ArticlePDF Available

Folate and Inflammation – links between folate and features of inflammatory conditions

Authors:

Abstract and Figures

Folate serves as a cofactor for one-carbon (1C) transfer reactions. These reactions are involved in the synthesis of DNA nucleotides, the amino acid methionine, and in the regulation of homocysteine (Hcy) levels. Emerging evidence suggests that these reactions have roles in the development and maintenance of inflammatory responses, with optimal folate availability having key importance in preventing endothelial dysfunction and DNA instability. Low folate levels are commonly observed in chronic inflammatory diseases, indicating that inadequate folate may be involved in the pathogenesis of inflammatory conditions or that chronic inflammation increases folate requirements. These findings highlight folate interventions as a potential treatment in inflammatory disorders. However, current understanding of folate and its influence on inflammatory phenotypes is limited. Evidence indicates that the relationship between folate and inflammation is dependent on several factors, including the timing of intervention, dosage, and interaction with environment and genes. These factors require further investigation before recommendations for folate intake can be made for the prevention and treatment of inflammation. This review outlines the emerging role of folate in inflammation and key factors that may influence this relationship.
Content may be subject to copyright.
Contents lists available at ScienceDirect
Journal of Nutrition & Intermediary Metabolism
journal homepage: www.elsevier.com/locate/jnim
Folate and Inammation links between folate and features of inammatory
conditions
Patrice Jones
a,b,
, Mark Lucock
a
, Christopher J. Scarlett
a
, Martin Veysey
a,c
, Emma L. Beckett
a,b
a
School of Environmental & Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
b
Public Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
c
Hull York Medical School, University of Hull, Hull, UK
HIGHLIGHTS
Optimal folate levels may prevent endothelial dysfunction in inammatory diseases.
Folate may also alter inammatory responses via DNA methylation and synthesis processes.
The link between folate and inammation varies based on several factors, such as timing of intervention.
Further studies are needed before making folate intake recommendations around inammation.
ARTICLE INFO
Keywords:
Folate
Inammation
Endothelial dysfunction
DNA methylation
DNA synthesis
ABSTRACT
Folate serves as a cofactor for one-carbon (1C) transfer reactions. These reactions are involved in the synthesis of
DNA nucleotides, the amino acid methionine, and in the regulation of homocysteine (Hcy) levels. Emerging
evidence suggests that these reactions have roles in the development and maintenance of inammatory re-
sponses, with optimal folate availability having key importance in preventing endothelial dysfunction and DNA
instability. Low folate levels are commonly observed in chronic inammatory diseases, indicating that in-
adequate folate may be involved in the pathogenesis of inammatory conditions or that chronic inammation
increases folate requirements. These ndings highlight folate interventions as a potential treatment in in-
ammatory disorders. However, current understanding of folate and its inuence on inammatory phenotypes is
limited. Evidence indicates that the relationship between folate and inammation is dependent on several
factors, including the timing of intervention, dosage, and interaction with environment and genes. These factors
require further investigation before recommendations for folate intake can be made for the prevention and
treatment of inammation. This review outlines the emerging role of folate in inammation and key factors that
may inuence this relationship.
1. Introduction
Folate is a B vitamin derived from natural sources, such as green
leafy vegetables, or from fortied foods or supplements. Folate-related
roles within the human body are reliant on reduced folate compounds
(i.e., tetrahydrofolates (THF)) holding and donating one-carbon (IC)
units for 1C transfer reactions. These reactions are required in the
synthesis of DNA nucleotides and the amino acid methionine, and the
subsequent regulation of homocysteine (Hcy) levels [1].
Folate-dependent 1C reactions have been extensively studied in the
context of cardiovascular disease and cancers, with increased risk of
these diseases in those with inadequate folate status [1]. In addition to
these well-established roles, interest in folate as a contributory factor in
inammation is emerging. This interest follows studies demonstrating
cardiovascular-related outcomes and chronic inammatory diseases
share characteristics which may relate to folate status, such as en-
dothelial dysfunction [2,3]. Endothelial dysfunction is dened as an
impairment in vasomotor tone regulation via imbalanced levels of va-
sodilator and vasoconstrictors, which results in the endothelium
shifting towards a pro-inammatory state [4,5]. Folate may prevent
endothelium dysfunction by maintaining levels of Hcy and vasodilator,
nitric oxide (NO) [46]. In addition, a link between chronic in-
ammatory conditions such as inammatory bowel diseases and ele-
vated risk of carcinomas is well-established, and likely involves the role
https://doi.org/10.1016/j.jnim.2019.100104
Received 29 June 2019; Received in revised form 31 October 2019; Accepted 6 November 2019
Corresponding author. School of Environmental & Life Sciences, University of Newcastle, Ourimbah, NSW, Australia.
E-mail address: Patrice.Jones@uon.edu.au (P. Jones).
Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
Available online 09 November 2019
2352-3859/ © 2019 Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
of folate in regulating DNA stability via mechanisms related to DNA
methylation, synthesis and repair [7,8].
Observational evidence suggests a relationship exists between folate
levels and risk of inammatory disease. Low serum folate and elevated
Hcy levels are commonly reported in patients with chronic in-
ammatory disorders, such as inammatory bowel disease and rheu-
matoid arthritis [9,10]. However, results of in vivo and in vitro studies
examining folate supplementation in inammation have been incon-
sistent. Interpretation of these studies is further complicated by several
factors that may alter the inuence folate has in inammation, such as
the timing of intervention, folate dosage, and interaction with en-
vironmental and genetic factors. These factors require further con-
sideration before recommendations for folate intake can be made for
the prevention and/or treatment of inammation.
This review outlines emerging roles of folate in inammation, en-
compassing regulation of endothelial dysfunction, via both Hcy-de-
pendent and Hcy-independent mechanisms, and the maintenance of
DNA stability through DNA methylation, synthesis and repair reactions.
Key factors that may modulate the activity of folate in these processes
are also discussed.
2. Regulation of Hcy
Hcy is a non-essential amino acid produced as an intermediate
product in the synthesis of essential amino acids methionine and cy-
steine. At elevated levels, Hcy can cause endothelial dysfunction, re-
sulting in a shift towards a pro-inammatory state [4].
Folate may modulate Hcy-induced endothelial dysfunction through
its role as a major regulator of Hcy. Folate, in the form of 5-methylTHF,
is used in the remethylation of Hcy to methionine (Fig. 1). This process
involves the irreversible action of 5,10-methylenetetrahydrofolate re-
ductase (MTHFR) in reducing 5,10-methyleneTHF to 5-methylTHF, and
then B
12
-dependent methionine synthase utilising 5-methylTHF as a
methyl donor in the remethylation of homocysteine to methionine
[11,12].
Hcy-induced endothelial dysfunction has been extensively studied
in atherosclerosis but is increasingly considered in other inammatory
disorders, such as diabetes (types 1 and 2) and inammatory bowel
disease, which also features endothelium dysfunction [2,3]. The major
mechanism by which Hcy induces endothelial dysfunction is through
decreasing production of the vasodilator NO, which may occur via
several mechanisms [4,5,1319].
NO is the major vasodilator within the endothelium and is produced
from L-arginine via endothelial nitric oxide synthase (eNOS). The ac-
tivity of eNOS may be hindered by actions of Hcy in increasing levels of
reactive oxygen species (ROS) and decreasing levels of eNOS cofactor,
tetrahydrobiopterin (BH4). Hcy can cause ROS production through
auto-oxidation of its sulfhydryl group [15] or through upregulating
ROS-generating NADPH oxidases [5,13,14]. Increased ROS production
leads to decreased bioavailability of BH4, which is readily oxidised to 7,
8-dihydrobiopterin (BH2) in the circulation [4,16]. Decreased BH4
bioavailability can further increase oxidative stress, where, in the ab-
sence of BH4, eNOS uncoupling occurs and eNOS produces the super-
oxide radical instead of nitric oxide [17].
Hcy may impact NO production through several other routes,
however these are less well-characterised. Endothelial cells supple-
mented in vitro with Hcy (0.52.5 nM) showed dose-dependent de-
creases in the expression of L-arginine transporters [18], suggesting
that Hcy may also inuence NO production via regulation of L-arginine
cell uptake. Hcy may also regulate eNOS activity by inuencing levels
of eNOS inhibitor, asymmetric dimethyl arginine (ADMA) [19]. In an in
vitro study, elevated Hcy caused increased ADMA levels and decreased
NO production via suppression of ADMA inhibitor dimethylarginine
dimethylaminohydrolase [19]. Impaired NO production is the major
mechanism by which Hcy may cause endothelial dysfunction, with
further roles of elevated Hcy levels in decreasing hydrogen sulphide
production and antioxidant activity, and increasing lipid peroxidation
also inuencing factors [4,5].
