations in risk among ethnicities (44). Moreover, NTD occurrence can be attributed to specific folate-gene interactions. The MTHFR c.677C>T polymorphism and other genetic variations can increase the folate requirement and susceptibility for an NTD-affected pregnancy. Prior to the fortification era, a case-control study showed that both red blood cell and serum folate concentrations were significantly lower in pregnant women with the T/T and C/T variants compared to the wild-type C/C genotype (24), suggesting inadequate folate metabolism with specific maternal genotypes. A meta-analysis of 34 case-control studies, including 3,018 case mothers and 8,746 control mothers, showed a positive association between the maternal MTHFR c.677C>T polymorphism and NTDs (45). Another MTHFR variant, an A-to-C change at position 1298, has also been associated with reduced MTHFR activity and increased NTD risk in some (46, 47), but not all (48), populations. Individuals heterozygous for both of these MTHFR variants (677C/T + 1298A/C) exhibit lower plasma folate and higher homocysteine concentrations than individuals with 677C/T + 1298A/A (49). Combined genotypes with homozygosity G/G for the reduced folate carrier transporter (RFC-1) polymorphism (c.80A>G) could further contribute to NTD occurrence (50). The degree of NTD risk was also assessed with additional MTHFR polymorphisms (c.116C>T, c.1793G>A) (51), as well as with mutations affecting other enzymes of the one-carbon metabolism, including methionine synthase (MTR c.2756A>G) (52), methionine synthase reductase (MTRR c.66A>G) (53), and methylenetetrahydrofolate dehydrogenase (MTHFD1 c.1958G>A) (54, 55). While maternal genotype can impact pregnancy outcome, it appears that gene-gene interactions between mother and fetus influence it further. The risk of NTD was increased by certain genetic combinations, including maternal (MTHFR c.677C>T)-fetal (MTHFR c.677C>T) and maternal (MTRR c.66A>G)-fetal (MTHFR c.677C>T) interactions (52, 53, 56). Finally, vitamin B12 status has been associated with NTD risk modification in the presence of specific polymorphisms in one-carbon metabolism (57).

Cardiovascular malformations

Congenital anomalies of the heart are a major cause of infant mortality but also cause deaths in adulthood (58). Using data from the European Registration of Congenital Anomalies and Twins (EUROCAT) database, a case-control study, involving 596 cases and 2,359 controls, found that consumption of at least 400 μg/day of folic acid during the periconceptual period (one month before conception through eight weeks' post-conception, covering the period of embryonic heart development) was associated with an 18% reduced risk of congenital heart defects (59). Meta-analyses of 20 to 25 case-control and family-based studies observed positive associations between maternal, fetal, or paternal MTHFR c.677C>T variant and incidence of congenital heart defects (60, 61). Additional studies are needed to elucidate the effects of gene-nutrient interactions on the risk of congenital heart defects; however, the currently available research indicates that adequate folate intake may play an important role.

Orofacial clefts

Maternal folate status during pregnancy may influence the risk of congenital anomalies called orofacial clefts, namely cleft lip with or without cleft palate (CL/P) (62). Most orofacial clefts are non-syndromic clefts, meaning no other congenital malformations are present (63). In a recent systematic review and meta-analysis of 39 observational studies, use of folic acid-containing supplements preconceptionally or during pregnancy was associated with a 42% lower risk of CL/P (95% CI, 0.49-0.70) (64). While some studies have suggested that polymorphisms in the cystathionine β-synthase (CBS) gene (c.699C>T) or MTHFR gene (c.677C>T) might be protective of orofacial clefts at low folate intakes (65, 66), this pooled analysis did not find an association between MTHFR polymorphisms and orofacial clefts (64). An earlier meta-analysis of observational studies evaluating mandatory programs of folic acid fortification globally found that folic acid fortification was only associated with a reduced risk of non-syndromic CL/P (5 studies) and not with orofacial clefts in total (24 studies), CL/P (16 studies), or cleft palate only (16 studies) (67). It is important to note that the observational studies to date are highly heterogeneous, which presents challenges when pooling study results in a meta-analysis.

Other adverse pregnancy outcomes

Low birth weight has been associated with increased risk of mortality during the first year of life and may also influence health outcomes during adulthood (68). A systematic review and meta-analysis of eight randomized controlled trials found a positive association between folic acid supplementation and birth weight; no association with length of gestation was observed (69). Additionally, a prospective cohort study of 306 pregnant adolescents associated low folate intakes and maternal folate status during the third trimester of pregnancy with higher incidence of small for gestational age births (birth weight <10th percentile) (70). Moreover, the maternal c.677C>T MTHFR genotype and increased homocysteine concentrations, considered an indicator of functional folate deficiency, have been linked to lower birth weights (71).

Elevated blood homocysteine concentrations have also been associated with increased incidence of miscarriage and other pregnancy complications, including preeclampsia and placental abruption (72). A large retrospective study showed that high plasma homocysteine in Norwegian women was strongly related to adverse outcomes and complications, including preeclampsia, premature delivery, and very low birth weight, in previous pregnancies (73). A meta-analysis of 51 prospective cohort studies linked the c.677C>T MTHFR variant with increased risk of preeclampsia in Caucasian and East Asian populations, reinforcing the notion that folate metabolism may play a role in the condition (74). However, it is not known whether maternal folic acid supplementation during pregnancy reduces the risk of preeclampsia, although results of a meta-analysis of 12 studies (11 cohort studies and 1 randomized controlled trial) suggested a lower risk (RR, 0.69; 95% CI 0.58-0.83) (75). A large international, multicenter, double-blind, placebo-controlled trial, the Folic Acid Clinical Trial (FACT), was initiated to evaluate whether high-dose folic acid supplementation throughout pregnancy could prevent preeclampsia and other adverse outcomes (e.g., maternal death, placental abruption, preterm delivery) (76). In this trial in 2,301 pregnant women at high risk of preeclampsia, 4.0-5.1 mg/day of supplemental folic acid from 8 to 16 weeks’ gestation until delivery did not lower risk of preeclampsia or any other adverse outcome when compared to placebo (subjects in the placebo group could take up to 1.1 mg/day of folic acid) (77).

Overall, findings of observational studies on the association of maternal folic acid supplementation and autism spectrum disorders in the offspring have been inconsistent (reviewed in 78, 79). Additionally, systematic reviews of observational studies have found no evidence of an association between folate exposure during pregnancy and childhood asthma or allergies (80, 81).

Adequate folate intake throughout pregnancy is known to protect against megaloblastic anemia (82). Thus, it seems reasonable to maintain folic acid supplementation throughout pregnancy, even after closure of the neural tube, in order to decrease the risk of other problems during pregnancy.

Cardiovascular disease

Homocysteine and cardiovascular disease

The results of more than 80 observational studies indicate that even moderately elevated concentrations of homocysteine in the blood increase the risk of cardiovascular disease (CVD) (4). Possible predispositions to vascular accidents have also been linked to genetic deficiencies in homocysteine metabolism in certain populations (83). The mechanism by which homocysteine may increase the risk of vascular disease has been the subject of a great deal of research, but it may involve adverse effects of homocysteine on blood clotting, arterial vasodilation, and thickening of arterial walls (84). Although increased homocysteine concentrations in the blood have been consistently associated with increased risk of CVD, it is unclear whether lowering circulating homocysteine will reduce CVD risk (see Folate and homocysteine). Elevated homocysteine may instead be a biomarker of cardiovascular disease rather than a causative factor (85).

Research had initially predicted that a prolonged decrease in serum homocysteine level of 3 micromoles/liter would lower the risk of CVD by up to 25% and be a reasonable treatment goal for individuals at high risk (86, 87). However, the analysis of clinical trials of B-vitamin supplementation has shown that lowering homocysteine concentrations did not prevent the occurrence of a second cardiovascular event in patients with existing CVD (88-90). Consequently, the American Heart Association recommends screening for elevated total homocysteine concentrations only in "high risk" individuals, for example, in those with personal or family history of premature cardiovascular disease, malnutrition or malabsorption syndromes, hypothyroidism, kidney failure, lupus, or for individuals taking certain medications (nicotinic acid, theophylline, bile acid-binding resins, methotrexate, and L-dopa.

Folate and homocysteine

Folate-rich diets have been associated with decreased risk of CVD, including coronary artery disease, myocardial infarction (heart attack), and stroke. A study that followed 1,980 Finnish men for 10 years found that those who consumed the most dietary folate had a 55% lower risk of an acute coronary event when compared to those who consumed the least dietary folate (91). Of the three B vitamins that regulate homocysteine concentrations, folic acid has been shown to have the greatest effect in lowering basal concentrations of homocysteine in the blood when there is no coexisting deficiency of vitamin B12 or vitamin B6 (see Nutrient interactions) (92). Increasing folate intake through folate-rich food or supplements has been found to reduce homocysteine concentrations (93). Moreover, blood homocysteine concentrations have declined since the FDA mandated folic acid fortification of the grain supply in the US (27). A meta-analysis of 25 randomized controlled trials, including almost 3,000 subjects, found that folic acid supplementation with 800 μg/day or more could achieve a maximal 25% reduction in plasma homocysteine concentrations. In this meta-analysis, daily doses of 200 μg and 400 μg of folic acid were associated with a 13% and 20% reduction in plasma homocysteine, respectively (94). A supplement regimen of 400 μg of folic acid, 2 mg of vitamin B6, and 6 μg of vitamin B12 has been advocated by the American Heart Association if an initial trial of a folate-rich diet (see Sources) is not successful in adequately lowering homocysteine concentrations (95).  

Several polymorphisms in folate/one-carbon metabolism modify homocysteine concentrations in blood (96). In particular, the effect of the c.677C>T MTHFR variant has been examined in relation to folic acid fortification policies worldwide. The analysis of randomized trials, including 59,995 subjects without a history of CVD, revealed that the difference in homocysteine concentrations between T/T and C/C genotypes was greater in low-folate regions compared to regions with food fortification policy (3.12 vs. 0.13 micromoles/liter) (97). Although folic acid supplementation effectively decreases homocysteine concentrations, it is not yet clear whether it also decreases risk for CVD. A meta-analysis of 19 randomized clinical trials, including 47,921 subjects with preexisting cardiovascular or renal disease, found that homocysteine lowering through folic acid and other B-vitamin supplementation failed to reduce the incidence of CVD despite significant reductions in plasma homocysteine concentrations (90). Other meta-analyses have confirmed the lack of causality between the lowering of homocysteine and the risk of CVD (96-98). Consequently, the American Heart Association removed its recommendation for using folic acid to prevent cardiovascular disease in high-risk women (99). However, more recent evidence suggests that homocysteine-lowering therapy with B vitamins may help prevent stroke in particular. A meta-analysis of three randomized controlled trials in patients with existing vascular disease found a benefit of B-vitamin supplementation on prevention of recurrent stroke (HR, 0.71; 95% CI, 0.58-0.88) (100). Other recent meta-analyses of randomized controlled trials have found that folic acid supplementation decreases the risk of stroke or cerebrovascular events in patients with existing CVD (101-103), as well as in those without CVD (104). A recent systematic review and meta-analysis of 21 randomized controlled trials — mostly in patients with existing CVD — found that folic acid supplementation slightly lowers systolic (1.10 mm Hg) and diastolic (-0.24 mm Hg) blood pressure (105).

