Next Article in Journal
Preventive Vitamin D Supplementation and Risk for COVID-19 Infection: A Systematic Review and Meta-Analysis
Previous Article in Journal
Associations between Neck Circumference, Mid-Upper Arm Circumference, Wrist Circumference, and High Blood Pressure among Lithuanian Children and Adolescents: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation

by
Maciej W. Socha
1,2,*,†,
Wojciech Flis
1,2,† and
Mateusz Wartęga
3
1
Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland
2
Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
3
Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2024, 16(5), 678; https://doi.org/10.3390/nu16050678
Submission received: 28 January 2024 / Revised: 26 February 2024 / Accepted: 27 February 2024 / Published: 28 February 2024
(This article belongs to the Special Issue Maternal Nutritional Intake and Child Health)

Abstract

:
Pregnancy is an extremely stressful period in a pregnant woman’s life. Currently, women’s awareness of the proper course of pregnancy and its possible complications is constantly growing. Therefore, a significant percentage of women increasingly reach for various dietary supplements during gestation. Some of the most popular substances included in multi-ingredient supplements are folic acid and choline. Those substances are associated with positive effects on fetal intrauterine development and fewer possible pregnancy-associated complications. Recently, more and more attention has been paid to the impacts of specific environmental factors, such as diet, stress, physical activity, etc., on epigenetic modifications, understood as changes occurring in gene expression without the direct alteration of DNA sequences. Substances such as folic acid and choline may participate in epigenetic modifications by acting via a one-carbon cycle, leading to the methyl-group donor formation. Those nutrients may indirectly impact genome phenotype by influencing the process of DNA methylation. This review article presents the current state of knowledge on the use of folic acid and choline supplementation during pregnancy, taking into account their impacts on the maternal–fetal unit and possible pregnancy outcomes, and determining possible mechanisms of action, with particular emphasis on their possible impacts on epigenetic modifications.

1. Introduction

The preconception period and the period of pregnancy are special times in every woman’s life. Pregnancy involves many physiological adaptations that may influence a woman’s behavior and health. Moreover, pregnancy adaptations often require additional supplementation during pregnancy. In recent years, there has been a noticeable increase in women’s awareness of the use of supplementation, both in the preconception period and during gestation [1]. Moreover, a significant improvement in the awareness of women (and often their partners) regarding the overall course of pregnancy and possible pregnancy-associated health risks can be observed [2]. The basis for the increase in this awareness is probably the wide availability of the Internet and numerous educational programs. Additionally, an increasing percentage of physicians draw attention to the necessity and positive effects of supplementation during pregnancy, which further encourages patients to take such actions. Therefore, an increasing percentage of pregnant women try to follow the principles of a proper diet, perform appropriate exercises, and ingest supplementary preparations. According to recent studies, a significant number of the surveyed women reach for supplementary preparations during pregnancy [3,4,5].
During pregnancy, the demand for nutrients increases dramatically, including an increased demand for vitamins, choline, folic acid, etc., which ensure the proper course of pregnancy and maintain proper fetal development [6]. The levels of many nutritional factors in the blood serum decrease (or remain at low, insufficient levels) during pregnancy unless they are supplemented [6,7]. Referring to recent research, it can be seen that consuming insufficient nutritional supplements may be associated with a wide range of pregnancy complications, such as Down syndrome, neural tube defects, anemia, pregnancy-induced hypertension (and preeclampsia), fetal growth restriction, gestational diabetes, and an increased percentage of cesarean deliveries [8,9]. Additionally, improper nutrition during pregnancy may also have long-term effects on the newborn child, such as asthma, congenital heart defects, allergies, and neurodevelopmental disorders [1,2,8,9,10,11].
Molecules provided with a proper diet are crucial in supporting metabolic functionality and activity, including cell proliferation and differentiation and cell signaling, which regulate tissue function, growth, and homeostasis [12]. Substances such as vitamins (A, D, K, and group B vitamins), choline, folates, inositol, etc., are involved in several stages of fetal, maternal, and placental interaction, working to enable a healthy gestation period. Recently, the topic of epigenetics in the context of maternal–fetal medicine has been gaining more and more attention. Substances provided with the diet, apart from their obvious direct impacts on cellular metabolism and homeostasis, may have indirect effects through epigenetic modifications of the genome. These substances may participate in epigenetic modifications by acting as methyl-group donors. Nutrients specific to epigenetic pathways may directly and indirectly impact the epigenome by influencing the processes of DNA methylation and histone modification [13,14]. These are primarily folic acid and choline [14]. Those molecules play a crucial role, one which indirectly affects the entire cycle of one-carbon groups and the biosynthesis of nucleotides. Because epigenetic modulation is one of the elementary programming processes, it seems obvious that incorrect folate or choline supplementation may be correlated with abnormalities in fetal programming and an increased tendency to fetal disorders [12,13,14,15].