3. Hcy-independent regulation of NO & ROS production
Folate has actions in regulating NO & ROS production that are in-
dependent of roles in regulating Hcy levels. These actions are not tra-
ditional 1C transfer reactions but arise due to structural similarities
between folates and biopterin compounds (i.e., between BH4 and THF,
and BH2 and DHF) [6,2023].
As outlined, eNOS cofactor BH4 is readily oxidised to BH2 in the
circulation. Folate-dependent dihydrofolate reductase (DHFR) has a
role in reducing BH2 back to BH4 via the BH4 salvage pathway (Fig. 2)
[20,21]. This pathway has importance in maintaining an adequate
BH4:BH2 ratio, with DHFR inhibition shown to reduce BH4 and in-
crease BH2 levels in vitro, leading to enzymatic uncoupling of eNOS and
increased ROS production [20,21].
Due to structural similarities between folates and biopterins, in the
same way, BH2 may interact with DHFR, folate compounds are found to
interact with eNOS [22]. 5-methylTHF is shown to bind to the active
site of eNOS [22], and mimic the BH4's action in inducing NO pro-
duction and reducing ROS production [22,24]. Through these me-
chanisms, folate supplementation (50 μM) has been found to increase
NO production in an in vitro study examining human pulmonary artery
endothelial cells under hypoxia [25]. This nding has been replicated
in human subjects, with local perfusion of folate (5 mM of 5-MTHF) into
the dermal space, and folate supplementation (6 mg/day) over 6 weeks
both shown to improve in NO-dependent vasodilation and endothelial
function in healthy older adults [25,26].
In addition to the roles of folate in regulating ROS production in-
directly via prevention of eNOS uncoupling, folate may reduce oxida-
tive stress by acting directly as an antioxidant [27,28]. Folic acid and
reduced folate forms (THF and 5-MTHF) have shown ROS scavenging
Fig. 1. Folate-dependent remethylation of Hcy to methionine.
The remethylation of Hcy involves the irreversible action of MTHFR in reducing
5,10-methyleneTHF to 5-methylTHF to be used by B
12
-dependent MTR in the
remethylation of Hcy to methionine. This process has dual importance in reg-
ulating Hcy and methionine levels. Hcy at elevated levels may cause endothelial
dysfunction while methionine is the substrate for SAM, a universal methyl
donor used in DNA methylation.
Abbreviations; THF; tetrahydrofolate, MTHFR; 5,10methyleneTHF reductase,
MTR; methionine synthase, Hcy; homocysteine, Met; methionine, SAM; S-ade-
nosyl methionine, MAT; adenosylmethionine synthetase, SAH; S-adenosyl hcy,
SAHH; S-adenosylhomocysteine hydrolase.
Created with BioRender.
P. Jones, et al. Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
2
activity in vitro [27,28]. However, whether this activity has biological
relevance remains under question, with it dicult to examine the direct
and indirect eects of folate on ROS production separately in in vivo
studies [6].
4. Maintaining DNA methylation
Methionine is produced through folate-dependent Hcy remethyla-
tion and is the substrate for S-adenosyl methionine (SAM), a universal
methyl donor used in methyl transferase processes such as DNA me-
thylation (Fig. 1)[12,29]. Through roles in inuencing SAM levels,
folate may modulate the bidirectional relationship between DNA me-
thylation and inammation, where hypomethylation of inammation-
related genes may predispose inammatory diseases [3032], and in-
ammation may promote DNA methylation processes [3335]. DNA
methylation changes are most dynamic during development and as
such, a consistent research interest has centred on the potential impact
dietary folate may have in utero on DNA methylation and future disease
risk [36]. Of importance to inammation, are several animal studies
that demonstrate maternal folate intake may inuence the risk of
obesity and colitis in ospring [3739].
Obesity is a low-grade inammatory disorder and a further risk
factor for chronic inammatory diseases [40]. A relationship is evident
between maternal weight gain and increased weight in children
[41,42], with these ndings replicated in several murine models
[3032]. Ospring of dams fed high fat obesogenic diets during preg-
nancy and lactation showed increased adiposity as adults. These o-
spring also possess increased insulin resistance and levels of pro-in-
ammatory cytokines [30,31,43], with this shown to increase
susceptibility to colitis in one model [43]. In a murine model, supple-
mentation of maternal diets with methyl donors (15 mg folic acid and
15 g choline/betaine/kg diet) ameliorated the eect of a high fat ma-
ternal diet on weight in ospring [37]. A further study in sheep found
the maternal restriction of methyl donors resulted in heavier adult
ospring that possessed altered immune responses and insulin re-
sistance [38]. In a murine model of colitis, the ospring of dams fed
methyl-donor decient diets had more severe colitis and overexpression
of pro-inammatory pathways [39]. These ndings indicate that folate
status can inuence the relationship between DNA methylation and
inammatory phenotype.
Inammation is shown to induce DNA methylation changes in a
multitude of in vitro and in vivo studies [4450]. In response to in-
ammation, the expression of DNA methyltransferases (DNMTs), S-
adenosylmethionine synthetase, and S-adenosylhomocysteine
hydrolase (SAHH) involved in regulating DNA methylation and me-
thionine production is increased [3335]. This results in substantial
increases in DNA methylation that may exacerbate inammatory re-
sponses and promote tumorigenesis [4450]. Increases in folate have
been shown to be associated with increases in DNMT expression in
human umbilical vein endothelial cells [51], with an increase in pro-
inammatory mediators reported in an murine model of folate re-
striction [52]. These ndings indicate inammatory responses may be
enhanced in cases of folate deciency [51,52]. In a murine model of
colitis, inhibition of cellular methylation via SAHH inhibition lead to
disease exacerbation, with intraperitoneal injections of folate acid
(50 mg/kg) ameliorating colitis severity via controlled DNA methyla-
tion promotion [34]. This nding demonstrates that both hypomethy-
lation and hypermethylation may cause pro-inammatory eects, and
that folate is an important factor balancing these eects.
5. Maintaining DNA synthesis & repair
Folate has a multifaceted role in nucleotide synthesis, with actions
in synthesising the thymine precursor, thymidylate, and purines. In
thymidylate synthesis, cytosolic serine hydroxymethyl transferase
(SHMT), thymidylate synthase (TYMS) and DHFR use 5,10-
methyleneTHF in the methylation of deoxyuridine monophosphate to
form thymidylate monophosphate. Folate in the form of 10-formylTHF
is also utilised in the synthesis of purine ring structures (Fig. 3)[12].
Nucleotide production is increasingly important during prolonged in-
ammation as it allows for DNA repair and the proliferation of immune
cells.
Increases in ROS during inammation can cause DNA mutations
which may further contribute to inammation and can lead to tumor-
igenesis [53]. Folate-dependent thymidylate is particularly important in
DNA repair, with involved enzymes, SHMT, TYMS and DHFR, forming a
multienzyme-complex that translocates to the nucleus during times of
DNA synthesis and repair [54,55]. In a randomised clinical trial ex-
amining arsenic-exposed individuals (n = 450), supplementation with
either 0.4 or 0.8 mg/day of folic acid for 8 weeks resulted in a sig-
nicant decrease in oxidative DNA damage [56]. In comparison, in vitro
and in vivo depletion studies both show folate deciency to exacerbate
DNA damage via reductions in thymine causing uracil to be mis-
incorporated in DNA, and DNA repair responses to be hindered
[5760].
Folate deciency is shown to reduce proliferation in multiple cell
types by causing nucleotide imbalances and subsequent accumulation
of cells in the S phase [6164]. Notably, changes in folate levels may
inuence the proliferation of T cells [64], with aberrant T-cell pro-
liferation associated with autoimmune chronic inammatory diseases
such as rheumatoid arthritis [65]. In cultured lymphocytes, folate de-
pletion reduced the proliferation of CD8
+
T lymphocytes and increased
the CD4
+
to CD8
+
T-cell ratio [64]. Supplementation of folate
(300 nM) to folate-depleted lymphocytes decreased the CD4
+
to CD8
+
ratio [64]. CD4
+
T (helper/inducer) and CD8
+
(cytotoxic/suppressor)
represent two T-cell subtypes, with an increased CD4
+
to CD8
+
ratio
indicative of increased immunodeciency, and a hallmark of auto-
immune inammatory diseases [65]. Further mouse model studies
support a role of folate in maintaining levels of CD4+Foxp3 regulatory
T cells (Tregs) [66,67], a regulatory CD4
+
subtype dysregulated in
chronic inammatory diseases such as inammatory bowel disease
[68]. Folate receptors have been found to be highly expressed in natural
Treg cells in mice, with blockage of these receptors resulting in de-
creased Treg levels [66,67].