Moreover, some studies have investigated the effect of folic acid supplementation on the development of atherosclerosis, a known risk factor for vascular accidents. The measurement of the carotid intima-media thickness (CIMT) is a surrogate endpoint for early atherosclerosis and a predictor for cardiovascular events (106). The meta-analysis of 10 randomized trials testing the effect of folic acid supplementation showed a significant reduction in CIMT in subjects with chronic kidney diseases and in those at risk for CVD, but not in healthy participants (107). Endothelial dysfunction is a common feature in atherosclerosis and vascular disease. High doses of folic acid (400-10,000 μg/day) have been associated with improvements in vascular health in both healthy and CVD subjects (108). A meta-analysis of 21 randomized, placebo-controlled trials found that folic acid supplementation improves endothelial function by increasing flow-mediated vasodilation (109).

Although some trials have failed to demonstrate any cardiovascular protection from folic acid supplementation, low folate intake is a known risk factor for vascular disease, and more research is needed to explore the role of folate in maintaining vascular health (110).  

Cancer

Cancer is thought to arise from DNA damage in excess of ongoing DNA repair and/or the inappropriate expression of critical genes. Because of the important roles played by folate in DNA and RNA synthesis and methylation, it is possible that inadequate folate intake contributes to genome instability and chromosome breakage that often characterize cancer development. In particular, DNA replication and repair are critical for genome maintenance, and the shortage in nucleotides caused by folate deficiency might lead to genome instability and DNA mutations. A decrease in 5,10-methylene THF can compromise the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP) by the enzyme thymidylate synthase (TYMS), causing uracil accumulation and thymine depletion. This could then lead to uracil misincorporation into DNA during replication or repair, and cause DNA damage, including point mutations and strand breaks (111). Since 5,10-methylene THF is also the MTHFR enzyme substrate, it is plausible that a reduction of MTHFR activity with the c.677C>T polymorphism may increase the use of 5,10-methylene THF for thymidylate synthesis and prevent DNA damage. However, this hypothesis might only be valid in a situation of folate deficiency (112). Conversely, it was argued that folic acid supplementation could fuel DNA synthesis, therefore promoting tumor growth. This is supported by the observation that TYMS can function like a tumor promoter (oncogene), while a reduction in TYMS activity is linked to a lower risk of cancer (113, 114). Additionally, antifolate molecules that block the thymidylate synthesis pathway are successfully used in cancer therapy (115). Folate also controls the homocysteine/methionine cycle and the pool of S-adenosylmethionine (SAM), the methyl donor for methylation reactions. Thus, folate deficiency may impair DNA and protein methylation and alter the expression of genes involved in DNA repair, cell proliferation, and cell death. Global DNA hypomethylation, a typical hallmark of cancer, causes genome instability and chromosome breaks (reviewed in 116).

The consumption of at least five servings of fruit and vegetables daily has been consistently associated with a decreased incidence of cancer (117, 118). Fruit and vegetables are excellent sources of folate, which may play a role in their anti-carcinogenic effect. Observational studies have found diminished folate status to be associated with site-specific cancers. While food fortification is mandatory in the US (since 1998; see Sources), concerns about the impact of high folic acid intakes on health have delayed the practice in several other countries (119). However, meta-analyses of folic acid intervention trials (supplemental doses ranging from 500 to 5,000 μg/day for at least one year) did not show any specific benefit or harm regarding total and site-specific cancer incidence (120, 121). In 2015, an expert panel convened by the US National Toxicology Program and the US Office of Dietary Supplements concluded that folic acid supplementation does not reduce cancer incidence in folate replete individuals and that further research is needed to determine whether supplemental folic acid might promote cancer growth (122).

Colorectal cancer

A pooled analysis of 13 prospective cohort studies, which followed a total of 725,134 individuals for a 7 to 20-year period, revealed a modest, inverse association between dietary and total (from food and supplements) folate intake and colon cancer risk. Specifically, a 2% decrease in colon cancer risk was estimated for every 100 μg/day increase in total folate intake (123). A large US prospective study, which followed 525,488 subjects, ages 50 to 71 years between 1995 and 2006, correlated higher intakes of dietary folate, supplemental folic acid, and total folate (diet and supplements combined) with decreased colorectal cancer (CRC) risk (124). However, when stratified by gender, there was no association between dietary folate intake and CRC risk in women (124, 125). A lack of association between CRC risk and dietary, supplemental, and total folate intakes was also reported in another prospective study that followed more than 90,000 US postmenopausal women during an 11-year period encompassing pre- and post-fortification periods (126). Yet, in the long-term follow up (followed for up to 24 years) of 86,320 women participating in the prospective Nurses’ Health Study, higher total intakes of folate at baseline, as well as higher supplemental folic acid intakes, were associated with lower risks of CRC (127).

Finally, a meta-analysis of 18 case-control studies found a slight reduction in CRC risk with folate from food (128). However, it is important to note that the case-control studies were highly heterogeneous, and that the authors stated that dietary fiber, vitamins, and alcohol intake could have confounded their results. Moreover, the lower limit of the highest quantile of folate intake was highly variable, ranging from 270 to 1,367 μg/day (128).

While most observational research shows a protective association of folate against colorectal cancer development, it has been suggested that high doses of supplemental folic acid may actually accelerate tumor growth in cancer patients (129). Whereas higher folate status within the normal dietary range is widely considered to be protective against cancer, some investigators remain concerned that exposure to excessively high folic acid intakes may increase the growth of pre-existing neoplasms (129). Several clinical trials addressed the effect of folic acid supplementation in patients with a history of colorectal adenoma, with trials finding a risk reduction or no effect of supplemental folic acid (130-133). A meta-analysis of three large randomized controlled trials in high-risk subjects did not demonstrate any increase in colorectal adenoma recurrence in subjects supplemented with 500 or 1,000 μg/day of folic acid for 24 to 42 months when compared with placebo treatment (134). The B-PROOF study was a randomized, double-blind, placebo-controlled trial examining whether supplemental folic acid and vitamin B12 affected risk of osteoporotic fracture in older adults (≥65 years) with hyperhomocysteinemia (135). In a secondary analysis of 2,524 older adults participating in B-PROOF, co-supplementation with folic acid (400 μg/day) and vitamin B12 (500 μg/day) for 2 to 3 years significantly increased risk of colorectal cancer (HR 1.77; 95% CI, 1.08-2.90) (136).

As suggested above, the MTHFR 677T/T genotype might prevent uracil misincorporation and protect DNA integrity and stability under low-folate conditions. A meta-analysis of 62 case-control and two cohort studies revealed that while the T/T variant reduces CRC risk by 12% compared to both C/T and C/C genotypes, the risk was decreased by 30% with high (348-1,583 μg/day) versus low total folate intakes (264-450 μg/day), irrespective of the genotype (137). A common polymorphism (c.2756A>G) in the MTR gene, which codes for methionine synthase, was also examined in relation with the risk of colorectal adenoma and cancer. Methionine synthase catalyzes the simultaneous conversion of homocysteine and 5-methylene THF into methionine and TFH, respectively. Meta-analyses of case-control studies have found no association between MTR variants and colorectal cancer risk (138, 139).

Alcohol consumption interferes with the absorption and metabolism of folate (18). One case-control and five prospective cohort studies have reported either reduction in CRC risk among nondrinkers compared to drinkers or a lack of association (128). In a large prospective study that followed more than 28,000 male health professionals for 22 years, intake of more than two alcoholic drinks (>30 grams of alcohol) per day augmented CRC risk by 42% during the pre-fortification period. CRC risk was not increased during the post-fortification period, suggesting that it is the combination of high alcohol and low folate intake that might increase CRC risk. Yet, another prospective study that followed more than 69,000 female nurses for 28 years did not report a significant increase in CRC risk with alcohol intake before and after the mandatory folic acid fortification (140). In some studies, individuals who are homozygous for the c.677C>T MTHFR polymorphism (T/T) have been found to be at decreased risk for colon cancer when folate intake is adequate. However, when folate intake is low and/or alcohol intake is high, individuals with the (T/T) genotype have been found to be at increased risk of colorectal cancer (141, 142).

Breast cancer

Several prospective cohort and case-control studies investigating whether folate intake is associated with breast cancer have reported mixed results (143). A meta-analysis of 39 prospective cohort and case-control studies found dietary folate intake to be inversely associated with risk of breast cancer, but data stratification revealed the association was evident in premenopausal women and not in postmenopausal women (144). Another meta-analysis of only prospective cohort studies (n=23) found a similar protective association in premenopausal women (145).

Moderate alcohol intake has been associated with increased risk of breast cancer in women (146). Results from prospective studies have also suggested that increased folate intake may reduce the risk of breast cancer in women who regularly consume alcohol (145, 147-150). Thus, high folate intake might be associated with a risk reduction only in women whose breast cancer risk is raised by alcohol consumption. A very large prospective study in more than 88,000 nurses reported that folic acid intake was not associated with breast cancer in women who consumed less than one alcoholic drink per day. However, in women consuming at least one alcoholic drink per day, folic acid intake of at least 600 μg daily resulted in about half the risk of breast cancer compared with women who consumed less than 300 μg of folic acid daily (149). Nevertheless, how alcohol consumption increases breast cancer risk is still subject to discussion (151).

Finally, meta-analyses evaluating the influence of polymorphisms in one-carbon metabolism on cancer risk found that specific variants in the genes encoding thymidylate synthase (152, 153), methionine synthase (154), methionine synthase reductase (154), and MTHFR (154) increase the risk of breast cancer in certain ethnic populations.

Childhood cancers

The incidence of Wilms' tumors (kidney cancer) and certain types of brain cancers (neuroblastoma, ganglioneuroblastoma, and ependymoma) in children has decreased since the mandatory fortification of the US grain supply in 1998 (155). However, incidence rates were unchanged between the pre- and post-fortification periods for leukemia – a predominant childhood malignancy. Overall, the findings of observational studies of maternal folic acid supplementation during pregnancy and childhood cancers have been mixed. A recent meta-analysis of case-control studies found a protective association of maternal folic acid supplementation during pregnancy with childhood acute lymphoblastic leukemia (ALL; OR, 0.75; 95% CI 0.66-0.86; 11 studies) but not with acute myeloid leukemia (5 studies) or childhood brain cancers (6 studies) (156).