The aim of this review is to present the current state of knowledge regarding the impacts of crucial substances such as choline and folic acid on maternal–fetal medicine and determine their possible mechanisms of action, with particular emphasis on the impact on epigenetic mechanisms.

2. Epigenetics

Currently, epigenetics refers to the changes occurring in gene expression that are not in direct correlation with changes in the DNA sequence itself [16]. The epigenetic mechanisms are responsible for specific gene expression without altering the genetic code. Environmental factors such as exposure to certain substances (provided with the diet), physical activity, or stressors can interact with genes and modulate their expression and function without altering the nucleotide sequence [17,18]. Epigenetic patterns may change throughout life—influenced by environmental exposures or nutritional status [18]. Changes in epigenetic patterns may lead to an alteration in the expression of specific genes, which can have downstream effects, such as changes in the risks of specific diseases, the metabolism, or behavioral patterns. Therefore, epigenetic signatures influenced by specific factors may determine human disease susceptibility, longevity, or, even, behavior [18,19,20] (Figure 1).
Epigenetic modifications involve DNA methylation, histone protein modification, or chromatin remodeling [21]. DNA methylation reaction is mediated by a family of DNA methyltransferases (DNMTs) and involves the addition of a methyl residue (-CH3 group) to the 5′ position of a cytosine, leading to 5-methylcytosine (5-mC) formation [22]. DNA methyltransferase exists in three forms (DNMT 1-3), with DNMT3 occurring in two isoforms—DNMT3a and DNMT3b. DNMT3a and DNMT3b methylate de novo DNA, while DNMT1 methylates the previously non-methylated hemi-DNA strand to generate a complementary DNA sequence [23]. Methylation occurs mainly within cytidine-guanine dinucleotide (CpG), which occurs repeatedly throughout the genome. The estimated range of CpG methylation is 70–80% throughout the DNA sequence in human cells [24]. Methylation of the CpG region located in the promoter of a specific gene increases the ability to bind other proteins to the methylated promoter region, leading to the blocking of the transcription of the specific gene [21,22]. Subsequently, the presence of methylated CpG dinucleotide residues prevents the transcriptional activity of specific genes that are expected to remain “silenced” in a cell-type-specific manner [25,26].
Epigenetic modification of histones is vastly more complex [27]. DNA double strand is densely packed into groups of approximately 146 base pairs, which are tangled around the proteins of histones. This complex with base pairs weaved around histones forms nucleosome, which is a subform of DNA packing [28]. From a chemical point of view, each nucleosome consists of the duplicated four core histones—H2A, H2B, H3, and H4. The entire nucleosome is covalently bonded by histone H1 [18].
There are several groups of enzymes that can modify histone proteins. Histone acetyltransferases (HATs) are enzymes that transfer the acetyl group onto the amino group of a lysine residue within the histone tail, leading to the positive charge of lysin neutralization [29]. In contrast, histone deacetylases (HDACs) work the opposite way—removing the acetyl residue restores the positive charge, which strengthens the connections of histones with the DNA [29,30]. Therefore, HATs can be classified as transcription activators, whereas HDACs are known as strong transcription suppressors. Another group of enzymes are histone phosphatases and kinases; these add or remove a phosphate residue from the hydroxyl group of the histone tail, leading to the removal or restoration of the positive charge, respectively. HATs and histone kinases are known as transcription activators, whereas HDACs and histone phosphatases are responsible for restraining transcriptional activity [22,23,24,25,26,27,28,29,30,31,32,33].
In addition to influencing the ionic charge of the protein (through acetylation and phosphorylation), epigenetic modification of histones can also take place through methylation [34]. Histone lysine methyltransferase (HKMT) and arginine methyltransferase (PRMT) are enzymes responsible for the methylation of histone tails, leading to gene transcription alteration [35]. Similarly to the process of DNA methylation, the addition of a methyl residue to the histone tail (by specific methyltransferases) leads to a change in the transcriptional activity of a specific region of the genome.