6. Factors inuencing inammatory roles of folate
With emerging roles in maintaining inammatory-related pathways,
the potential usefulness of folate supplementation in the prevention and
treatment of inammation is being explored. However, data from
Fig. 2. The BH4 salvage pathway.
eNOS cofactor BH4 is readily oxidised to BH2 in circulation. In cases of de-
creased BH4 bioavailability, eNOS uncoupling occurs, and eNOS produces ROS
instead of NO. BH2 may be reduced back to BH4 via folate-dependent DHFR via
the BH4 salvage pathway. This pathway maintains an adequate BH4: BH2 ratio
to support NO production.
Abbreviations; eNOS; endothelial nitric oxide synthetase, BH4; tetra-
hydrobiopterin, BH2; 7, 8-dihydrobiopterin, DHFR; dihydrofolate reductase.
Figure created with BioRender.
P. Jones, et al. Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
3
animal and human studies examining the inuence of folate supple-
mentation on inammatory biomarkers is inconsistent and varies sig-
nicantly based on factors such as timing of intervention (i.e., pre-
ventative vs. treatment interventions), folate dose, levels of other
dietary factors, and genetic variance in folate enzymes.
Several human intervention studies demonstrate folate supple-
mentation eectively lowers Hcy levels but this has varied inuence on
markers of endothelial dysfunction depending on time of intervention
[6975]. Hcy-lowering had no eect on markers of endothelial dys-
function in studies examining healthy subjects or subjects with current
inammatory conditions (25 mg mg/day of folic acid, 6 weeks2
years) [6972] but did improve endothelial function in subjects with
elevated Hcy (510 mg/day folic acid over 68 weeks) [74,75]. These
ndings indicate folate supplementation is a more eective pre-
ventative measure rather than a potential treatment, particularly in
individuals with elevated Hcy at risk of future inammatory conditions.
Folate interventions for prolonged periods or in advanced disease
stages may cause folate to promote inammation by encouraging
aberrant methylation and cell proliferation activity. Intraperitoneal
injections of folate acid (50 mg/kg) ameliorated colitis severity in one
murine model of colitis [34] but in further murine models supple-
mentation of methyl donors to maternal diets (5 mg/kg folic acid, 5 g
betaine and 5.76 g choline/kg diet) was found to induce susceptibility
to colitis in ospring [76,77]. Supplementation of methyl donors to
maternal diets (15 mg folic acid and 15 g choline/betaine/kg diet) was
shown to combat the eect of a high fat maternal diets on weight gain
in ospring in one model [37], but the opposite eect was seen in cases
of excess folate supplementation (20 g folic acid/kg diet) [78]. This
suggests level of folate dose may also largely determine the inuence
folate may have in inammation.
As methyl donor supplementation is not a targeted approach, af-
fecting methylation of both anti-inammatory and pro-inammatory
genes, it is possible that prolonged or excessive exposure to methyl
donors or intervention at specicdisease stage may promote DNA
methylation patterns that contribute to inammation [79]. Prolonged
or excessive exposure to folate during advanced diseases stages may
exacerbate inammation by also encouraging aberrant cell prolifera-
tion of pro-inammatory and cancerous cell types [80]. Notably,
methotrexate is an anti-folate medication with action in deterring
proliferation of pro-inammatory cells by inhibition of the role of folate
in DNA synthesis. Methotrexate (MTX) is commonly prescribed in
rheumatoid arthritis and other inammatory disorders [81]. However,
increased Hcy levels and risk of cardiovascular disease is commonly
reported in methotrexate users [82] with current consensus that users
should regularly use folate supplementation to oset these risks (re-
commended 5 mg folic acid following MTX treatments) [81].
The interpretation of current ndings is made dicult by the co-
existence of several dietary and genetic factors that regulate folate or
folate-related processes. These factors are not always considered in
study designs but are important nutrient-nutrient and nutrient-gene
interactions that may determine study outcomes. In addition to folate,
adequate levels of vitamin B
12
and vitamin B
6
, and methyl donor cho-
line are required for Hcy-regulation, DNA methylation, and DNA
synthesis [12,83]. Consequently, results of interventions can vary de-
pending on whether folate supplementation was considered in isolation
or in combination with relevant B vitamins and methyl donors. The
occurrence of functional polymorphisms in folate-dependent enzymes
may also inuence the actions of folate in inammation. MTHFR-C677T
is the most extensively studied polymorphism in folate metabolism and
is associated with signicant reductions in MTHFR activity and folate
status [84,85]. Results of in vivo and in vitro studies indicate MTHFR-
C677T as a major genetic determinant of Hcy levels [8689], with this
variant also linked to changes in NO production [90], DNA methylation
[91,92] and DNA damage [93,94] through interaction with folate
status. Other functional polymorphisms in rate-limiting enzymes such
as TYMS and DHFR are associated with changes in folate levels [85]
and could theoretically alter inammatory roles, but are yet to be ex-
amined. Considering the current available evidence, further con-
sideration of factors such as timing of intervention, folate dose and
interactions with dietary and genetic factors is needed in future studies
Fig. 3. Roles of folate in nucleotide synthesis.
Folate synthesises thymine precursor, thymidylate,
and purine ring structures. In thymidylate synth-
esis, SHMT generates 5,10methyleneTHF which is
then used by TYMS in methylation of dUMP to
dTMP. DHFR regenerates oxidised DHF back to
THF. Folate in the form of 10-formylTHF is utilised
in the synthesis of purines, where GAR and AICAR
transformylases transfer one carbon units from 10-
formylTHF to purine ring structures. MTHFD1 has
action in generating the dierent THF forms of
folate supporting these reactions, as well as reac-
tions in Hcy remethylation.
Abbreviations; THF; tetrahydrofolate, MTHFD1;
methylenetetrahydrofolate dehydrogenase 1, GAR
Glycinamide ribonucleotide; AICAR 5-aminoimi-
dazole-4-carboxamide ribonucleotide; SHMT;
serine hydroxymethyl transferase, DHFR; dihy-
drofolate reductase, TYMS; thymidylate synthase,
dUMP; deoxyuridine monophosphate, dTMP;
deoxythymidine monophosphate, Hcy; homo-
cysteine.
Created with BioRender.
P. Jones, et al. Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
4
to elucidate the roles of folate in inammation.
7. Conclusion
There is a growing body of evidence supporting the role of optimal
folate availability in the development and maintenance of processes
linked to inammation. Folate may prevent endothelial dysfunction by
Hcy-dependent and Hcy-independent regulation of NO levels.
Additionally, actions of folate in DNA methylation, repair and synthesis
processes may inuence the inammatory phenotype via epigenetic
changes and modulation of cell proliferation. However, much of our
current knowledge in how folate may inuence inammation is based
on ndings from in vitro studies and murine models, which have not
been yet been translated into investigations of human cohorts. The
interpretation of current ndings is further complicated by several
confounders that may alter the inuence of folate, which need to be
further examined before folate recommendations around inammation
can be made.
Author statement
Conception and design of review by PJ and EB. Drafting, revision,
and approval of nal submission by all authors.
Declaration of competing interest
The authors declare no conict of interest.
Acknowledgements
Patrice Jones is supported by an Australian Government Research
Training Program scholarship and a Hunter Medical Research Institute
(HMRI) Greaves Family Scholarship. Emma Beckett is supported by a
NHMRC Early Career Fellowship.
References
[1] P.J. Stover, Physiology of folate and vitamin B12 in health and disease, Nutr. Rev.
62 (6 Pt 2) (2004) S3S12 discussion S13.
[2] C.M. Steyers, F.J. Miller, Endothelial dysfunction in chronic inammatory diseases,
Int. J. Mol. Sci. 15 (7) (2014) 1132411349.
[3] A.S. De Vriese, et al., Endothelial dysfunction in diabetes, Br. J. Pharmacol. 130 (5)
(2000) 963974.
[4] W.K.C. Lai, M.Y. Kan, Homocysteine-induced endothelial dysfunction, Ann. Nutr.
Metabol. 67 (1) (2015) 112.
[5] R. Esse, et al., The contribution of homocysteine metabolism disruption to en-
dothelial dysfunction: state-of-the-art, Int. J. Mol. Sci. 20 (4) (2019) 867.
[6] A.E. Stanhewicz, W.L. Kenney, Role of folic acid in nitric oxide bioavailability and
vascular endothelial function, Nutr. Rev. 75 (1) (2017) 6170.
[7] C.N. Bernstein, et al., Cancer risk in patients with inammatory bowel disease: a
population-based study, Cancer 91 (4) (2001) 854862.