Additionally, several meta-analyses have found little to no protective association of MTHFR polymorphisms; however, the most recent meta-analysis of 66 case-control studies found a reduction in the risk of ALL with the c.677C>T variant, driven mainly by a protective association in Caucasians and Asians (157).

Alzheimer's disease and cognitive impairment

Alzheimer’s disease (AD) is the most common form of dementia, affecting more than 5 million people 65 years or older in the US (158). β-amyloid plaque deposition, Tau protein-forming tangles, and increased cell death in the brain of AD patients have been associated with cognitive decline and memory loss. One study associated increased consumption of fruit and vegetables, which are abundant sources of folate, with a reduced risk of developing dementia and AD in women (159). In the Baltimore Longitudinal Study of Aging that followed 579 nondemented older adults for an average of 9.3 years, daily folate intakes of at least 400 μg at baseline were associated with a 55% lower risk of developing Alzheimer’s disease compared to lower intakes (160). Through its role in nucleic acid synthesis and methyl donor provision for methylation reactions, folate is critical for normal brain development and function, not only throughout pregnancy and postnatally but also later in life (161). In one cross-sectional study of elderly women, AD patients had significantly higher homocysteine and lower red blood cell folate concentrations compared to healthy individuals. However, there was no difference in the level of serum folate between groups, suggesting that long-term folate status, rather than recent folate intake, may be associated with the risk of AD (162).

Several investigators have described associations between increased homocysteine concentrations and cognitive impairment in the elderly (163), but prospective cohort studies examining dietary or total daily folate intake and cognition have reported mixed results (164-169). Higher homocysteine concentrations were found in individuals suffering from dementia, including AD and vascular dementia, compared to healthy subjects (170, 171).

Over the past few decades, hyperhomocysteinemia in older adults has become recognized as a modifiable risk factor for cognitive decline, dementia, and Alzheimer’s disease (172-174). A Cochrane review of 14 placebo-controlled trials found that B-vitamin supplementation (folic acid, vitamin B6, vitamin B12) in older adults who were primarily "cognitively healthy" (but many had cardiovascular disease or other conditions) had no significant benefits on global cognitive function (175). Moreover, a recent meta-analysis of 14 randomized, placebo-controlled trials in older adults found that any benefits of B-vitamin supplementation were limited to slowing cognitive decline (176). Stratification of the data revealed that cognitive benefits were seen only in those without dementia at baseline and in trials of more than one year in duration (176). However, not every trial included in these meta-analyses supplemented with folic acid.

Some trials have examined the effects of B-vitamin supplementation in older adults experiencing cognitive impairment, with a few finding cognitive benefit. A two-year randomized, placebo-controlled trial in 168 elderly subjects with mild cognitive impairment described the benefits of a daily regimen of 800 μg of folic acid, 500 μg of vitamin B12, and 20 mg of vitamin B6 (177, 178). Atrophy of specific brain regions affected by AD was observed in individuals of both groups, and this atrophy correlated with cognitive decline; however, the B-vitamin treatment group experienced a smaller loss of gray matter compared to the placebo group (0.5% vs. 3.7%). A greater benefit was seen in subjects with higher baseline homocysteine concentrations, suggesting the importance of lowering circulating homocysteine in prevention of cognitive decline and dementia. Three randomized controlled trials conducted in Chinese adults ages 65 years or older found that supplementation with 400 μg/day of folic acid for 6 to 24 months improved some measures of cognitive function compared to a no-treatment control/conventional treatment (179-181), suggesting that supplemental folic acid in populations without mandated folic acid fortification may confer cognitive benefit. Large double-blind, placebo-controlled trials are needed to understand whether folic acid supplementation late in life might help decrease age-related cognitive impairments or the risk of Alzheimer’s disease and other dementias. Yet, it is prudent to ensure adequate intakes of folate and other B vitamins throughout life for brain — and overall — health.  

Disease Treatment

Metabolic diseases

Folinic acid (see Figure 1; also known as leucovorin), a tetrahydrofolic acid derivative, is used in the clinical management of rare inborn errors that affect folate transport or metabolism (reviewed in 182). Such conditions are of autosomal recessive inheritance, meaning only individuals receiving two copies of the mutated gene (one from each parent) develop the disease. Folinic acid can be administered orally, intravenously, or intramuscularly.

Hereditary folate malabsorption

Hereditary folate malabsorption is caused by mutations in the SLC46A1 gene coding for the folate transporter PCFT and typically affects gastrointestinal folate absorption and folate transport into the brain (183). Patients present with low to undetectable concentrations of folate in serum and cerebrospinal fluid; anemia (often macrocytic), thrombocytopenia, and/or pancytopenia (low number of all blood cells); impaired immune responses that increase susceptibility to infections; and a general failure to thrive (184, 185). Neurologic symptoms, including developmental delays, cognitive disorders, and seizures, have also been observed (185, 186). Clinical improvements have been recorded following provision of folinic acid parenterally (187, 188) as well as intramuscularly (188, 189).

Cerebral Folate Deficiency (CFD) syndrome

CFD is characterized by low levels of folate coenzymes in cerebrospinal fluid despite often normal concentrations of folate in blood. Folate transport across the blood-brain barrier is compromised in CFD and has been linked either to the presence of autoantibodies blocking the folate receptor-alpha (FRα) or to mutations in the FOLR1 gene encoding FRα (190-193). Neurological abnormalities, along with visual and hearing impairments, have been described in children with CFD; autism spectrum disorder (ASD) is present in some cases. CFD has also been described in adults presenting with neurological symptoms (194).

Folinic acid can enter the brain and normalize the level of folate coenzymes and has been shown to normalize folate concentrations and improve various social interactions in CFD, including mood, behavior, and verbal communication in children with ASD (190, 195, 196).

Dihydrofolate reductase (DHFR) deficiency

DHFR is the NADPH-dependent enzyme that catalyzes the reduction of dihydrofolic acid to tetrahydrofolic acid. DHFR is also required to convert folic acid to DHF. DHFR deficiency is characterized by megaloblastic anemia and cerebral folate deficiency causing intractable seizures and mental deficits. Although folinic acid treatment can alleviate the symptoms of DHFR deficiency, early diagnosis is essential to prevent irreversible brain damage and improve clinical outcomes (197, 198).

Sources

Food sources

Green leafy vegetables (foliage) are rich sources of folate and provide the basis for its name. Citrus fruit juices, legumes, and fortified foods are also excellent sources of folate (1); the folate content of fortified cereal varies greatly. A number of folate-rich foods are listed in Table 2, along with their folate content in micrograms (μg). For more information on the nutrient content of specific foods, search USDA's FoodData Central.

Table 2. Some Food Sources of Folate and Folic Acid
Food Serving Folate (μg DFEs)
Lentils (cooked, boiled) ½ cup 179
Garbanzo beans (chickpeas, boiled) ½ cup 141
Asparagus (boiled) ½ cup (~6 spears) 134
Spinach (boiled) ½ cup 132
Lima beans (boiled) ½ cup 78
Orange juice 6 fl. oz. 56
Bread (enriched) 1 slice 50*
Spaghetti (enriched, cooked) 1 cup 180*
White rice (enriched, cooked) 1 cup 153-180*
*To help prevent neural tube defects, the US FDA required the addition of 1.4 milligrams (mg) of folic acid per kilogram (kg) of grain to be added to refined grain products, which were already enriched with niacin, thiamin, riboflavin, and iron, as of January 1, 1998. The addition of nutrients to food in order to prevent a nutritional deficiency or restore nutrients lost in processing is known as fortification. The FDA initially estimated that this level of fortification would increase dietary intake by an average of 100 μg folic acid/day (28). However, further evaluations based on observational studies suggested increases twice that predicted by the FDA (33). On April 14, 2016, the FDA approved the voluntary fortification of corn masa flour up to 1.5 mg of folic acid per kg corn flour (199).

Supplements

The principal form of supplemental folate is folic acid. It is available as a single-ingredient supplement and also included in combination products, such as B-complex vitamins and multivitamins. Doses of 1 mg or greater require a prescription (200). 5-Methyltetrahydrofolate is also available as a supplement (201).

Additionally, folinic acid, a tetrahydrofolic acid derivative, is used to manage certain metabolic diseases (see Disease Treatment). Further, the US FDA has approved the supplementation of folate in oral contraceptives. The addition of levomefolate calcium (the calcium salt of MeTHF; 451 μg/tablet) to oral contraceptives is intended to raise folate status in women of childbearing age (202). According to a US national survey, only 24% of non-pregnant women aged 15-44 years are meeting the current recommendation of 400 μg/day of folic acid (203).

Safety

Toxicity

No adverse effects have been associated with the consumption of excess folate from food. Concerns regarding safety are limited to synthetic folic acid intake. Deficiency of vitamin B12, though often undiagnosed, may affect a significant number of people, especially older adults (see the separate article on Vitamin B12). One symptom of vitamin B12 deficiency is megaloblastic anemia, which is indistinguishable from that associated with folate deficiency (see Deficiency). There is concern that large doses of folic acid given to an individual with an undiagnosed vitamin B12 deficiency could correct megaloblastic anemia without correcting the underlying vitamin B12 deficiency, leaving the individual at risk of developing irreversible neurologic damage. Such cases of neurologic progression in vitamin B12 deficiency have been mostly seen in case studies at folic acid doses of 5,000 μg (5 mg) and above. In order to be very sure of preventing irreversible neurologic damage in vitamin B12-deficient individuals, the Food and Nutrition Board of the US National Academy of Medicine advises that all adults limit their intake of folic acid (supplements and fortification) to 1,000 μg (1 mg) daily (Table 3). The Board also noted that vitamin B12 deficiency is very rare in women in their childbearing years, making the consumption of folic acid at or above 1,000 μg/day unlikely to cause problems (1); however, there are limited data on the effects of large doses (78).

Table 3. Tolerable Upper Intake Level (UL) for Folic Acid
Age Group UL (μg/day)
Infants 0-12 months Not possible to establish*
Children 1-3 years 300
Children 4-8 years 400
Children 9-13 years 600
Adolescents 14-18 years 800
Adults 19 years and older 1,000
*Source of intake should be from food and formula only.

The saturation of DHFR metabolic capacity by oral doses of folic acid has been associated with the appearance of unmetabolized folic acid in blood (204). Exposure to folic acid in the food supply is also associated with unmetabolized folic acid in the blood. A US national cross-sectional survey, NHANES 2007-2008, detected unmetabolized folic acid in 96% of serum samples of Americans (≥1 year of age) (205). In an ancillary study of the Folic Acid Clinical Trial, maternal supplementation with 4.0-5.1 mg/day of folic acid during pregnancy (starting at 8 to 16 weeks’ gestation) led to higher maternal blood concentrations of unmetabolized folic acid and 5-methylTHF at 24 to 26 weeks’ gestation compared to dosages of 1.1 mg/day or less (206). However, no differences were seen in other markers of one-carbon metabolism, including serum concentrations of THF, 5-formylTHF, 5,10-methenylTHF, vitamin B12, and pyridoxal 5’-phosphate (206). A randomized controlled trial in 126 pregnant women found that continuing folic acid supplementation of 400 μg/day beyond the first trimester of pregnancy (i.e., gestational weeks 14 throughout the second and third trimesters) did not increase blood concentrations of unmetabolized folic acid at 36 weeks’ gestation compared to placebo despite increases in plasma total folate, plasma 5-MTHF, and red blood cell folate (207). Maternal folic acid supplementation throughout pregnancy also did not increase unmetabolized folic acid detected in neonatal cord blood samples compared to placebo (207).