Regulation of gene transcription may also occur as a result of changes in the spatial conformation of chromatin [36]. A process called chromatin condensation allows a formation of a tightly packed, histone-rich structure to form separate chromosomes. The greatest compaction occurs during the metaphase of cell division [37]. Tight packaging of the genome leads to the production of heterochromatin, which becomes inaccessible to transcription factors, leading to the silencing of specific genes. In turn, loosening the chromatin structure results in the formation of euchromatin, which is available to transcription factors [38]. Chromatin remodeling, leading to gene silencing or activation, is under a strict control provided by an ATP-dependent protein complex or by histone-modifying proteins such as Polycomb-group proteins [21].
The ATP-dependent chromatin-remodeling protein complex, known as SWI/SNF, has catalytic activity in detaching specific histones from DNA, leading to the loosening of the chromatin structure. In turn, Polycomb proteins participate in chromatin condensation, leading to gene silencing. They produce protein multimers—protein repressive complexes (PRC), which, when bound to histones, lead to their mono-ubiquitination, which causes tighter packing of chromatin, leading to the inhibition of gene transcription and specific gene silencing [39,40,41].
DNA methylation seems to be not only the best-studied DNA modification pathway but also the most important mechanism of epigenetic genome modification [42]. The DNA methylation process cannot occur without molecules known as methyl-group donors. The main cellular donor of methyl groups is S-adenosylmethionine (SAM) [42]. Apart from playing a crucial role in DNA methylation, SAM is greatly involved in other cellular biochemical pathways, such as antioxidant formation, the synthesis of neurotransmitters, or phosphatidylcholine synthesis [43]. This molecule is synthesized during the “one-carbon metabolism”—a biochemical pathway responsible for the maintenance of cellular homeostasis, with additional participation of methyl-group donors [44]. These reactions support a vast group of interweaving metabolic pathways, such as synthesis of neurotransmitters, nucleotide metabolism, and/or lipids biosynthesis. The main biochemical pathways involved in a one-carbon cycle are methionine and folate cycles, among which the methyl-group donors such as choline, betaine, vitamins B6 and B12, and folate are crucial players [45].
In the cytosol, SAM is formed by adenylation of methionine catalyzed by methionine S-adenosyltransferase (MAT) [46,47]. Methionine is supplied through the diet or is de novo generated from homocysteine (hCy). After donating its CH3-group to a specific acceptor, SAM is transformed into S-adenosylhomocysteine (SAH), which is a strong metyltransferares competitive antagonist [48]. SAH is subsequently hydrolyzed to homocysteine and adenine. Adenine can subsequently enter into the purine-ring synthesis reaction [46,49]. In turn, homocysteine can be further transformed in several pathways. First, hCy can be transsulfurated, where it combines with serine to form cystathionine in a reaction catalyzed by cystathionine β-synthase (CBS) [50]. Secondly, hCy can convert to methionine, which will enhance further SAM production. Homocysteine can be converted to methionine via two biochemical pathways [47,50,51]. Betaine-homocysteine methyltransferase (BHMT) catalyzes the transfer of a methyl group from betaine (which is a metabolite of choline) to hCy, leading to the formation of methionine and dimethylglycine [52]. This pathway is particularly active in the muscles and liver, due to the high content of betaine and choline in these organs [47]. The second methionine regenerating pathway includes the folic acid pathway [53]. 5-methyltetrahydrofolate-homocysteine methyltransferase (MS) generates methionine by the transfer of the 5-methyltetrahydrofolate (5-mTHF) methyl moiety to hCy, leading to the methionine and tetrahydrofolate (THF) formation [54]. Both a decrease in the concentration of choline (and its metabolites) and folic acid lead to a significant increase in the concentration of homocysteine in the cell [46,54]. This suggests that methionine, folate, and choline metabolic pathways interweave and strongly interact at the point where hCy is metabolized to methionine. Transmethylation reactions occurring in the one-carbon cycle are strictly regulated by both external factors and the metabolites themselves. The ability to regenerate methionine from homocysteine in the folic acid pathway is strongly dependent on vitamin B12 availability, which is a cofactor of MS [46]. Overproduction of SAM causes inhibition of BHMT-dependent methionine regeneration [52]. Moreover, excess SAM inhibits methylenetetrahydrofolate reductase (MTHFR), leading to a decrease in the concentration of 5-mTHF, which participates in the regeneration of methionine [55]. Taking the above into consideration, it can be assumed that the cell’s ability to synthesize SAM is strictly dependent on the current availability of methionine, choline (and betaine), and 5-methyltetrahydrofolate (5-mTHF) [56]. Without ensuring an adequate supply of the substrates participating in the one-carbon pathway, it becomes impossible to generate an appropriate number of methyl-group donors (such as SAM), which are a link between cell metabolism and DNA methylation. Subsequently, this imbalance may translate into the impairment of the process of epigenetic modification of the genome [46,52,56].