[8] S. Danese, A. Mantovani, Inammatory bowel disease and intestinal cancer: a
paradigm of the Yin-Yang interplay between inammation and cancer, Oncogene
29 (23) (2010) 33133323.
[9] Y. Pan, et al., Associations between folate and vitamin B12 levels and inammatory
bowel disease: a meta-analysis, Nutrients 9 (4) (2017) 382.
[10] P.E. Lazzerini, et al., Hyperhomocysteinemia, inammation and autoimmunity,
Autoimmun. Rev. 6 (7) (2007) 503509.
[11] H.J. Blom, Y. Smulders, Overview of homocysteine and folate metabolism. With
special references to cardiovascular disease and neural tube defects, J. Inherit.
Metab. Dis. 34 (1) (2011) 7581.
[12] P.J. Stover, M.S. Field, Tracking of intracellular folates, Adv. Nutr. Int. Rev. J. 2
(4) (2011) 325331.
[13] K. Bedard, K.-H. Krause, The NOX Family of ROS-generating NADPH oxidases:
Physiology and pathophysiology, Physiol. Rev. 87 (1) (2007) 245313.
[14] S.H. Chan, et al., Exercise intervention attenuates hyperhomocysteinemia-induced
aortic endothelial oxidative injury by regulating SIRT1 through mitigating NADPH
oxidase/LOX-1 signaling, Redox Biol. 14 (2018) 116125.
[15] I.F.W. McDowell, D. Lang, Homocysteine and endothelial dysfunction: a link with
cardiovascular disease, J. Nutr. 130 (2) (2000) 369S-372S.
[16] N.J. Alp, K.M. Channon, Regulation of endothelial nitric oxide synthase by tetra-
hydrobiopterin in vascular disease, Arterioscler. Thromb. Vasc. Biol. 24 (3) (2004)
413420.
[17] G. Topal, et al., Homocysteine induces oxidative stress by uncoupling of NO
synthase activity through reduction of tetrahydrobiopterin. Free radical biology &
medicine, 36 (12) (2004) 15321541.
[18] L. Jin, et al., Homocysteine induces endothelial dysfunction via inhibition of argi-
nine transport, J. Physiol. Pharmacol. 58 (2) (2007) 191206.
[19] L.H. Liu, et al., Protection of DDAH2 overexpression against homocysteine-induced
impairments of DDAH/ADMA/NOS/NO pathway in endothelial cells, Cell. Physiol.
Biochem. 30 (6) (2012) 14131422.
[20] M.J. Crabtree, A.B. Hale, K.M. Channon, Dihydrofolate reductase protects en-
dothelial nitric oxide synthase from uncoupling in tetrahydrobiopterin deciency,
Free Radic. Biol. Med. 50 (11) (2011) 16391646.
[21] M.J. Crabtree, et al., Critical role for tetrahydrobiopterin recycling by dihydrofolate
reductase in regulation of endothelial nitric-oxide synthase coupling: relative im-
portance of the de novo biopterin synthesis versus salvage pathways, J. Biol. Chem.
284 (41) (2009) 2812828136.
[22] M.E. Hyndman, et al., Interaction of 5-methyltetrahydrofolate and tetra-
hydrobiopterin on endothelial function, Am. J. Physiol. Heart Circ. Physiol. (6)
(2002) 282 p. H2167-72.
[23] S. Kaufman, Some metabolic relationships between biopterin and folate: implica-
tions for the methyl trap hypothesis, Neurochem. Res. 16 (9) (1991) 10311036.
[24] E.S. Stroes, et al., Folic acid reverts dysfunction of endothelial nitric oxide synthase,
Circ. Res. 86 (11) (2000) 11291134.
[25] K. Chalupsky, et al., Folic acid promotes recycling of tetrahydrobiopterin and
protects against hypoxia-induced pulmonary hypertension by recoupling en-
dothelial nitric oxide synthase, Antioxidants Redox Signal. 23 (14) (2015)
10761091.
[26] A.E. Stanhewicz, L.M. Alexander, W.L. Kenney, Folic acid supplementation im-
proves microvascular function in older adults through nitric oxide-dependent me-
chanisms, Clin. Sci. (Lond.) 129 (2) (2015) 159167.
[27] R. Joshi, et al., Free radical scavenging behavior of folic acid: evidence for possible
antioxidant activity, Free Radic. Biol. Med. 30 (12) (2001) 13901399.
[28] B.M. Rezk, et al., Tetrahydrofolate and 5-methyltetrahydrofolate are folates with
high antioxidant activity. Identication of the antioxidant pharmacophore, FEBS
(Fed. Eur. Biochem. Soc.) Lett. 555 (3) (2003) 601605.
[29] L.M. Stead, et al., Methylation demand and homocysteine metabolism, Adv. Enzym.
Regul. 44 (1) (2004) 321333.
[30] G.J. Howie, et al., Maternal nutritional history predicts obesity in adult ospring
independent of postnatal diet, J. Physiol. 587 (Pt 4) (2009) 905915.
[31] N.G. Ashino, et al., Maternal high-fat feeding through pregnancy and lactation
predisposes mouse ospring to molecular insulin resistance and fatty liver, J. Nutr.
Biochem. 23 (4) (2012) 341348.
[32] Z. Vucetic, et al., Maternal high-fat diet alters methylation and gene expression of
dopamine and opioid-related genes, Endocrinology 151 (10) (2010) 47564764.
[33] H.M. O'Hagan, et al., Oxidative damage targets complexes containing DNA me-
thyltransferases, SIRT1, and polycomb members to promoter CpG Islands, Cancer
Cell 20 (5) (2011) 606619.
[34] D.J. Kominsky, et al., An endogenously anti-inammatory role for methylation in
mucosal inammation identied through metabolite proling, J. Immunol. 186
(11) (2011) 65056514.
[35] E. Foran, et al., Upregulation of DNA methyltransferasemediated gene silencing,
anchorage-independent growth, and Migration of colon cancer Cells by interleukin-
6, Mol. Cancer Res. 8 (4) (2010) 471481.
[36] R. Barouki, et al., Developmental origins of non-communicable disease: implica-
tions for research and public health, Environ. Health 11 (2012) 42.
[37] J. Carlin, R. George, T.M. Reyes, Methyl donor supplementation blocks the adverse
eects of maternal high fat diet on ospring physiology, PLoS One 8 (5) (2013)
e63549.
[38] K.D. Sinclair, et al., DNA methylation, insulin resistance, and blood pressure in
ospring determined by maternal periconceptional B vitamin and methionine
status, Proc. Natl. Acad. Sci. 104 (49) (2007) 1935119356.
[39] M. Chen, et al., Methyl decient diet aggravates experimental colitis in rats, J. Cell
Mol. Med. 15 (11) (2011) 24862497.
[40] M.S. Ellulu, et al., Obesity and inammation: the linking mechanism and the
complications, Arch. Med. Sci. : AMS 13 (4) (2017) 851863.
[41] E. Oken, et al., Gestational weight gain and child adiposity at age 3 years, Am. J.
Obstet. Gynecol. 196 (4) (2007) 322 e1-8.
[42] D.A. Lawlor, et al., Epidemiologic evidence for the fetal overnutrition hypothesis:
ndings from the mater-university study of pregnancy and its outcomes, Am. J.
Epidemiol. 165 (4) (2007) 418424.
[43] R. Xie, et al., Maternal high fat diet alters gut microbiota of ospring and exacer-
bates DSS-induced colitis in adulthood, Front. Immunol. 9 (2018) 26082608.
[44] M.A. Hahn, et al., Methylation of polycomb target genes in intestinal cancer is
mediated by inammation, Cancer Res. 68 (24) (2008) 1028010289.
[45] H. Wehbe, et al., Interleukin-6 contributes to growth in cholangiocarcinoma cells by
aberrant promoter methylation and gene expression, Cancer Res. 66 (21) (2006)
1051710524.
[46] J.P. Issa, et al., Accelerated age-related CpG island methylation in ulcerative colitis,
Cancer Res. 61 (9) (2001) 35733577.
[47] T. Maekita, et al., High levels of aberrant DNA methylation in Helicobacter pylori-
infected gastric mucosae and its possible association with gastric cancer risk, Clin.
Cancer Res. 12 (3 Pt 1) (2006) 989995.
[48] F.Y. Wang, et al., Aberrant DNA methylation in ulcerative colitis without neoplasia,
Hepato-Gastroenterology 55 (81) (2008) 6265.
[49] K. Tominaga, et al., Prediction of colorectal neoplasia by quantitative methylation
analysis of estrogen receptor gene in nonneoplastic epithelium from patients with
ulcerative colitis, Clin. Cancer Res. 11 (24 Pt 1) (2005) 88808885.