Evidence of adverse health effects of unmetabolized folic acid is quite limited. Hematologic abnormalities and poorer cognition have been associated with the presence of unmetabolized folic acid in vitamin B12-deficient older adults (≥60 years) (208, 209). A small study conducted in postmenopausal women also raised concerns about the effect of exposure to unmetabolized folic acid on immune function (210). Recent observational studies have found no link between circulating maternal unmetabolized folate during pregnancy and allergic disease in infancy (211) or autistic traits or language impairment in early childhood (212). In a small, randomized, open-label trial in 38 women of reproductive age receiving 30 weeks of daily multivitamin supplements, daily supplementation with either 1.1 mg or 5 mg of folic acid resulted in the transient appearance of unmetabolized folic acid in blood over the first 12 weeks of supplementation (213). However, unmetabolized folic acid concentrations returned to baseline levels at the end of the study, suggesting that adaptive mechanisms eventually converted folic acid to reduced forms of folate. Nonetheless, the use of supplemental 5-methyltetrahydrofolate may provide an alternative to prevent any potential negative effects of unconverted folic acid in older adults.

Drug interactions

When nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin or ibuprofen, are taken in very large therapeutic dosages (i.e., to treat severe arthritis), they may interfere with folate metabolism. In contrast, routine use of NSAIDs has not been found to adversely affect folate status. The anticonvulsant, phenytoin, has been shown to inhibit the intestinal absorption of folate, and several studies have associated decreased folate status with long-term use of the anticonvulsants, phenytoin, phenobarbital, and primidone (214). However, few studies controlled for differences in dietary folate intake between anticonvulsant users and nonusers. Also, taking folic acid at the same time as the cholesterol-lowering agents, cholestyramine and colestipol, may decrease the absorption of folic acid (200). Methotrexate is a folic acid antagonist used to treat a number of diseases, including cancer, rheumatoid arthritis, and psoriasis. Some of the side effects of methotrexate are similar to those of severe folate deficiency, and supplementation with folic or folinic acid is used to reduce antifolate toxicity. Other antifolate molecules currently used in cancer therapy include aminopterin, pemetrexed, pralatrexate, and raltitrexed (11, 115). Further, a number of other medications have been shown to have antifolate activity, including trimethoprim (an antibiotic), pyrimethamine (an antimalarial), triamterene (a blood pressure medication), and sulfasalazine (a treatment for ulcerative colitis). Early studies of oral contraceptives (birth control pills) containing high doses of estrogen indicated adverse effects on folate status; however, this finding has not been supported in more recent studies that used low-dose oral contraceptives and controlled for dietary folate (215).

Linus Pauling Institute Recommendation

The available scientific evidence shows that adequate folate intake prevents neural tube defects and other poor outcomes of pregnancy; is helpful in lowering the risk of some forms of cancer, especially in genetically susceptible individuals; and may lower the risk of stroke. The Linus Pauling Institute recommends that adults take a daily multivitamin/mineral supplement, which typically contains 400 μg of folic acid, the Daily Value (DV). Even with a larger than average intake of folic acid from fortified food, it is unlikely that an individual's daily folic acid intake would regularly exceed the tolerable upper intake level of 1,000 μg/day established by the Institute of Medicine (see Safety).

Older adults (>50 years)

The recommendation for 400 μg/day of supplemental folic acid as part of a daily multivitamin/mineral supplement, in addition to a folate-rich diet, is especially important for older adults because blood homocysteine concentrations tend to increase with age (see Disease Prevention).


Authors and Reviewers

Originally written in 2000 by:
Jane Higdon, Ph.D.
Linus Pauling Institute
Oregon State University

Updated in April 2002 by:
Jane Higdon, Ph.D.
Linus Pauling Institute
Oregon State University

Updated in September 2007 by:
Victoria J. Drake, Ph.D.
Linus Pauling Institute
Oregon State University

Updated in June 2014 by:
Barbara Delage, Ph.D.
Linus Pauling Institute
Oregon State University

Updated in October 2023 by:
Victoria J. Drake, Ph.D.
Linus Pauling Institute
Oregon State University

Reviewed in December 2023 by:
Martha S. Field, Ph.D.
Division of Nutritional Sciences
Cornell University

Copyright 2000-2024  Linus Pauling Institute


References

1. Food and Nutrition Board, Institute of Medicine. Folate. Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B-6, Folate, Vitamin B-12, Pantothenic Acid, Biotin, and Choline. Washington D.C.: National Academy Press; 1998:196-305.  (National Academy Press)

2. Choi SW, Mason JB. Folate and carcinogenesis: an integrated scheme. J Nutr. 2000;130(2):129-132.  (PubMed)

3. Bailey LB, Gregory JF, 3rd. Folate metabolism and requirements. J Nutr. 1999;129(4):779-782.  (PubMed)

4. Gerhard GT, Duell PB. Homocysteine and atherosclerosis. Curr Opin Lipidol. 1999;10(5):417-428.  (PubMed)

5. 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(3):613-621.  (PubMed)

6. Jacques PF, Kalmbach R, Bagley PJ, et al. The relationship between riboflavin and plasma total homocysteine in the Framingham Offspring cohort is influenced by folate status and the C677T transition in the methylenetetrahydrofolate reductase gene. J Nutr. 2002;132(2):283-288.  (PubMed)

7. Wilson CP, McNulty H, Ward M, et al. Blood pressure in treated hypertensive individuals with the MTHFR 677TT genotype is responsive to intervention with riboflavin: findings of a targeted randomized trial. Hypertension. 2013;61(6):1302-1308.  (PubMed)

8. Wilson CP, Ward M, McNulty H, et al. Riboflavin offers a targeted strategy for managing hypertension in patients with the MTHFR 677TT genotype: a 4-y follow-up. Am J Clin Nutr. 2012;95(3):766-772.  (PubMed)

9. McNulty H, Dowey le RC, Strain JJ, et al. Riboflavin lowers homocysteine in individuals homozygous for the MTHFR 677C->T polymorphism. Circulation. 2006;113(1):74-80.  (PubMed)

10. McNulty H, Ward M, Hoey L, Hughes CF, Pentieva K. Addressing optimal folate and related B-vitamin status through the lifecycle: health impacts and challenges. Proc Nutr Soc. 2019;78(3):449-462.  (PubMed)

11. Shulpekova Y, Nechaev V, Kardasheva S, et al. The concept of folic acid in health and disease. Molecules. 2021;26(12):3731.  (PubMed)

12. Desmoulin SK, Hou Z, Gangjee A, Matherly LH. The human proton-coupled folate transporter: Biology and therapeutic applications to cancer. Cancer Biol Ther. 2012;13(14):1355-1373.  (PubMed)

13. Solanky N, Requena Jimenez A, D'Souza SW, Sibley CP, Glazier JD. Expression of folate transporters in human placenta and implications for homocysteine metabolism. Placenta. 2010;31(2):134-143.  (PubMed)

14. Halsted CH, Villanueva JA, Devlin AM, Chandler CJ. Metabolic interactions of alcohol and folate. J Nutr. 2002;132(8 Suppl):2367S-2372S.  (PubMed)

15. Pfeiffer CM, Sternberg MR, Schleicher RL, Rybak ME. Dietary supplement use and smoking are important correlates of biomarkers of water-soluble vitamin status after adjusting for sociodemographic and lifestyle variables in a representative sample of U.S. adults. J Nutr. 2013;143(6):957S-965S.  (PubMed)

16. Stark KD, Pawlosky RJ, Sokol RJ, Hannigan JH, Salem N, Jr. Maternal smoking is associated with decreased 5-methyltetrahydrofolate in cord plasma. Am J Clin Nutr. 2007;85(3):796-802.  (PubMed)

17. Hutson JR, Stade B, Lehotay DC, Collier CP, Kapur BM. Folic acid transport to the human fetus is decreased in pregnancies with chronic alcohol exposure. PLoS One. 2012;7(5):e38057.  (PubMed)

18. Herbert V. Folic acid. In: Shils M, Olson, J.A., Shike M., Ross A.C., ed. Modern Nutrition in Health and Disease. Baltimore: Williams & Wilkins; 1999:433-446. 

19. Stabler SP. Clinical folate deficiency. In: Bailey LB, ed. Folate in Health and Disease. 2nd ed. Boca Raton, FL: CRC press, Taylor & Francis Group; 2010:409-428. 

20. Bailey LB. Dietary reference intakes for folate: the debut of dietary folate equivalents. Nutr Rev. 1998;56(10):294-299.  (PubMed)

21. Bailey LB, Gregory JF, 3rd. Polymorphisms of methylenetetrahydrofolate reductase and other enzymes: metabolic significance, risks and impact on folate requirement. J Nutr. 1999;129(5):919-922.  (PubMed)

22. Wilcken B, Bamforth F, Li Z, et al. Geographical and ethnic variation of the 677C>T allele of 5,10 methylenetetrahydrofolate reductase (MTHFR): findings from over 7000 newborns from 16 areas world wide. J Med Genet. 2003;40(8):619-625.  (PubMed)

23. Guenther BD, Sheppard CA, Tran P, Rozen R, Matthews RG, Ludwig ML. The structure and properties of methylenetetrahydrofolate reductase from Escherichia coli suggest how folate ameliorates human hyperhomocysteinemia. Nat Struct Biol. 1999;6(4):359-365.  (PubMed)

24. Molloy AM, Daly S, Mills JL, et al. Thermolabile variant of 5,10-methylenetetrahydrofolate reductase associated with low red-cell folates: implications for folate intake recommendations. Lancet. 1997;349(9065):1591-1593.  (PubMed)

25. Rozen R. Genetic predisposition to hyperhomocysteinemia: deficiency of methylenetetrahydrofolate reductase (MTHFR). Thromb Haemost. 1997;78(1):523-526.  (PubMed)

26. Kauwell GP, Wilsky CE, Cerda JJ, et al. Methylenetetrahydrofolate reductase mutation (677C-->T) negatively influences plasma homocysteine response to marginal folate intake in elderly women. Metabolism. 2000;49(11):1440-1443.  (PubMed)

27. Shane B. Folic acid, vitamin B-12, and vitamin B-6. In: Stipanuk M, ed. Biochemical and Physiological Aspects of Human Nutrition. Philadelphia: W.B. Saunders Co.; 2000:483-518.  