Currently, much attention is given to the influence of external factors, such as dietary habits, on the development of the fetus from the moment of conception [57]. Moreover, it is believed that gestational (or pre-conceptional) exposure to environmental factors may epigenetically determine the phenotype or disease-susceptibility of the offspring [58]. After fertilization of the oocyte by the sperm, the DNA methylation pattern of the genome is erased, and then the de novo methylation process of the newly formed zygote occurs in order to have pluripotent cells [18,56]. During this process, a completely new DNA methylation pattern will be created, one which will be passed on to subsequent cells. DNA methylation is a key process for normal physiological development and one that plays an important role in gene expression during future cell differentiation. Additionally, during this time, female embryos will have their X chromosomes inactivated, leaving only one copy of each gene linked to the X chromosome to be expressed [18,57,58]. Taking all the above into consideration, we believe that the period of cellular differentiation occurring immediately after fertilization is the crucial moment in which proper DNA (and histones) methylation is able to ensure the proper, uninterrupted development of the fetus. Therefore, we believe that ensuring appropriate supplementation (leading to the proper supply of methyl-group donors) during this period is crucial for proper fetal development. For the rest of this review, we would like to focus on choline and folic acid and their possible roles in fetal development, emphasizing their possible impacts on epigenetic programming.

3. Choline

Choline, being a quaternary ammonium cation, is a water-soluble molecule that is crucial for proper homeostasis [59]. This micronutrient serves as the starting material for several important metabolites which are key players in fetal development. There is an increased demand for choline during pregnancy, especially in late gestation [60]. Referring to the research, it can be determined that pregnant women currently consume too little choline [60]. Insufficient choline ingestion can lead to serious pregnancy complications such as premature birth, preeclampsia, and fetal growth restriction (FGR) [59]. Therefore, a sufficient choline intake is pivotal for proper fetal development and homeostasis. An adequate intake level of 425 mg/day has been established for women, with up to 550 mg/day during pregnancy and lactation [1,61].
Choline (and its derivatives) are involved in a variety of processes, such as the biogenesis of cell membranes, cell divisions, DNA methylation, lipid transport, and myelination of nerve axons [49]. Choline can be supplied with food and can also be synthesized de novo [62]. The highest amounts of choline can be observed in red meat, eggs, and fish [62]. The endogenous synthesis of choline occurs mainly in the liver [62]. Endogenous choline is mostly obtained from phosphatidylethanolamine. Phosphatidylethanolamine N-methyltransferase (PEMT) sequentially methylates phosphatidylethanolamine to phosphatidylcholine using three molecules of SAM as a methyl donor [62,63]. Then, the newly formed phosphatidylcholine is enzymatically cleaved by phospholipases to choline or can participate in subsequent cell membrane biogenesis. Despite the consumption of three SAM molecules in the choline synthesis process, the concentration of S-adenosylmethionine is maintained via the BHMT pathway [64]. Interestingly, estrogen can upregulate PEMT expression in the human liver, leading to increased choline production during pregnancy [65,66]. This may suggest that the upregulation of endogenous choline de novo synthesis in a pregnant woman’s liver occurs during the gestation period.
A portion of choline is acetylated into acetylcholine (Ach) by the enzymatic activity of choline acetyltransferase (ChAT) [67,68]. Subsequently, acetylcholine is stored in neuronal membranes, where it acts as a neurotransmitter [69]. This is particularly pivotal in neurons with increased choline consumption, such as cholinergic autonomic neurons. The central nervous system cannot synthesize choline itself. It is provided by two main sources. The first is Ach degradation to free choline by acetylcholine esterase (AchE). The second pathway includes hydrolysis of phosphatidylcholine by specific phospholipases. Subsequently, free choline is stored in neuronal membranes for further Ach synthesis [69,70,71,72].