[50] T. Moriyama, et al., Hypermethylation of p14 (ARF) may be predictive of colitic
P. Jones, et al. Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
5
cancer in patients with ulcerative colitis, Dis. Colon Rectum 50 (9) (2007)
13841392.
[51] S. Cui, et al., Folic acid modulates VPO1 DNA methylation levels and alleviates
oxidative stress-induced apoptosis in vivo and in vitro, Redox Biol. 19 (2018)
8191.
[52] A.F. Kolb, L. Petrie, Folate deciency enhances the inammatory response of
macrophages, Mol. Immunol. 54 (2) (2013) 164172.
[53] S. Kawanishi, et al., Crosstalk between DNA Damage and Inammation in the
multiple Steps of carcinogenesis, Int. J. Mol. Sci. 18 (8) (2017) 1808.
[54] D.D. Anderson, C.F. Woeller, P.J. Stover, Small ubiquitin-like modier-1 (SUMO-1)
modication of thymidylate synthase and dihydrofolate reductase, Clin. Chem. Lab.
Med. 45 (12) (2007) 17601763.
[55] D.D. Anderson, et al., Serine hydroxymethyltransferase anchors de Novo thymidy-
late synthesis pathway to nuclear lamina for DNA synthesis, J. Biol. Chem. 287 (10)
(2012) 70517062.
[56] X. Guo, et al., Protective eect of folic acid on oxidative DNA damage: a rando-
mized, double-blind, and placebo controlled clinical trial, Medicine (45) (2015) 94
e1872-e1872.
[57] G.P. Basten, et al., Sensitivity of markers of DNA stability and DNA repair activity to
folate supplementation in healthy volunteers, Br. J. Canc. 94 (12) (2006)
19421947.
[58] S.J. Duthie, et al., Folate deciency in vitro induces uracil misincorporation and
DNA hypomethylation and inhibits DNA excision repair in immortalized normal
human colon epithelial cells, Nutr. Cancer 37 (2) (2000) 245251.
[59] S.J. James, L. Yin, Diet-induced DNA damage and altered nucleotide metabolism in
lymphocytes from methyl-donor-decient rats, Carcinogenesis 10 (7) (1989)
12091214.
[60] S.J. Duthie, A. Hawdon, DNA instability (strand breakage, uracil misincorporation,
and defective repair) is increased by folic acid depletion in human lymphocytes in
vitro, FASEB J. 12 (14) (1998) 14911497.
[61] W.Y. Zhu, P.W. Melera, Basal levels of metallothionein I and II expression in mouse
embryo broblasts enhance growth in low folate through a cell cycle mediated
pathway, Cell Biol. Int. 25 (12) (2001) 12611269.
[62] M.J. Koury, J.O. Price, G.G. Hicks, Apoptosis in megaloblastic anemia occurs during
DNA synthesis by a p53-independent, nucleoside-reversible mechanism, Blood 96
(9) (2000) 32493255.
[63] R.-F.S. Huang, et al., Folate deciency induces a cell cycle-specic apoptosis in
HepG2 cells, J. Nutr. 129 (1) (1999) 2531.
[64] C. Courtemanche, et al., Folate deciency inhibits the proliferation of primary
human CD8+ T lymphocytes in vitro, J. Immunol. 173 (5) (2004) 31863192.
[65] A. Skapenko, et al., The role of the T cell in autoimmune inammation, Arthritis
Res. Ther. (2005) S4S14 7 Suppl 2(Suppl 2).
[66] T. Yamaguchi, et al., Control of immune responses by antigen-specic regulatory T
cells expressing the folate receptor, Immunity 27 (1) (2007) 145159.
[67] M. Kinoshita, et al., Dietary folic acid promotes survival of Foxp3+Regulatory T
cells in the colon, J. Immunol. 189 (6) (2012) 28692878.
[68] B. Singh, et al., Control of intestinal inammation by regulatory T cells, Immunol.
Rev. 182 (1) (2001) 190200.
[69] A.C. Peeters, et al., The eect of homocysteine reduction by B-vitamin supple-
mentation on markers of endothelial dysfunction, Thromb. Haemost. 92 (5) (2004)
10861091.
[70] P. Dusitanond, et al., Homocysteine-lowering treatment with folic acid, cobalamin,
and pyridoxine does not reduce blood Markers of inammation, endothelial dys-
function, or Hypercoagulability in patients with previous transient ischemic Attack
or stroke, Stroke 36 (1) (2005) 144146.
[71] E.G. Vermeulen, et al., Homocysteine-lowering treatment with folic acid plus vi-
tamin B6 lowers urinary albumin excretion but not plasma markers of endothelial
function or C-reactive protein: further analysis of secondary end-points of a ran-
domized clinical trial, Eur. J. Clin. Investig. 33 (3) (2003) 209215.
[72] Ø. Bleie, et al., Homocysteine-lowering therapy does not aect inammatory
markers of atherosclerosis in patients with stable coronary artery disease, J. Intern.
Med. 262 (2) (2007) 244253.
[73] M. USUI, et al., Endothelial dysfunction by acute hyperhomocyst(e)inaemia: re-
storation by folic acid, Clin. Sci. 96 (3) (1999) 235239.
[74] Bellamy, et al., Oral folate enhances endothelial function in hyperhomocystei-
naemic subjects, Eur. J. Clin. Investig. 29 (8) (1999) 659662.
[75] K.S. Woo, et al., Folic acid improves arterial endothelial function in adults with
hyperhomocystinemia, J. Am. Coll. Cardiol. 34 (7) (1999) 20022006.
[76] T.D. Schaible, et al., Maternal methyl-donor supplementation induces prolonged
murine ospring colitis susceptibility in association with mucosal epigenetic and
microbiomic changes, Hum. Mol. Genet. 20 (9) (2011) 16871696.
[77] S.A. Mir, et al., Prenatal methyl-donor supplementation augments colitis in young
adult mice, PLoS One (8) (2013) 8 p. e73162-e73162.
[78] E. Pannia, et al., A high multivitamin diet fed to Wistar rat dams during pregnancy
increases maternal weight gain later in life and alters homeostatic, hedonic and
peripheral regulatory systems of energy balance, Behav. Brain Res. 278 (2015)
111.
[79] R.J. ONeill, P.B. Vrana, C.S. Rosenfeld, Maternal methyl supplemented diets and
eects on ospring health, Front. Genet. (5) (2014) 289.
[80] J. Sauer, J.B. Mason, S.-W. Choi, Too much folate: a risk factor for cancer and
cardiovascular disease? Curr. Opin. Clin. Nutr. Metab. Care 12 (1) (2009) 3036.
[81] S.L. Whittle, R.A. Hughes, Folate supplementation and methotrexate treatment in
rheumatoid arthritis: a review, Rheumatology 43 (3) (2004) 267271.
[82] M.T. Nurmohamed, Cardiovascular risk in rheumatoid arthritis, Autoimmun. Rev. 8
(8) (2009) 663667.
[83] R. Obeid, The metabolic burden of methyl donor deciency with focus on the be-
taine homocysteine methyltransferase pathway, Nutrients 5 (9) (2013) 34813495.
[84] P. Frosst, et al., A candidate genetic risk factor for vascular disease: a common
mutation in methylenetetrahydrofolate reductase, Nat. Genet. 10 (1) (1995)
111113.
[85] M. Hiraoka, Y. Kagawa, Genetic polymorphisms and folate status, Congenital.
Anom. 57 (5) (2017) 142149.
[86] L. Bathum, et al., Genetic and environmental inuences on plasma homocysteine:
results from a Danish twin study, Clin. Chem. 53 (5) (2007) 971979.
[87] H. Refsum, et al., The hordaland homocysteine study: a community-based study of
homocysteine, its determinants, and associations with disease, J. Nutr. 136 (6)
(2006) 1731S1740S.
[88] L. Peyrin-Biroulet, et al., Association of MTRR 66A > G polymorphism with su-
peroxide dismutase and disease activity in patients with Crohn's disease, Am. J.
Gastroenterol. 103 (2) (2008) 399406.
[89] X. Qin, et al., MTHFR C677T and MTR A2756G polymorphisms and the homo-
cysteine lowering ecacy of dierent doses of folic acid in hypertensive Chinese
adults, Nutr. J. 11 (2012) 2.
[90] C. Antoniades, et al., MTHFR 677 C > T Polymorphism reveals functional im-
portance for 5-methyltetrahydrofolate, not homocysteine, in regulation of vascular
redox state and endothelial function in human atherosclerosis, Circulation 119 (18)
(2009) 25072515.