28. Eskes TK. Open or closed? A world of difference: a history of homocysteine research. Nutr Rev. 1998;56(8):236-244.  (PubMed)

29. Czeizel AE, Dudas I, Vereczkey A, Banhidy F. Folate deficiency and folic acid supplementation: the prevention of neural-tube defects and congenital heart defects. Nutrients. 2013;5(11):4760-4775.  (PubMed)

30. Kancherla V. Neural tube defects: a review of global prevalence, causes, and primary prevention. Childs Nerv Syst. 2023;39(7):1703-1710.  (PubMed)

31. MRC Vitamin Study Research Group. Prevention of neural tube defects: results of the Medical Research Council Vitamin Study. Lancet. 1991;338(8760):131-137.  (PubMed)

32. Czeizel AE, Dudas I. Prevention of the first occurrence of neural-tube defects by periconceptional vitamin supplementation. N Engl J Med. 1992;327(26):1832-1835.  (PubMed)

33. Quinlivan EP, Gregory JF, 3rd. Effect of food fortification on folic acid intake in the United States. Am J Clin Nutr. 2003;77(1):221-225.  (PubMed)

34. National Birth Defects Prevention Network. Neural Tube Defect Ascertainment Project. Available at: NTD fact sheet 01-10 for website.pdf. Accessed 12/16/14.

35. Williams J, Mai CT, Mulinare J, et al. Updated estimates of neural tube defects prevented by mandatory folic Acid fortification - United States, 1995-2011. MMWR Morb Mortal Wkly Rep. 2015;64(1):1-5.  (PubMed)

36. Kancherla V, Wagh K, Priyadarshini P, Pachon H, Oakley GP, Jr. A global update on the status of prevention of folic acid-preventable spina bifida and anencephaly in year 2020: 30-Year anniversary of gaining knowledge about folic acid's prevention potential for neural tube defects. Birth Defects Res. 2022;114(20):1392-1403.  (PubMed)

37. Kancherla V, Botto LD, Rowe LA, et al. Preventing birth defects, saving lives, and promoting health equity: an urgent call to action for universal mandatory food fortification with folic acid. Lancet Glob Health. 2022;10(7):e1053-e1057.  (PubMed)

38. Talaulikar VS, Arulkumaran S. Folic acid in obstetric practice: a review. Obstet Gynecol Surv. 2011;66(4):240-247.  (PubMed)

39. American College of Obstetricians and Gynecologists (ACOG). Neural tube defects. Washington, DC. 2003. http://www.guideline.gov/content.aspx?id=3994. Accessed 12/19/14.

40. McNulty B, Pentieva K, Marshall B, et al. Women's compliance with current folic acid recommendations and achievement of optimal vitamin status for preventing neural tube defects. Hum Reprod. 2011;26(6):1530-1536.  (PubMed)

41. Nilsen RM, Vollset SE, Gjessing HK, et al. Patterns and predictors of folic acid supplement use among pregnant women: the Norwegian Mother and Child Cohort Study. Am J Clin Nutr. 2006;84(5):1134-1141.  (PubMed)

42. Ray JG, Singh G, Burrows RF. Evidence for suboptimal use of periconceptional folic acid supplements globally. BJOG. 2004;111(5):399-408.  (PubMed)

43. Rogers LM, Cordero AM, Pfeiffer CM, et al. Global folate status in women of reproductive age: a systematic review with emphasis on methodological issues. Ann N Y Acad Sci. 2018;1431(1):35-57.  (PubMed)

44. Copp AJ, Stanier P, Greene ND. Neural tube defects: recent advances, unsolved questions, and controversies. Lancet Neurol. 2013;12(8):799-810.  (PubMed)

45. Yang Y, Chen J, Wang B, Ding C, Liu H. Association between MTHFR C677T polymorphism and neural tube defect risks: A comprehensive evaluation in three groups of NTD patients, mothers, and fathers. Birth Defects Res A Clin Mol Teratol. 2015;103(6):488-500.  (PubMed)

46. De Marco P, Calevo MG, Moroni A, et al. Study of MTHFR and MS polymorphisms as risk factors for NTD in the Italian population. J Hum Genet. 2002;47(6):319-324.  (PubMed)

47. Boduroglu K, Alanay Y, Alikasifoglu M, Aktas D, Tuncbilek E. Analysis of MTHFR 1298A>C in addition to MTHFR 677C>T polymorphism as a risk factor for neural tube defects in the Turkish population. Turk J Pediatr. 2005;47(4):327-333.  (PubMed)

48. Wang Y, Liu Y, Ji W, et al. Variants in MTHFR gene and neural tube defects susceptibility in China. Metab Brain Dis. 2015;30(4):1017-1026.  (PubMed)

49. van der Put NM, Gabreels F, Stevens EM, et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998;62(5):1044-1051.  (PubMed)

50. De Marco P, Calevo MG, Moroni A, et al. Reduced folate carrier polymorphism (80A-->G) and neural tube defects. Eur J Hum Genet. 2003;11(3):245-252.  (PubMed)

51. O'Leary VB, Mills JL, Parle-McDermott A, et al. Screening for new MTHFR polymorphisms and NTD risk. Am J Med Genet A. 2005;138A(2):99-106.  (PubMed)

52. Christensen B, Arbour L, Tran P, et al. Genetic polymorphisms in methylenetetrahydrofolate reductase and methionine synthase, folate levels in red blood cells, and risk of neural tube defects. Am J Med Genet. 1999;84(2):151-157.  (PubMed)

53. Relton CL, Wilding CS, Pearce MS, et al. Gene-gene interaction in folate-related genes and risk of neural tube defects in a UK population. J Med Genet. 2004;41(4):256-260.  (PubMed)

54. Brody LC, Conley M, Cox C, et al. A polymorphism, R653Q, in the trifunctional enzyme methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase/formyltetrahydrofolate synthetase is a maternal genetic risk factor for neural tube defects: report of the Birth Defects Research Group. Am J Hum Genet. 2002;71(5):1207-1215.  (PubMed)

55. Meng J, Han L, Zhuang B. Association between MTHFD1 polymorphisms and neural tube defect susceptibility. J Neurol Sci. 2015;348(1-2):188-194.  (PubMed)

56. van der Put NM, van den Heuvel LP, Steegers-Theunissen RP, et al. Decreased methylene tetrahydrofolate reductase activity due to the 677C-->T mutation in families with spina bifida offspring. J Mol Med (Berl). 1996;74(11):691-694.  (PubMed)

57. Wilson A, Platt R, Wu Q, et al. A common variant in methionine synthase reductase combined with low cobalamin (vitamin B12) increases risk for spina bifida. Mol Genet Metab. 1999;67(4):317-323.  (PubMed)

58. Gilboa SM, Salemi JL, Nembhard WN, Fixler DE, Correa A. Mortality resulting from congenital heart disease among children and adults in the United States, 1999 to 2006. Circulation. 2010;122(22):2254-2263.  (PubMed)

59. van Beynum IM, Kapusta L, Bakker MK, den Heijer M, Blom HJ, de Walle HE. Protective effect of periconceptional folic acid supplements on the risk of congenital heart defects: a registry-based case-control study in the northern Netherlands. Eur Heart J. 2010;31(4):464-471.  (PubMed)

60. Yin M, Dong L, Zheng J, Zhang H, Liu J, Xu Z. Meta analysis of the association between MTHFR C677T polymorphism and the risk of congenital heart defects. Ann Hum Genet. 2012;76(1):9-16.  (PubMed)

61. Wang W, Wang Y, Gong F, Zhu W, Fu S. MTHFR C677T polymorphism and risk of congenital heart defects: evidence from 29 case-control and TDT studies. PLoS One. 2013;8(3):e58041.  (PubMed)

62. Badovinac RL, Werler MM, Williams PL, Kelsey KT, Hayes C. Folic acid-containing supplement consumption during pregnancy and risk for oral clefts: a meta-analysis. Birth Defects Res A Clin Mol Teratol. 2007;79(1):8-15.  (PubMed)

63. Watkins SE, Meyer RE, Strauss RP, Aylsworth AS. Classification, epidemiology, and genetics of orofacial clefts. Clin Plast Surg. 2014;41(2):149-163.  (PubMed)

64. Zhou Y, Sinnathamby V, Yu Y, et al. Folate intake, markers of folate status and oral clefts: An updated set of systematic reviews and meta-analyses. Birth Defects Res. 2020;112(19):1699-1719.  (PubMed)

65. Boyles AL, Wilcox AJ, Taylor JA, et al. Folate and one-carbon metabolism gene polymorphisms and their associations with oral facial clefts. Am J Med Genet A. 2008;146A(4):440-449.  (PubMed)

66. Boyles AL, Wilcox AJ, Taylor JA, et al. Oral facial clefts and gene polymorphisms in metabolism of folate/one-carbon and vitamin A: a pathway-wide association study. Genet Epidemiol. 2009;33(3):247-255.  (PubMed)

67. Millacura N, Pardo R, Cifuentes L, Suazo J. Effects of folic acid fortification on orofacial clefts prevalence: a meta-analysis. Public Health Nutr. 2017;20(12):2260-2268.  (PubMed)

68. Wilcox AJ. On the importance--and the unimportance--of birthweight. Int J Epidemiol. 2001;30(6):1233-1241.  (PubMed)

69. Fekete K, Berti C, Trovato M, et al. Effect of folate intake on health outcomes in pregnancy: a systematic review and meta-analysis on birth weight, placental weight and length of gestation. Nutr J. 2012;11:75.  (PubMed)

70. Baker PN, Wheeler SJ, Sanders TA, et al. A prospective study of micronutrient status in adolescent pregnancy. Am J Clin Nutr. 2009;89(4):1114-1124.  (PubMed)

71. Lee HA, Park EA, Cho SJ, et al. Mendelian randomization analysis of the effect of maternal homocysteine during pregnancy, as represented by maternal MTHFR C677T genotype, on birth weight. J Epidemiol. 2013;23(5):371-375.  (PubMed)

72. Scholl TO, Johnson WG. Folic acid: influence on the outcome of pregnancy. Am J Clin Nutr. 2000;71(5 Suppl):1295S-1303S.  (PubMed)

73. Vollset SE, Refsum H, Irgens LM, et al. Plasma total homocysteine, pregnancy complications, and adverse pregnancy outcomes: the Hordaland Homocysteine study. Am J Clin Nutr. 2000;71(4):962-968.  (PubMed)

74. Wang XM, Wu HY, Qiu XJ. Methylenetetrahydrofolate reductase (MTHFR) gene C677T polymorphism and risk of preeclampsia: an updated meta-analysis based on 51 studies. Arch Med Res. 2013;44(3):159-168.  (PubMed)