Choline is the precursor for several cell membrane phospholipids, such as phosphatidylethanolamine, sphingomyelin, and phosphatidylcholine. These choline-derived phospholipids are responsible for maintaining the structural and functional integrity of cellular membranes, including neuronal membranes [73]. They also participate in other physiological processes essential for normal brain development, such as signaling, cell proliferation, and myelination [73,74,75].
Apart from the obvious direct impacts of choline (and its derivatives) on specific cell metabolic pathways, it is also worthwhile to consider the indirect impacts of choline on epigenetic mechanisms. Recently, choline has emerged as a promising possible modifier of the genomic sequence that modulates gene expression by affecting the concentration of SAM, which is the main cellular methyl donor molecule participating in epigenetic mechanisms such as DNA and histone methylation [21]. As mentioned earlier, de novo choline synthesis consumes three SAM molecules. However, choline, through its indirect effects, is one of the factors that significantly increases the availability of SAM in the cell [45,63]. Recent studies have shown that choline contributes to the methylation of homocysteine, leading to a reduction in its concentration [76]. Moreover, oral choline supplementation was associated with a significant decrease in hCy concentrations [76]. We believe that this is possible mainly due to the choline metabolite—betaine. Betaine is a molecule containing three reactive methyl groups bonded with a nitrogen atom [77]. It is synthesized through choline oxidation via choline dehydrogenase and betaine aldehyde dehydrogenase [78]. One of three methyl groups previously associated with betaine is transferred to homocysteine, leading to methionine and dimethylglycine (DMG) formation in a reaction catalyzed by betaine homocysteine N-methyltransferase (BHMT) [78,79]. Then, methionine can transform into SAM, which will donate its methyl groups to a specific acceptor. DMG is subsequently partially eliminated through urine and partially metabolized into sarcosine and glycine [78,79,80].
As mentioned earlier, choline intake by pregnant women is largely insufficient. Studies have clearly shown that both supplementation and choline deficiency during various stages of human development have consequences on the brain function, neuronal activity, and cognitive functions of adult offspring [81]. Firstly, prenatal choline supplementation positively influenced the performance of memory-related tasks conducted later in the offspring’s life [82]. Moreover, a choline-deficient diet led to significant impairment in learning and memory skills in mice [83]. Additionally, intrauterine exposure of the developing fetus to choline has a positive effect on the proliferation of progenitor cells, while inhibiting apoptosis of hippocampal cells, leading to the improvement of hippocampal functioning [82,84,85,86]. Finally, choline supplementation positively impacted visuospatial and auditory memory in offspring [84,85,86]. It has also been shown that maternal choline supplementation during pregnancy can exert a neuroprotective effect and attenuate cognitive impairment associated with seizures [87,88,89,90]. Furthermore, it has been shown that neural tube defects (previously mainly associated with folic acid depletion) can be associated with choline supplementation [91]. According to the research, patients with low choline levels had a significantly increased risk of neural tube defects (NTD) [92,93,94,95].
It also turns out that choline supplementation can also alleviate symptoms related to pathologies occurring during pregnancy. Fetal alcohol exposure can lead to congenital fetal alcohol spectrum disorders (FASD), which are characterized by developmental disabilities such as congenital impairment, seizures, and poor growth [96]. Choline supplementation in offspring with FASD symptoms was associated with better verbal memory, fewer behavioral symptoms of attention deficit hyperactivity (ADHD), and increased non-verbal intelligence [97]. However, choline supply had no effect on the motion disorders occurring in this group of disorders [98].
The potential involvement of choline in autism spectrum disorder (ASD) has also been postulated [99]. This developmental disorder is characterized by difficulties in social communication and interaction, with additional restrictions and patterns apparent in social behavior. Referring to the research, these disorders may be caused by improper provision of nutrients during pregnancy [99,100]. Metabolic mapping of deep brain structures revealed that patients with ASD had significantly lower concentrations of choline in gray- and white-matter structures, compared with patients without developmental disorders [100,101]. Moreover, it was also shown that patients with ASD had significantly lower concentrations of choline (and its metabolites) in their blood plasma [102]. This may suggest that choline may be one of the crucial players ensuring proper brain development and may also prove to be an important biomarker of the severity of cognitive disorders.