[91] K.P. Shelnutt, et al., Methylenetetrahydrofolate reductase 677C> T poly-
morphism aects DNA methylation in response to controlled folate intake in young
women, J. Nutr. Biochem. 15 (9) (2004) 554560.
[92] S. Friso, et al., A common mutation in the 5,10-methylenetetrahydrofolate re-
ductase gene aects genomic DNA methylation through an interaction with folate
status, Proc. Natl. Acad. Sci. U. S. A. 99 (8) (2002) 56065611.
[93] N. Botto, et al., Genetic polymorphisms in folate and homocysteine metabolism as
risk factors for DNA damage, Eur. J. Hum. Genet. 11 (9) (2003) 671678.
[94] M. Kimura, et al., Methylenetetrahydrofolate reductase C677T polymorphism, folic
acid and riboavin are important determinants of genome stability in cultured
human lymphocytes, J. Nutr. 134 (1) (2004) 4856.
P. Jones, et al. Journal of Nutrition & Intermediary Metabolism 18 (2019) 100104
6
... 5 Moreover, serum folic acid levels have been reported to have prognostic value in inflammatory diseases. 6 Despite this, folic acid fortification has sparked controversy regarding its effectiveness in preventing major cardiovascular adverse events such as stroke, despite a significant inverse dose-response relationship between the fall in homocysteine after treatment. 5 Notwithstanding growing evidence on the use of folic acid and homocysteine as prognostic markers in different cardiovascular diseases, there are no studies assessing their role in patients with aortic aneurysm/aortic dissection. ...
Article
Full-text available
Background and Aims Folate metabolites have been identified as prognostic markers in cardiovascular, oncologic, and congenital diseases. However, few studies have explored the association between serum folic acid levels and aortic rupture. The aim of this study was to evaluate the predictive value of serum folic acid level for pre-operative adverse events in patients with symptomatic aortic aneurysm/aortic dissection. Methods and Results Patients with symptomatic aortic aneurysm/aortic dissection, presenting to the emergency department (ED), were enrolled in the study. Both univariate and multivariate analyses were performed to identify predictors of pre-operative aortic rupture. The optimal cut-off value for folic acid was determined, and a prognostic nomogram was subsequently developed for potential clinical application. Among 639 symptomatic aortic aneurysm/aortic dissection patients, 8.61% experienced rupture prior to surgery/intervention, with a significantly higher incidence observed in patients with higher serum folic acid levels. Multivariate logistic regression analysis revealed that folic acid levels ≥9ng/mL were an independent risk factor for pre-operative aortic rupture (OR=2.41). A nomogram was developed based on the combination of Stanford type A aortic dissection, folic acid level, white blood cell count, serum potassium, and random blood glucose, demonstrating excellent discrimination ability with the area under the curve of 0.801. Conclusion This large-sample, prospective study suggested that the serum folic acid level was an independent predictor of pre-operative aortic rupture in patients with symptomatic aortic aneurysm/aortic dissection. The developed nomogram highlighted the prognostic value of folic acid and other objective clinical markers, offering a practical tool for early risk stratification, particularly in the ED setting.
... There is an association between folate and inflammation, as the optimal level of folate can prevent endothelial dysfunction and DNA instability. Low levels of folate are commonly observed in chronic inflammatory diseases, indicating that insufficient folate may play a role in the pathogenesis of inflammatory conditions or that chronic inflammation increases folate requirements [50]. Erythrocyte folate is a good indicator of folate status, and biomarkers of inflammation in women have been associated with erythrocyte folate [14]. ...
Article
Full-text available
Objectives: The aim was to simultaneously investigate inflammatory biomarkers, neopterin, the kynurenine/tryptophan (Kyn/Trp) pathway, vitamin D (VitD), vitamin D binding protein (VDBP), and erythrocyte folate, in cerebral palsy (CP). Methods: A case–control study was conducted at Mersin University Hospital. Three- to ten-year-old patients with spastic CP (n = 50) and age- and gender-matched healthy controls (n = 55) were included. Serum levels of neopterin, Trp, Kyn and 25OHD, plasma VDBP, and erythrocyte folate concentrations were measured. Indoleamine-2,3-dioxygenase 1 (IDO-1) enzyme activity was evaluated according to the Kyn/Trp ratio. Comparison and correlation analyses were performed. Results: The levels of neopterin, Trp, and Kyn were lower in children with CP than in healthy controls (p = 0.037, p < 0.001, and p = 0.003, respectively). IDO1 was not significantly different between the CP and control groups (p = 0.214). The levels of VitD and VDBP were higher in children with CP (p < 0.001 and p = 0.001, respectively). The level of erythrocyte folate was also higher in children with CP (p < 0.001). No significant correlation was found between age and inflammatory biomarkers in the CP group. Neopterin was correlated with the level of Gross Motor Function Classification System (GMFCS) level (r = 0.292, p = 0.044), while there was no significant correlation between the other biomarkers and the level of GMFCS in the CP group. Conclusions: Inflammatory biomarkers of neopterin and Kyn are lower, inflammatory biomarkers of VDBP and erythrocyte folate are higher, and anti-inflammatory VitD is higher in children with spastic CP compared to healthy children. More knowledge is needed to demonstrate inflammatory and anti-inflammatory status in children with CP.
... A recent study 15 indicated that the uncontrolled upregulation of BH4 induces mitochondrial dysfunction, cognitive impairment, 16 immune dysregulation, 17 progression of inflammatory and autoimmune diseases, 18 and chronic pain. 18 Nevertheless, the metabolic conversion of BH4 to BH2 upregulates the synthesis of reactive nitrogen species, 19,20 directly contributing to the inflammatory response in cells. 21 Recently, our study 22 has demonstrated that ME/CFS subjects with OI (ME + OI) are associated with the elevated expression of BH4 suggesting the potential role of upregulated BH4 in the pathogenesis of OI. ...
Article
Full-text available
Background Tetrahydrobiopterin (BH4) and its oxidized derivative dihydrobiopterin (BH2) were found to be strongly elevated in ME/CFS patients with orthostatic intolerance (ME + OI). Objective However, the molecular mechanism of biopterin biogenesis is poorly understood in ME + OI subjects. Here, we report that the activation of the non-oxidative pentose phosphate pathway (PPP) plays a critical role in the biogenesis of biopterins (BH4 and BH2) in ME + OI subjects. Research Design and Results Microarray-based gene screening followed by real-time PCR-based validation, ELISA assay, and finally enzyme kinetic studies of glucose-6-phosphate dehydrogenase (G6PDH), transaldolase (TALDO1), and transketolase (TK) enzymes revealed that the augmentation of anaerobic PPP is critical in the regulations of biopterins. To further investigate, we devised a novel cell culture strategy to induce non-oxidative PPP by treating human microglial cells with ribose-5-phosphate (R5P) under a hypoxic condition of 85%N2/10%CO2/5%O2 followed by the analysis of biopterin metabolism via ELISA, immunoblot, and dual immunocytochemical analyses. Moreover, the siRNA knocking down of the taldo1 gene strongly inhibited the bioavailability of phosphoribosyl pyrophosphate (PRPP), reduced the expressions of purine biosynthetic enzymes, attenuated GTP cyclohydrolase 1 (GTPCH1), and suppressed subsequent production of BH4 and its metabolic conversion to BH2 in R5P-treated and hypoxia-induced C20 human microglia cells. These results confirmed that the activation of non-oxidative PPP is indeed required for the upregulation of both BH4 and BH2 via the purine biosynthetic pathway. To test the functional role of ME + OI plasma-derived biopterins, exogenously added plasma samples of ME + OI plasma with high BH4 upregulated inducible nitric oxide synthase (iNOS) and nitric oxide (NO) in human microglial cells indicating that the non-oxidative PPP-induced-biopterins could stimulate inflammatory response in ME + OI patients. Conclusion Taken together, our current research highlights that the induction of non-oxidative PPP regulates the biogenesis of biopterins contributing to ME/CFS pathogenesis.
Article
Full-text available
Humans must obtain vitamin B 9 (folate) from plant-based diet. The sources as well as the effect of food processing are discussed in detail. Industrial production, fortification and biofortification, kinetics, and physiological role in humans are described. As folate deficiency leads to several pathological states, current opinions toward prevention through fortification are discussed. Claimed risks of increased folate intake are mentioned as well as analytical ways for measurement of folate.