75. Liu C, Liu C, Wang Q, Zhang Z. Supplementation of folic acid in pregnancy and the risk of preeclampsia and gestational hypertension: a meta-analysis. Arch Gynecol Obstet. 2018;298(4):697-704.  (PubMed)

76. Wen SW, Champagne J, Rennicks White R, et al. Effect of folic acid supplementation in pregnancy on preeclampsia: the folic acid clinical trial study. J Pregnancy. 2013;2013:294312.  (PubMed)

77. Wen SW, White RR, Rybak N, et al. Effect of high dose folic acid supplementation in pregnancy on pre-eclampsia (FACT): double blind, phase III, randomised controlled, international, multicentre trial. BMJ. 2018;362:k3478.  (PubMed)

78. Field MS, Stover PJ. Safety of folic acid. Ann N Y Acad Sci. 2018;1414(1):59-71.  (PubMed)

79. Hoxha B, Hoxha M, Domi E, et al. Folic acid and autism: a systematic review of the current state of knowledge. Cells. 2021;10(8):1976.  (PubMed)

80. Crider KS, Cordero AM, Qi YP, Mulinare J, Dowling NF, Berry RJ. Prenatal folic acid and risk of asthma in children: a systematic review and meta-analysis. Am J Clin Nutr. 2013;98(5):1272-1281.  (PubMed)

81. Brown SB, Reeves KW, Bertone-Johnson ER. Maternal folate exposure in pregnancy and childhood asthma and allergy: a systematic review. Nutr Rev. 2014;72(1):55-64.  (PubMed)

82. Lassi ZS, Salam RA, Haider BA, Bhutta ZA. Folic acid supplementation during pregnancy for maternal health and pregnancy outcomes. Cochrane Database Syst Rev. 2013;3:CD006896.  (PubMed)

83. Ding R, Lin S, Chen D. The association of cystathionine beta synthase (CBS) T833C polymorphism and the risk of stroke: a meta-analysis. J Neurol Sci. 2012;312(1-2):26-30.  (PubMed)

84. Seshadri N, Robinson K. Homocysteine, B vitamins, and coronary artery disease. Med Clin North Am. 2000;84(1):215-237, x.  (PubMed)

85. Smith AD, Refsum H. Homocysteine - from disease biomarker to disease prevention. J Intern Med. 2021;290(4):826-854.  (PubMed)

86. Wald DS, Law M, Morris JK. Homocysteine and cardiovascular disease: evidence on causality from a meta-analysis. BMJ. 2002;325(7374):1202.  (PubMed)

87. Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: a meta-analysis. JAMA. 2002;288(16):2015-2022.  (PubMed)

88. Clarke R, Halsey J, Lewington S, et al. Effects of lowering homocysteine levels with B vitamins on cardiovascular disease, cancer, and cause-specific mortality: Meta-analysis of 8 randomized trials involving 37 485 individuals. Arch Intern Med. 2010;170(18):1622-1631.  (PubMed)

89. Clarke R, Halsey J, Bennett D, Lewington S. Homocysteine and vascular disease: review of published results of the homocysteine-lowering trials. J Inherit Metab Dis. 2011;34(1):83-91.  (PubMed)

90. Huang T, Chen Y, Yang B, Yang J, Wahlqvist ML, Li D. Meta-analysis of B vitamin supplementation on plasma homocysteine, cardiovascular and all-cause mortality. Clin Nutr. 2012;31(4):448-454.  (PubMed)

91. Voutilainen S, Rissanen TH, Virtanen J, Lakka TA, Salonen JT. Low dietary folate intake is associated with an excess incidence of acute coronary events: The Kuopio Ischemic Heart Disease Risk Factor Study. Circulation. 2001;103(22):2674-2680.  (PubMed)

92. Brattstrom L. Vitamins as homocysteine-lowering agents. J Nutr. 1996;126(4 Suppl):1276S-1280S.  (PubMed)

93. Rader JI. Folic acid fortification, folate status and plasma homocysteine. J Nutr. 2002;132(8 Suppl):2466S-2470S.  (PubMed)

94. 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(4):806-812.  (PubMed)

95. Malinow MR, Bostom AG, Krauss RM. Homocyst(e)ine, diet, and cardiovascular diseases: a statement for healthcare professionals from the Nutrition Committee, American Heart Association. Circulation. 1999;99(1):178-182.  (PubMed)

96. van Meurs JB, Pare G, Schwartz SM, et al. Common genetic loci influencing plasma homocysteine concentrations and their effect on risk of coronary artery disease. Am J Clin Nutr. 2013;98(3):668-676.  (PubMed)

97. Holmes MV, Newcombe P, Hubacek JA, et al. Effect modification by population dietary folate on the association between MTHFR genotype, homocysteine, and stroke risk: a meta-analysis of genetic studies and randomised trials. Lancet. 2011;378(9791):584-594.  (PubMed)

98. Clarke R, Bennett DA, Parish S, et al. Homocysteine and coronary heart disease: meta-analysis of MTHFR case-control studies, avoiding publication bias. PLoS Med. 2012;9(2):e1001177.  (PubMed)

99. Mosca L, Benjamin EJ, Berra K, et al. Effectiveness-based guidelines for the prevention of cardiovascular disease in women--2011 update: a guideline from the American Heart Association. J Am Coll Cardiol. 2011;57(12):1404-1423.  (PubMed)

100. Park J-H, Saposnik G, Ovbiagele B, Markovic D, Towfighi A. Effect of B-vitamins on stroke risk among individuals with vascular disease who are not on antiplatelets: A meta-analysis. Int J Stroke.11(2):206-211.  (PubMed)

101. Tian T, Yang KQ, Cui JG, Zhou LL, Zhou XL. Folic acid supplementation for stroke prevention in patients with cardiovascular disease. Am J Med Sci. 2017;354(4):379-387.  (PubMed)

102. Wang WW, Wang XS, Zhang ZR, He JC, Xie CL. A meta-analysis of folic acid in combination with anti-hypertension drugs in patients with hypertension and hyperhomocysteinemia. Front Pharmacol. 2017;8:585.  (PubMed)

103. Wang Y, Jin Y, Wang Y, et al. The effect of folic acid in patients with cardiovascular disease: A systematic review and meta-analysis. Medicine (Baltimore). 2019;98(37):e17095.  (PubMed)

104. Li Y, Huang T, Zheng Y, Muka T, Troup J, Hu FB. Folic acid supplementation and the risk of cardiovascular diseases: a meta-analysis of randomized controlled trials. J Am Heart Assoc. 2016;5(8):e003768.  (PubMed)

105. Asbaghi O, Salehpour S, Rezaei Kelishadi M, et al. Folic acid supplementation and blood pressure: a GRADE-assessed systematic review and dose-response meta-analysis of 41,633 participants. Crit Rev Food Sci Nutr. 2023;63(13):1846-1861.  (PubMed)

106. Lorenz MW, Markus HS, Bots ML, Rosvall M, Sitzer M. Prediction of clinical cardiovascular events with carotid intima-media thickness: a systematic review and meta-analysis. Circulation. 2007;115(4):459-467.  (PubMed)

107. Qin X, Xu M, Zhang Y, et al. Effect of folic acid supplementation on the progression of carotid intima-media thickness: a meta-analysis of randomized controlled trials. Atherosclerosis. 2012;222(2):307-313.  (PubMed)

108. de Bree A, van Mierlo LA, Draijer R. Folic acid improves vascular reactivity in humans: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2007;86(3):610-617.  (PubMed)

109. Zamani M, Rezaiian F, Saadati S, et al. The effects of folic acid supplementation on endothelial function in adults: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutr J. 2023;22(1):12.  (PubMed)

110. McNeil CJ, Beattie JH, Gordon MJ, Pirie LP, Duthie SJ. Nutritional B vitamin deficiency disrupts lipid metabolism causing accumulation of proatherogenic lipoproteins in the aorta adventitia of ApoE null mice. Mol Nutr Food Res. 2012;56(7):1122-1130.  (PubMed)

111. Blount BC, Mack MM, Wehr CM, et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci U S A. 1997;94(7):3290-3295.  (PubMed)

112. Narayanan S, McConnell J, Little J, et al. Associations between two common variants C677T and A1298C in the methylenetetrahydrofolate reductase gene and measures of folate metabolism and DNA stability (strand breaks, misincorporated uracil, and DNA methylation status) in human lymphocytes in vivo. Cancer Epidemiol Biomarkers Prev. 2004;13(9):1436-1443.  (PubMed)

113. Rahman L, Voeller D, Rahman M, et al. Thymidylate synthase as an oncogene: a novel role for an essential DNA synthesis enzyme. Cancer Cell. 2004;5(4):341-351.  (PubMed)

114. Hubner RA, Liu JF, Sellick GS, Logan RF, Houlston RS, Muir KR. Thymidylate synthase polymorphisms, folate and B-vitamin intake, and risk of colorectal adenoma. Br J Cancer. 2007;97(10):1449-1456.  (PubMed)

115. Desmoulin SK, Wang L, Polin L, et al. Functional loss of the reduced folate carrier enhances the antitumor activities of novel antifolates with selective uptake by the proton-coupled folate transporter. Mol Pharmacol. 2012;82(4):591-600.  (PubMed)

116. 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(1):21-38.  (PubMed)

117. Butrum RR, Clifford CK, Lanza E. NCI dietary guidelines: rationale. Am J Clin Nutr. 1988;48(3 Suppl):888-895.  (PubMed)

118. World Cancer Research Fund/American Institute for Cancer Research. Diet, Nutrition, Physical Activity, and Cancer: a Global Perspective 2018. Available at: https://www.wcrf.org/wp-content/uploads/2021/02/Summary-of-Third-Expert-Report-2018.pdf. Accessed 12/21/2023.

119. Crider KS, Bailey LB, Berry RJ. Folic acid food fortification-its history, effect, concerns, and future directions. Nutrients. 2011;3(3):370-384.  (PubMed)

120. Qin X, Cui Y, Shen L, et al. Folic acid supplementation and cancer risk: A meta-analysis of randomized controlled trials. Int J Cancer. 2013;133(5):1033-1041.  (PubMed)

121. Vollset SE, Clarke R, Lewington S, et al. Effects of folic acid supplementation on overall and site-specific cancer incidence during the randomised trials: meta-analyses of data on 50,000 individuals. Lancet. 2013;381(9871):1029-1036.  (PubMed)

122. National Toxicology Program. Identifying research needs for assessing safe use of high intakes of folic acid. NTP Monograph. Research Triangle Park, NC: National Toxicology Program, 2015.  