It is hypothesized that perinatal choline supplementation may prevent the occurrence of cognitive disorders (such as ASD) or mental illnesses such as schizophrenia [103]. Expression of the CHRNA7 gene and its receptor product, the α7-nicotinic receptor (among other receptors), are significantly reduced in specific disorders, especially in ASD or schizophrenia [103]. α7-nicotinic acetylcholine receptor is strongly involved in the maturation of GABA synapse inhibitors. Referring to the research, it has been noted that gestational choline supplementation was associated with the enhancement of the expression of the CHRNA7 gene and its product [103,104]. The offspring of patients who ingested choline during pregnancy showed significantly less social withdrawal and significantly fewer attention problems, compared with the placebo group [104]. We believe that choline supplementation can result in more effective maturation of GABA receptors, which may reduce excessive synaptic activation in the future and thus lead to a reduced risk of behavioral disorders or mental illnesses.
Undoubtedly, there is a connection between choline supplementation and proper brain development. Possible explanations for this phenomenon seem to be multifactorial. Firstly, phospholipids (which are derivatives of choline) are an essential component of the emerging cell membranes—their presence is crucial during the period of rapid cell division and myelination of newly divided neurons. Secondly, acetylcholine ensures the proper development of brain progenitor cells and enables proper cholinergic conduction, which is crucial in the developing brain. Third, as mentioned earlier, choline can directly influence growth stimulation while inhibiting apoptosis of deep brain structures such as the hippocampus. Finally, choline may exert an indirect effect on brain development through epigenetic mechanisms as a methyl-group donor. Studies have shown that a low-choline diet during pregnancy was associated with significant fetal decline of hippocampal DNA methylation of the Cdkn3 (cyclin-dependent kinase inhibitor 3) gene and increased expression of its protein product, kinase-associated phosphatase (Kap), a cell cycle regulator that inhibits cell proliferation [105].
The effects of choline supplementation on the symptoms associated with genetic neurological disorders such as Down syndrome were also investigated. A rodent model of Down syndrome (DS) showed great depletion of cholinergic neurons of the basal forebrain, leading to impairment in cognitive functions, memory, and attention functions—symptoms often presented in Alzheimer’s disease [63]. Interestingly, maternal perinatal choline ingestion in the mentioned rodent model significantly improved cognitive functions in the offspring with DS [106,107]. Moreover, choline supplementation seems to not only have a positive effect on alleviating the cognitive symptoms of Down syndrome, but also may help reduce the risk of its occurrence. Referring to the conducted research, women who delivered a child with DS had significantly lower folic acid and choline concentrations [108]. Additionally, these patients had significantly higher homocysteine concentrations, compared to women with a child without cognitive disorders [108]. These data are extremely interesting for several reasons. First, these data show promising possible treatment options for alleviating Down syndrome symptoms in the offspring. Secondly, they indicate the interdependence between the metabolisms of folic acid and choline and confirm the fact that the biochemical pathways of the one-carbon cycle are interconnected and interwoven. Finally, these data may suggest that the appropriate concentration of methyl groups may translate into the risk of DS. As mentioned earlier, both choline and folic acid participate in the one-carbon pathway, which (among other things) is responsible for generating methyl residue donors. In cases of an insufficient supply of folic acid and choline, methionine synthesis decreases with an increase in hCy concentration. Improper methionine concentration translates into insufficient SAM production, which causes a decrease in the number of available methyl groups. As a result, hypomethylation of the kinetochore may occur—the latter is a protein connecting the centromere with the karyokinetic spindle fibers and determines the proper movement of chromosomes in the metaphase and anaphase of cell division [109]. Insufficient hypomethylation of the kinetochore impairs its function, which may lead to abnormal cell division, leading to the development of Down syndrome. Taking all of the above into consideration, we believe that choline (being strongly involved in the production of SAM) may significantly contribute to the reduction of cognitive symptoms occurring in a number of neurodevelopmental diseases and may also prove to be a factor that will have a positive impact in reducing the risk of Down syndrome in offspring. However, this topic requires further thorough research.
In addition to the described impact on fetal development, choline may also contribute to the proper course of pregnancy itself, which may translate into a reduction in the rate of pregnancy complications. Choline supplementation in the third trimester of pregnancy was associated with a significant decrease in the concentration and expression of Fms-like tyrosine kinase-1 factors (sFlt-1), which are the main antiangiogenic factors involved in the preeclampsia pathogenetic pathway [110]. Additionally, choline supplementation has been linked with positive modulation of oxidative stress, apoptosis, and inflammatory response in human placental trophoblast cells [111,112]. Both overexpression of sFlt-1 and inflammatory response (with the assistance of oxidative stress) occur within the preeclampsia pathogenetic pathway. We believe that choline, by significantly reducing the concentration of sFlt-1 and inhibiting the development of the inflammatory reaction, may contribute to the inhibition of the development of preeclampsia.