Article
Celiac disease (CD) is an autoimmune disorder that presents with gastrointestinal symptoms including diarrhea, weight loss, and abdominal bloating due to the inflammation in the small intestine. It has been associated with various extraintestinal manifestations, including mucocutaneous findings such as dermatitis herpetiformis, anemia, dental enamel defects, osteoporosis, and arthritis. Studies have revealed an increasing association between CD and cardiovascular diseases (CVDs), including atherosclerosis, cardiomyopathy, and arrhythmia. Chronic inflammation, nutritional deficiencies from malabsorption, endothelial dysfunction, thrombophilic autoantibodies, thrombocytosis, and protein C and S deficiency have been proposed as the probable mechanisms for the association between the 2 conditions. This article aims to provide a review of the pathophysiological mechanism of celiac disease causing various CVDs and to compare and contrast the existing studies suggesting both favorable and unfavorable CVD outcomes in patients with CD.
Article
Background Folate has an important role in the functioning of the musculoskeletal system, including modulation of inflammation, immunity, cartilage regeneration, prevention of osteoporosis, and maintenance of muscle strength, but evidence on the association between folate intake and knee pain, functional scores, and radiographic progression in patients with knee osteoarthritis (OA) is still limited. Methodology Our population‐based cohort was extracted from the osteoarthritis initiative (OAI), focusing on individuals with prevalent radiographic knee OA (with a Kellgren–Lawrence score ≥2). Folate consumption was determined using the food frequency questionnaire. Data regarding the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores and radiographic readings were collected over 48 months. We analyzed the compiled data using generalized additive mixed models. Results Our cohort consisted of 1472 OA patients (626 men and 846 women, mean [SD] age 62.35 [8.92]). At the 48‐month follow‐up, we observed a significant correlation between higher folate intake and a slower progression of knee pain and functional scores, as evidenced by a statistically significant decrease in the WOMAC total score, WOMAC pain subscale score, and WOMAC function/disability subscale score (p < .05). The fully adjusted models estimated a reduction of −0.028 points per 50 μg/1000 kcal of daily folate intake on the WOMAC pain subscale, −0.117 points on the WOMAC function subscale, and −0.160 points on the total WOMAC scale. Furthermore, our nonparametric fit analysis suggested that a higher intake of folate might decelerate the radiographic progression of OA. Stratified analyses indicated that an increase in folate consumption might particularly benefit men, older adults, overweight and obese individuals, and those with a higher dietary fiber intake. Conclusion Higher folate intake is correlated with improved knee function and reduced pain in patients with knee OA and might deter the radiographic progression of OA. The benefits appear to be more pronounced in men, older adults, overweight and obese individuals, and those with a higher dietary fiber intake.
Article
Folate uptake is largely mediated by folate receptor (FR)β, encoded by FOLR2 gene, in myeloid immune cells such as granulocytes, monocytes, and especially in macrophages that constitute the reticuloendothelial system (RES) and infiltrate the tumor microenvironment. Since the myeloid immune compartment dynamically changes during tumorigenesis, it is critical to assess the infiltration status of the tumors by FRβ-expressing myeloid cells to better define the targeting efficacy of folate-functionalized drug delivery systems. On the other hand, clearance by RES is a major limitation for the targeting efficacy of nanoparticles decorated with folate. Therefore, the aims of this study are (i) to determine the amount and subtypes of FRβ⁺ myeloid cells infiltrating the tumors at different stages, (ii) to compare the amount and subtype of FRβ⁺ myeloid cells in distinct organs of tumor-bearing and healthy animals, (iii) to test if the cancer-targeting efficacy and biodistribution of a prototypic folate-functionalized nanoparticle associates with the density of FRβ⁺ myeloid cells. Here, we report that myeloid cell infiltration was enhanced and FRβ was upregulated at distinct stages of tumorigenesis in a mouse breast cancer model. The CD206⁺ subset of macrophages highly expressed FRβ, prominently both in tumor-bearing and healthy mice. In tumor-bearing mice, the amount of all myeloid cells, but particularly granulocytes, was remarkably increased in the tumor, liver, lungs, spleen, kidneys, lymph nodes, peritoneal cavity, bone marrow, heart, and brain. Compared with macrophages, the level of FRβ was moderate in granulocytes and monocytes. The density of FRβ⁺ immune cells in the tumor microenvironment was not directly associated with the tumor-targeting efficacy of the folate-functionalized cyclodextrin nanoparticles. The lung was determined as a preferential site of accumulation for folate-functionalized nanoparticles, wherein FRβ⁺CD206⁺ macrophages significantly engulfed cyclodextrin nanoparticles. In conclusion, our results demonstrate that the tumor formation augments the FR levels and alters the infiltration and distribution of myeloid immune cells in all organs which should be considered as a major factor influencing the targeting efficacy of nanoparticles for drug delivery.
Article
Full-text available
Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid formed during the metabolism of the essential amino acid methionine. Hcy is considered a risk factor for atherosclerosis and cardiovascular disease (CVD), but the molecular basis of these associations remains elusive. The impairment of endothelial function, a key initial event in the setting of atherosclerosis and CVD, is recurrently observed in hyperhomocysteinemia (HHcy). Various observations may explain the vascular toxicity associated with HHcy. For instance, Hcy interferes with the production of nitric oxide (NO), a gaseous master regulator of endothelial homeostasis. Moreover, Hcy deregulates the signaling pathways associated with another essential endothelial gasotransmitter: hydrogen sulfide. Hcy also mediates the loss of critical endothelial antioxidant systems and increases the intracellular concentration of reactive oxygen species (ROS) yielding oxidative stress. ROS disturb lipoprotein metabolism, contributing to the growth of atherosclerotic vascular lesions. Moreover, excess Hcy maybe be indirectly incorporated into proteins, a process referred to as protein N-homocysteinylation, inducing vascular damage. Lastly, cellular hypomethylation caused by build-up of S-adenosylhomocysteine (AdoHcy) also contributes to the molecular basis of Hcy-induced vascular toxicity, a mechanism that has merited our attention in particular. AdoHcy is the metabolic precursor of Hcy, which accumulates in the setting of HHcy and is a negative regulator of most cell methyltransferases. In this review, we examine the biosynthesis and catabolism of Hcy and critically revise recent findings linking disruption of this metabolism and endothelial dysfunction, emphasizing the impact of HHcy on endothelial cell methylation status.
Article
Full-text available
Background: Accumulating evidence shows that high fat diet is closely associated with inflammatory bowel disease. However, the effects and underlying mechanisms of maternal high fat diet (MHFD) on the susceptibility of offspring to colitis in adulthood lacks confirmation. Methods: C57BL/6 pregnant mice were given either a high fat (60 E% fat, MHFD group) or control diet [10 E% fat, maternal control diet (MCD) group] during gestation and lactation. The intestinal development, mucosal barrier function, microbiota, and mucosal inflammation of 3-week old offspring were assessed. After weaning all mice were fed a control diet until 8 weeks of age when the microbiota was analyzed. Offspring were also treated with 2% DSS solution for 5 days and the severity of colitis was assessed. Results: The offspring in MHFD group were significantly heavier than those in MCD group only at 2–4 weeks of age, while no differences were found in the body weight between two groups at other measured time points. Compared with MCD group, MHFD significantly inhibited intestinal development and disrupted barrier function in 3-week old offspring. Although H&E staining showed no obvious microscopic inflammation in both groups of 3-week old offspring, increased production of inflammatory cytokines indicated low-grade inflammation was induced in MHFD group. Moreover, fecal analysis of the 3-week old offspring indicated that the microbiota compositions and diversity were significantly changed in MHFD group. Interestingly after 5 weeks consumption of control diet in both groups, the microbiota composition of offspring in MHFD group was still different from that in MCD group, although the bacterial diversity was partly recovered at 8 weeks of age. Finally, after DSS treatment in 8-week old offspring, MHFD significantly exacerbated the severity of colitis and increased the production of proinflammatory cytokine. Conclusions: Our data reveal that MHFD in early life can inhibit intestinal development, induce dysbiosis and low-grade inflammation and lead to the disruption of intestinal mucosal barrier in offspring, and enhance DSS-induced colitis in adulthood.
Article
Full-text available
Endothelial cell injury and apoptosis play a primary role in the pathogenesis of atherosclerosis. Moreover, accumulating evidence indicates that oxidative injury is an important risk factor for endothelial cell damage. In addition, low folate levels are considered a contributing factor to promotion of vascular disease because of the deregulation of DNA methylation. We aimed to investigate the effects of folic acid on injuries induced by oxidative stress that occur via an epigenetic gene silencing mechanism in ApoE knockout mice fed a high-fat diet and in human umbilical vein endothelial cells treated with oxidized low-density lipoprotein (ox-LDL). We assessed how folic acid influenced the levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG, an oxidative DNA damage marker) and cellular apoptosis in in vivo and in vitro models. Furthermore, we analyzed DNA methyltransferase (DNMT) activity, vascular peroxidase 1 (VPO1) expression, and promoter methylation in human umbilical vein endothelial cells. Our data showed that folic acid reduced 8-OHdG levels and decreased apoptosis in the aortic tissue of ApoE-/- mice. Likewise, our in vitro experiments showed that folic acid protects against endothelial dysfunction induced by ox-LDL by reducing reactive oxygen species (ROS)-derived oxidative injuries, 8-OHdG content, and the apoptosis ratio. Importantly, this effect was indirectly caused by increased DNMT activity and altered DNA methylation at VPO1 promoters, as well as changes in the abundance of VPO1 expression. Collectively, we conclude that folic acid supplementation may prevent oxidative stress-induced apoptosis and suppresses ROS levels through downregulating VPO1 as a consequence of changes in DNA methylation, which may contribute to beneficial effects on endothelial function.