123. Kim DH, Smith-Warner SA, Spiegelman D, et al. Pooled analyses of 13 prospective cohort studies on folate intake and colon cancer. Cancer Causes Control. 2010;21(11):1919-1930.  (PubMed)

124. Gibson TM, Weinstein SJ, Pfeiffer RM, et al. Pre- and postfortification intake of folate and risk of colorectal cancer in a large prospective cohort study in the United States. Am J Clin Nutr. 2011;94(4):1053-1062.  (PubMed)

125. Stevens VL, McCullough ML, Sun J, Jacobs EJ, Campbell PT, Gapstur SM. High levels of folate from supplements and fortification are not associated with increased risk of colorectal cancer. Gastroenterology. 2011;141(1):98-105, 105 e101.  (PubMed)

126. Zschabitz S, Cheng TY, Neuhouser ML, 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(2):332-343.  (PubMed)

127. Wang F, Wu K, Li Y, et al. Association of folate intake and colorectal cancer risk in the postfortification era in US women. Am J Clin Nutr. 2021;114(1):49-58.  (PubMed)

128. Kennedy DA, Stern SJ, Moretti M, et al. Folate intake and the risk of colorectal cancer: a systematic review and meta-analysis. Cancer Epidemiol. 2011;35(1):2-10.  (PubMed)

129. Kim YI. Folate: a magic bullet or a double edged sword for colorectal cancer prevention? Gut. 2006;55(10):1387-1389.  (PubMed)

130. Paspatis GA, Kalafatis E, Oros L, Xourgias V, Koutsioumpa P, Karamanolis DG. Folate status and adenomatous colonic polyps. A colonoscopically controlled study. Dis Colon Rectum. 1995;38(1):64-67; discussion 67-68.  (PubMed)

131. Jaszewski R, Misra S, Tobi M, et al. Folic acid supplementation inhibits recurrence of colorectal adenomas: a randomized chemoprevention trial. World J Gastroenterol. 2008;14(28):4492-4498.  (PubMed)

132. Wu K, Platz EA, Willett WC, et al. A randomized trial on folic acid supplementation and risk of recurrent colorectal adenoma. Am J Clin Nutr. 2009;90(6):1623-1631.  (PubMed)

133. Logan RF, Grainge MJ, Shepherd VC, Armitage NC, Muir KR, ukCAP Trial Group. Aspirin and folic acid for the prevention of recurrent colorectal adenomas. Gastroenterology. 2008;134(1):29-38.  (PubMed)

134. Figueiredo JC, Mott LA, Giovannucci E, et al. Folic acid and prevention of colorectal adenomas: a combined analysis of randomized clinical trials. Int J Cancer. 2011;129(1):192-203.  (PubMed)

135. van Wijngaarden JP, Dhonukshe-Rutten RA, van Schoor NM, et al. Rationale and design of the B-PROOF study, a randomized controlled trial on the effect of supplemental intake of vitamin B12 and folic acid on fracture incidence. BMC Geriatr. 2011;11:80.  (PubMed)

136. Oliai Araghi S, Kiefte-de Jong JC, van Dijk SC, et al. Folic acid and vitamin B12 supplementation and the risk of cancer: long-term follow-up of the B Vitamins for the Prevention of Osteoporotic Fractures (B-PROOF) trial. Cancer Epidemiol Biomarkers Prev. 2019;28(2):275-282.  (PubMed)

137. Kennedy DA, Stern SJ, Matok I, et al. Folate intake, MTHFR polymorphisms, and the risk of colorectal cancer: a systematic review and meta-analysis. J Cancer Epidemiol. 2012;2012:952508.  (PubMed)

138. Ding W, Zhou DL, Jiang X, Lu LS. Methionine synthase A2756G polymorphism and risk of colorectal adenoma and cancer: evidence based on 27 studies. PLoS One. 2013;8(4):e60508.  (PubMed)

139. Haerian MS, Haerian BS, Molanaei S, et al. MTRR rs1801394 and its interaction with MTHFR rs1801133 in colorectal cancer: a case-control study and meta-analysis. Pharmacogenomics. 2017;18(11):1075-1084.  (PubMed)

140. Nan H, Lee JE, Rimm EB, Fuchs CS, Giovannucci EL, Cho E. Prospective study of alcohol consumption and the risk of colorectal cancer before and after folic acid fortification in the United States. Ann Epidemiol. 2013;23(9):558-563.  (PubMed)

141. Slattery ML, Potter JD, Samowitz W, Schaffer D, Leppert M. Methylenetetrahydrofolate reductase, diet, and risk of colon cancer. Cancer Epidemiol Biomarkers Prev. 1999;8(6):513-518.  (PubMed)

142. Ma J, Stampfer MJ, Giovannucci E, et al. Methylenetetrahydrofolate reductase polymorphism, dietary interactions, and risk of colorectal cancer. Cancer Res. 1997;57(6):1098-1102.  (PubMed)

143. Larsson SC, Giovannucci E, Wolk A. Folate and risk of breast cancer: a meta-analysis. J Natl Cancer Inst. 2007;99(1):64-76.  (PubMed)

144. Ren X, Xu P, Zhang D, et al. Association of folate intake and plasma folate level with the risk of breast cancer: a dose-response meta-analysis of observational studies. Aging (Albany NY). 2020;12(21):21355-21375.  (PubMed)

145. Zeng J, Wang K, Ye F, et al. Folate intake and the risk of breast cancer: an up-to-date meta-analysis of prospective studies. Eur J Clin Nutr. 2019;73(12):1657-1660.  (PubMed)

146. Brooks PJ, Zakhari S. Moderate alcohol consumption and breast cancer in women: from epidemiology to mechanisms and interventions. Alcohol Clin Exp Res. 2013;37(1):23-30.  (PubMed)

147. Rohan TE, Jain MG, Howe GR, Miller AB. Dietary folate consumption and breast cancer risk. J Natl Cancer Inst. 2000;92(3):266-269.  (PubMed)

148. Sellers TA, Kushi LH, Cerhan JR, et al. Dietary folate intake, alcohol, and risk of breast cancer in a prospective study of postmenopausal women. Epidemiology. 2001;12(4):420-428.  (PubMed)

149. Zhang S, Hunter DJ, Hankinson SE, et al. A prospective study of folate intake and the risk of breast cancer. JAMA. 1999;281(17):1632-1637.  (PubMed)

150. Fagherazzi G, Vilier A, Boutron-Ruault MC, Mesrine S, Clavel-Chapelon F. Alcohol consumption and breast cancer risk subtypes in the E3N-EPIC cohort. Eur J Cancer Prev. 2015;24(3):209-214.  (PubMed)

151. Freudenheim JL. Alcohol's effects on breast cancer in women. Alcohol Res. 2020;40(2):11.  (PubMed)

152. Wang J, Wang B, Bi J, Di J. The association between two polymorphisms in the TYMS gene and breast cancer risk: a meta-analysis. Breast Cancer Res Treat. 2011;128(1):203-209.  (PubMed)

153. Weiner AS, Boyarskikh UA, Voronina EN, et al. Polymorphisms in the folate-metabolizing genes MTR, MTRR, and CBS and breast cancer risk. Cancer Epidemiol. 2012;36(2):e95-e100.  (PubMed)

154. Mo W, Ding Y, Zheng Y, Zou D, Ding X. Associations between folate metabolism enzyme polymorphisms and breast cancer: A meta-analysis. Breast J. 2020;26(3):484-487.  (PubMed)

155. Linabery AM, Johnson KJ, Ross JA. Childhood cancer incidence trends in association with US folic acid fortification (1986-2008). Pediatrics. 2012;129(6):1125-1133.  (PubMed)

156. Wan Ismail WR, Abdul Rahman R, Rahman NAA, Atil A, Nawi AM. The protective effect of maternal folic acid supplementation on childhood cancer: a systematic review and meta-analysis of case-control studies. J Prev Med Public Health. 2019;52(4):205-213.  (PubMed)

157. Frikha R. Assessment of the relationship between methylenetetrahydrofolate reductase polymorphism and acute lymphoblastic leukemia: Evidence from an updated meta-analysis. J Oncol Pharm Pract. 2020;26(7):1598-1610.  (PubMed)

158. Matthews KA, Xu W, Gaglioti AH, et al. Racial and ethnic estimates of Alzheimer's disease and related dementias in the United States (2015-2060) in adults aged ≥65 years. Alzheimers Dement. 2019;15(1):17-24.  (PubMed)

159. Hughes TF, Andel R, Small BJ, et al. Midlife fruit and vegetable consumption and risk of dementia in later life in Swedish twins. Am J Geriatr Psychiatry. 2010;18(5):413-420.  (PubMed)

160. Corrada MM, Kawas CH, Hallfrisch J, Muller D, Brookmeyer R. Reduced risk of Alzheimer's disease with high folate intake: the Baltimore Longitudinal Study of Aging. Alzheimers Dement. 2005;1(1):11-18.  (PubMed)

161. Weir DG, Scott JM. Brain function in the elderly: role of vitamin B12 and folate. Br Med Bull. 1999;55(3):669-682.  (PubMed)

162. Faux NG, Ellis KA, Porter L, et al. Homocysteine, vitamin B12, and folic acid levels in Alzheimer's disease, mild cognitive impairment, and healthy elderly: baseline characteristics in subjects of the Australian Imaging Biomarker Lifestyle study. J Alzheimers Dis. 2011;27(4):909-922.  (PubMed)

163. Van Dam F, Van Gool WA. Hyperhomocysteinemia and Alzheimer's disease: A systematic review. Arch Gerontol Geriatr. 2009;48(3):425-430.  (PubMed)

164. Morris MC, Evans DA, Bienias JL, et al. Dietary folate and vitamin B12 intake and cognitive decline among community-dwelling older persons. Arch Neurol. 2005;62(4):641-645.  (PubMed)

165. Morris MC, Evans DA, Schneider JA, Tangney CC, Bienias JL, Aggarwal NT. Dietary folate and vitamins B-12 and B-6 not associated with incident Alzheimer's disease. J Alzheimers Dis. 2006;9(4):435-443.  (PubMed)

166. Qin B, Xun P, Jacobs DR, Jr., et al. Intake of niacin, folate, vitamin B-6, and vitamin B-12 through young adulthood and cognitive function in midlife: the Coronary Artery Risk Development in Young Adults (CARDIA) study. Am J Clin Nutr. 2017;106(4):1032-1040.  (PubMed)

167. Agnew-Blais JC, Wassertheil-Smoller S, Kang JH, et al. Folate, vitamin B-6, and vitamin B-12 intake and mild cognitive impairment and probable dementia in the Women's Health Initiative Memory Study. J Acad Nutr Diet. 2015;115(2):231-241.  (PubMed)

168. Sheng LT, Jiang YW, Pan XF, et al. Association between dietary intakes of B vitamins in midlife and cognitive impairment in late-life: the Singapore Chinese Health Study. J Gerontol A Biol Sci Med Sci. 2020;75(6):1222-1227.  (PubMed)