Article
Full-text available
Inflammation can be induced by chronic infection, inflammatory diseases and physicochemical factors. Chronic inflammation is estimated to contribute to approximately 25% of human cancers. Under inflammatory conditions, inflammatory and epithelial cells release reactive oxygen (ROS) and nitrogen species (RNS), which are capable of causing DNA damage, including the formation of 8-oxo-7,8-dihydro-2′-deoxyguanosine and 8-nitroguanine. We reported that 8-nitroguanine was clearly formed at the sites of cancer induced by infectious agents including Helicobacter pylori, inflammatory diseases including Barrett’s esophagus, and physicochemical factors including asbestos. DNA damage can lead to mutations and genomic instability if not properly repaired. Moreover, DNA damage response can also induce high mobility group box 1-generating inflammatory microenvironment, which is characterized by hypoxia. Hypoxia induces hypoxia-inducible factor and inducible nitric oxide synthase (iNOS), which increases the levels of intracellular RNS and ROS, resulting DNA damage in progression with poor prognosis. Furthermore, tumor-producing inflammation can induce nuclear factor-κB, resulting in iNOS-dependent DNA damage. Therefore, crosstalk between DNA damage and inflammation may play important roles in cancer development. A proposed mechanism for the crosstalk may explain why aspirin decreases the long-term risk of cancer mortality.
Article
Full-text available
Moderate hyperhomocysteinemia-induced low folate status is an independent risk factor for cardiovascular disease, dementia, and depression. Folate is an essential cofactor in the one-carbon metabolism pathway and is necessary in amino acid metabolism, purine and thymidylate synthesis, and DNA methylation. In the folate cycle and homocysteine metabolism, folate, vitamin B12, vitamin B6, and vitamin B2 are important cofactors. Many enzymes are involved in folate transport and uptake, the folate pathway, and homocysteine (Hcy) metabolism, and various polymorphisms have been documented in these enzymes. Serum folate and total Hcy (tHcy) levels are influenced by folate intake and genetic polymorphisms in 5,10-methylenetertahydrofolate reductase (MTHFR) such as C677T. The prevalence of the MTHFR 677TT genotype varies across ethnic groups and regions, with a frequency of approximately 15% in Japanese populations. Individuals with the TT genotype have significantly higher tHcy levels and lower folate levels in serum than those with the CT and TT genotypes. However, administration of folic acid has been shown to eliminate these differences. Moreover, data have suggested that interventions based on genotype may be effective for motivating individuals to change their lifestyle and improve their nutrition status. Accordingly, in this review, we discuss the effects of MTHFR C677T polymorphisms on serum tHcy and folate levels with folic acid intervention and evaluate approaches for overcoming folic acid deficiency and related symptoms.
Article
Full-text available
Background: Inflammatory bowel disease (IBD) patients may be at risk of vitamin B12 and folate insufficiencies, as these micronutrients are absorbed in the small intestine, which is affected by IBD. However, a consensus has not been reached on the association between IBD and serum folate and vitamin B12 concentrations. Methods: In this study, a comprehensive search of multiple databases was performed to identify studies focused on the association between IBD and serum folate and vitamin B12 concentrations. Studies that compared serum folate and vitamin B12 concentrations between IBD and control patients were selected for inclusion in the meta-analysis. Results: The main outcome was the mean difference in serum folate and vitamin B12 concentrations between IBD and control patients. Our findings indicated that the average serum folate concentration in IBD patients was significantly lower than that in control patients, whereas the mean serum vitamin B12 concentration did not differ between IBD patients and controls. In addition, the average serum folate concentration in patients with ulcerative colitis (UC) but not Crohn's disease (CD) was significantly lower than that in controls. This meta-analysis identified a significant relationship between low serum folate concentration and IBD. Conclusions: Our findings suggest IBD may be linked with folate deficiency, although the results do not indicate causation. Thus, providing supplements of folate and vitamin B12 to IBD patients may improve their nutritional status and prevent other diseases.
Article
Deficiency of folate or vitamin B12 (cobalamin) causes megaloblastic anemia, a disease characterized by pancytopenia due to the excessive apoptosis of hematopoietic progenitor cells. Clinical and experimental studies of megaloblastic anemia have demonstrated an impairment of DNA synthesis and repair in hematopoietic cells that is manifested by an increased percentage of cells in the DNA synthesis phase (S phase) of the cell cycle, compared with normal hematopoietic cells. Both folate and cobalamin are required for normal de novo synthesis of thymidylate and purines. However, previous studies of impaired DNA synthesis and repair in megaloblastic anemia have concerned mainly the decreased intracellular levels of thymidylate and its effects on nucleotide pools and misincorporation of uracil into DNA. An in vitro model of folate-deficient erythropoiesis was used to study the relationship between the S-phase accumulation and apoptosis in megaloblastic anemia. The results indicate that folate-deficient erythroblasts accumulate in and undergo apoptosis in the S phase when compared with control erythroblasts. Both the S-phase accumulation and the apoptosis were induced by folate deficiency in erythroblasts fromp53 null mice. The complete reversal of the S-phase accumulation and apoptosis in folate-deficient erythroblasts required the exogenous provision of specific purines or purine nucleosides as well as thymidine. These results indicate that decreased de novo synthesis of purines plays as important a role as decreased de novo synthesis of thymidylate in the pathogenesis of megaloblastic anemia.
Article
Coronary artery disease (CAD) is a critical cardiovascular disease and a cause of high morbidity and mortality in this world. Hyperhomocysteinemia (HHcy) has been suggested as a risk factor for CAD. In addition, SIRT1 (sirtuin 1) has been reported to play a protective role in a variety of diseases, especially in the cardiovascular system. The main purpose of this study was to investigate the effects of exercise training on apoptosis and inflammation in HHcy animals. We also tested whether exercise protected against Hhcy-induced dysfunction of endothelium through modulation of SIRT1. C57BL mice (8 in each group) were fed with or without 1% L-methionine (w/w) in water for 4 months to induce HHcy. We found that Hhcy repressed SIRT1 and AMPK expression and increased NADPH oxidase activity. Plasma MDA, endothelium LOX-1 and p-p38 were up-regulated by Hhcy induction. NF-κB and it downstream molecules were activated under Hhcy situation, thereby promoting pro-inflammatory responses. Moreover, we also reported that Hhcy caused endothelium apoptosis involving Akt inhibition and mitochondria-dependent apoptotic pathways. Exercise training significantly protected against endothelium from Hhcy caused oxidative injuries. In addition, EX527 (SIRT1 inhibitor) reduced the therapeutic effects by exercise. Our results had indicated that exercise training prevent the development of atherosclerosis through SIRT1 activation and oxidative stress inhibition under Hhcy situation.
Article
Folic acid is a member of the B-vitamin family and is essential for amino acid metabolism. Adequate intake of folic acid is vital for metabolism, cellular homeostasis, and DNA synthesis. Since the initial discovery of folic acid in the 1940s, folate deficiency has been implicated in numerous disease states, primarily those associated with neural tube defects in utero and neurological degeneration later in life. However, in the past decade, epidemiological studies have identified an inverse relation between both folic acid intake and blood folate concentration and cardiovascular health. This association inspired a number of clinical studies that suggested that folic acid supplementation could reverse endothelial dysfunction in patients with cardiovascular disease (CVD). Recently, in vitro and in vivo studies have begun to elucidate the mechanism(s) through which folic acid improves vascular endothe- lial function. These studies, which are the focus of this review, suggest that folic acid and its active metabolite 5-methyl tetrahydrofolate improve nitric oxide (NO) bioavailability by increasing endothelial NO synthase coupling and NO production as well as by directly scavenging superoxide radicals. By improving NO bioavailabil- ity, folic acid may protect or improve endothelial function, thereby preventing or reversing the progression of CVD in those with overt disease or elevated CVD risk. © The Author(s) 2016. Published by Oxford University Press on behalf of the International Life Sciences Institute.