169. Zanin Palchetti C, Gomes Goncalves N, Vidal Ferreira N, et al. Dietary folate intake and its association with longitudinal changes in cognition function. Clin Nutr ESPEN. 2023;55:332-339.  (PubMed)

170. Wald DS, Kasturiratne A, Simmonds M. Serum homocysteine and dementia: meta-analysis of eight cohort studies including 8669 participants. Alzheimers Dement. 2011;7(4):412-417.  (PubMed)

171. Ho RC, Cheung MW, Fu E, et al. Is high homocysteine level a risk factor for cognitive decline in elderly? A systematic review, meta-analysis, and meta-regression. Am J Geriatr Psychiatry. 2011;19(7):607-617.  (PubMed)

172. McCaddon A, Miller JW. Assessing the association between homocysteine and cognition: reflections on Bradford Hill, meta-analyses, and causality. Nutr Rev. 2015;73(10):723-735.  (PubMed)

173. Smith AD, Refsum H. Homocysteine, B vitamins, and cognitive impairment. Annu Rev Nutr. 2016;36:211-239.  (PubMed)

174. Smith AD, Refsum H, Bottiglieri T, et al. Homocysteine and dementia: an International Consensus Statement. J Alzheimers Dis. 2018;62(2):561-570.  (PubMed)

175. Rutjes AW, Denton DA, Di Nisio M, et al. Vitamin and mineral supplementation for maintaining cognitive function in cognitively healthy people in mid and late life. Cochrane Database Syst Rev. 2018;12(12):CD011906.  (PubMed)

176. Wang Z, Zhu W, Xing Y, Jia J, Tang Y. B vitamins and prevention of cognitive decline and incident dementia: a systematic review and meta-analysis. Nutr Rev. 2022;80(4):931-949.  (PubMed)

177. Smith AD, Smith SM, de Jager CA, et al. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PLoS One. 2010;5(9):e12244.  (PubMed)

178. Douaud G, Refsum H, de Jager CA, et al. Preventing Alzheimer's disease-related gray matter atrophy by B-vitamin treatment. Proc Natl Acad Sci U S A. 2013;110(23):9523-9528.  (PubMed)

179. Ma F, Wu T, Zhao J, et al. Folic acid supplementation improves cognitive function by reducing the levels of peripheral inflammatory cytokines in elderly Chinese subjects with MCI. Sci Rep. 2016;6:37486.  (PubMed)

180. Ma F, Wu T, Zhao J, et al. Effects of 6-month folic acid supplementation on cognitive function and blood biomarkers in mild cognitive impairment: a randomized controlled trial in China. J Gerontol A Biol Sci Med Sci. 2016;71(10):1376-1383.  (PubMed)

181. Ma F, Li Q, Zhou X, et al. Effects of folic acid supplementation on cognitive function and Abeta-related biomarkers in mild cognitive impairment: a randomized controlled trial. Eur J Nutr. 2019;58(1):345-356.  (PubMed)

182. Watkins D, Rosenblatt DS. Update and new concepts in vitamin responsive disorders of folate transport and metabolism. J Inherit Metab Dis. 2012;35(4):665-670.  (PubMed)

183. Zhao R, Min SH, Qiu A, et al. The spectrum of mutations in the PCFT gene, coding for an intestinal folate transporter, that are the basis for hereditary folate malabsorption. Blood. 2007;110(4):1147-1152.  (PubMed)

184. Borzutzky A, Crompton B, Bergmann AK, et al. Reversible severe combined immunodeficiency phenotype secondary to a mutation of the proton-coupled folate transporter. Clin Immunol. 2009;133(3):287-294.  (PubMed)

185. Goldman ID. Hereditary folate malabsorption. In: Adam MP, Mirzaa GM, Pagon RA, et al., eds. GeneReviews((R)). Seattle (WA); 1993.  (PubMed)

186. Sofer Y, Harel L, Sharkia M, Amir J, Schoenfeld T, Straussberg R. Neurological manifestations of folate transport defect: case report and review of the literature. J Child Neurol. 2007;22(6):783-786.  (PubMed)

187. Diop-Bove N, Kronn D, Goldman ID. Hereditary folate malabsorption. In: Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT, Stephens K, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2008.  (PubMed)

188. Lubout CMA, Goorden SMI, van den Hurk K, et al. Successful treatment of hereditary folate malabsorption with intramuscular folinic acid. Pediatr Neurol. 2020;102:62-66.  (PubMed)

189. Online Mendelian Inheritance in Man, OMIM®. Johns Hopkins University, Baltimore, MD. MIM Number: 229050: 7/9/2016: . World Wide Web URL: https://www.omim.org/entry/229050.

190. Frye RE, Sequeira JM, Quadros EV, James SJ, Rossignol DA. Cerebral folate receptor autoantibodies in autism spectrum disorder. Mol Psychiatry. 2013;18(3):369-381.  (PubMed)

191. Grapp M, Just IA, Linnankivi T, et al. Molecular characterization of folate receptor 1 mutations delineates cerebral folate transport deficiency. Brain. 2012;135(Pt 7):2022-2031.  (PubMed)

192. Kanmaz S, Simsek E, Yilmaz S, Durmaz A, Serin HM, Gokben S. Cerebral folate transporter deficiency: a potentially treatable neurometabolic disorder. Acta Neurol Belg. 2023;123(1):121-127.  (PubMed)

193. Ramaekers VT, Quadros EV. Cerebral folate deficiency syndrome: early diagnosis, intervention and treatment strategies. Nutrients. 2022;14(15):3096.  (PubMed)

194. Masingue M, Benoist JF, Roze E, et al. Cerebral folate deficiency in adults: A heterogeneous potentially treatable condition. J Neurol Sci. 2019;396:112-118.  (PubMed)

195. Ramaekers VT, Blau N, Sequeira JM, Nassogne MC, Quadros EV. Folate receptor autoimmunity and cerebral folate deficiency in low-functioning autism with neurological deficits. Neuropediatrics. 2007;38(6):276-281.  (PubMed)

196. Ramaekers VT, Hausler M, Opladen T, Heimann G, Blau N. Psychomotor retardation, spastic paraplegia, cerebellar ataxia and dyskinesia associated with low 5-methyltetrahydrofolate in cerebrospinal fluid: a novel neurometabolic condition responding to folinic acid substitution. Neuropediatrics. 2002;33(6):301-308.  (PubMed)

197. Banka S, Blom HJ, Walter J, et al. Identification and characterization of an inborn error of metabolism caused by dihydrofolate reductase deficiency. Am J Hum Genet. 2011;88(2):216-225.  (PubMed)

198. Cario H, Smith DE, Blom H, et al. Dihydrofolate reductase deficiency due to a homozygous DHFR mutation causes megaloblastic anemia and cerebral folate deficiency leading to severe neurologic disease. Am J Hum Genet. 2011;88(2):226-231.  (PubMed)

199. US Food & Drug Admininstration. FDA News Release. FDA approves folic acid fortification of corn masa flour. April 14, 2016. Available at: https://www.fda.gov/news-events/press-announcements/fda-approves-folic-acid-fortification-corn-masa-flour. Accessed 7/17/2023.

200. Folate. In: Hendler SS, Rorvik, D.R., ed. PDR for Nutritional Supplements. 2nd ed. Montvale: Physicians' Desk Reference Inc.; 2008.  

201. US Department of Health and Human Services, National Institutes of Health, Office of Dietary Supplements. Dietary Supplement Label Database (DSLD). [Internet]. [cited 7/17/2023]. Available from: https://dsld.nlm.nih.gov/dsld/.

202. Wiesinger H, Eydeler U, Richard F, et al. Bioequivalence evaluation of a folate-supplemented oral contraceptive containing ethinylestradiol/drospirenone/levomefolate calcium versus ethinylestradiol/drospirenone and levomefolate calcium alone. Clin Drug Investig. 2012;32(10):673-684.  (PubMed)

203. Tinker SC, Cogswell ME, Devine O, Berry RJ. Folic acid intake among U.S. women aged 15-44 years, National Health and Nutrition Examination Survey, 2003-2006. Am J Prev Med. 2010;38(5):534-542.  (PubMed)

204. 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(6):1790-1795.  (PubMed)

205. Pfeiffer CM, Sternberg MR, Fazili Z, et al. Unmetabolized folic acid is detected in nearly all serum samples from US children, adolescents, and adults. J Nutr. 2015;145(3):520-531.  (PubMed)

206. Murphy MSQ, Muldoon KA, Sheyholislami H, et al. Impact of high-dose folic acid supplementation in pregnancy on biomarkers of folate status and 1-carbon metabolism: An ancillary study of the Folic Acid Clinical Trial (FACT). Am J Clin Nutr. 2021;113(5):1361-1371.  (PubMed)

207. Pentieva K, Selhub J, Paul L, et al. Evidence from a randomized trial that exposure to supplemental folic acid at recommended levels during pregnancy does not lead to increased unmetabolized folic acid concentrations in maternal or cord blood. J Nutr. 2016;146(3):494-500.  (PubMed)

208. Morris MS, Jacques PF, Rosenberg IH, Selhub J. Folate and vitamin B-12 status in relation to anemia, macrocytosis, and cognitive impairment in older Americans in the age of folic acid fortification. Am J Clin Nutr. 2007;85(1):193-200.  (PubMed)

209. Morris MS, Jacques PF, Rosenberg IH, Selhub J. Circulating unmetabolized folic acid and 5-methyltetrahydrofolate in relation to anemia, macrocytosis, and cognitive test performance in American seniors. Am J Clin Nutr. 2010;91(6):1733-1744.  (PubMed)

210. Troen AM, Mitchell B, Sorensen B, et al. Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr. 2006;136(1):189-194.  (PubMed)

211. Best KP, Green TJ, Sulistyoningrum DC, et al. Maternal late-pregnancy serum unmetabolized folic acid concentrations are not associated with infant allergic disease: a prospective cohort study. J Nutr. 2021;151(6):1553-1560.  (PubMed)

212. Husebye ESN, Wendel AWK, Gilhus NE, Riedel B, Bjork MH. Plasma unmetabolized folic acid in pregnancy and risk of autistic traits and language impairment in antiseizure medication-exposed children of women with epilepsy. Am J Clin Nutr. 2022;115(5):1432-1440.  (PubMed)

213. Tam C, O'Connor D, Koren G. Circulating unmetabolized folic acid: relationship to folate status and effect of supplementation. Obstet Gynecol Int. 2012;2012:485179.  (PubMed)

214. Apeland T, Mansoor MA, Strandjord RE. Antiepileptic drugs as independent predictors of plasma total homocysteine levels. Epilepsy Res. 2001;47(1-2):27-35.  (PubMed)

215. Wilson SM, Bivins BN, Russell KA, Bailey LB. Oral contraceptive use: impact on folate, vitamin B(6), and vitamin B(1)(2) status. Nutr Rev. 2011;69(10):572-583.  (PubMed)