It is also believed that choline may positively affect fetal growth [113]. Referring to the research, rodents with an insufficient placenta, after choline supplementation, showed higher betaine concentrations, which, in turn, positively correlated with fetal weight gain in early pregnancy [114,115]. It is believed that choline may exert its effect on fetal growth via the insulin-like growth factor (IGF) axis [113]. Choline supplementation, by influencing DNA methylation and increasing gene expression, is associated with increased expression of specific IGF proteins, which leads to improved fetal growth [116].
Choline supplementation may also affect the hypothalamic–pituitary–adrenal (HPA) fetal axis. Excessive stimulation of the HPA axis during pregnancy may translate into increased risks of preeclampsia and fetal growth restriction [117]. Moreover, increased activity of the HPA axis (caused, for example, by the use of glucocorticosteroids or maternal stress) may lead to increased susceptibility to the development of hypertension and diabetes as an adult [118]. Apart from the negative impact of HPA axis stimulation on somatic disorders, it is believed that HPA hyperstimulation may be connected with psychiatric disorders and long-term neurobehavioral outcomes [119,120,121]. The latest research has shown that maternal choline supplementation led to the alteration (via the donation of methyl groups) of the methylation of genes, such as corticotropin-releasing hormone gene (Crh) and glucocorticoid gene (Nr3c1), in the placental tissue [122]. Subsequently, these changes led to a decrease in the cortisol concentration in the umbilical cord blood [122]. The above data show that choline prenatal supplementation may be a potent novel therapeutical option in pregnancies where excessive maternal stress leads to HPA axis hyperactivity. However, this interesting topic requires further thorough research.
Taking all the above into consideration, we believe that choline can exert positive effects on placentation and trophoblast cells by reducing the inflammatory response, reducing the expression of anti-angiogenic factors, and increasing the secretion of specific growth factors. These data clearly show that choline intake may be beneficial for pregnancies complicated by placental insufficiencies such as preeclampsia or fetal growth restriction. Overall, given the compelling data and encouraging results from the above studies, further examination of the relationships between maternal choline intake and placental function, fetal brain development, and future cognitive function is required.

4. Folic Acid

The term “folates” is a general term encompassing folic acid and its derivative compounds, such as dihydro, tetrahydro, and methyl, having specific metabolic activities in the cell [123]. Natural sources of folates include fruits, meat, and green, leafy vegetables [123]. Additionally, folic acid (the synthetic form of folates) is used as a fortifying factor in dietary supplements [124].
Folic acid (as well as its metabolites), in addition to its effective direct impact on a wide range of metabolic processes occurring in the cell, is also a well-known epigenetic modifier. Apart from participating in the synthesis of the purine ring, it also indirectly participates in the donation of methyl groups in epigenetic transformations [125]. After enzymatic cleavage of ingested nutrients in the intestine, folic acid enters the cells (mainly the liver, kidney, and skeletal muscles), where it is reduced to dihydrofolate (DHF), and then to tetrahydrofolate (THF) by dihydrofolate reductase (DHFR) [125]. Subsequently, serine hydroxymethyltransferase (SHMT) catalyzes the reaction of conversion of THF to 5,10-methylenetetrahydrofolate (5,10-mTHF). This reaction requires serine, glycine, and vitamin B6, as coenzymes, to occur properly. Then, 5,10-mTHF is reduced to 5-mTHF in a reaction catalyzed by methylenetetrahydrofolate reductase (MTHFR) [126]. In this metabolic form, folic acid enters the one-carbon cycle, where it actively participates in the generation of methyl residue donors. THF and 5-mTHF are among the most active metabolites of folic acid [125,126,127]. As mentioned earlier, the one-carbon cycle consists of intersecting biochemical pathways (Figure 2). Generally, these pathways can be divided into folate-dependent and folate-independent pathways. The folate-independent pathway consists of a series of reactions involving choline and its metabolites, whereas the folate-dependent pathway consists of a series of reactions involving hCy, 5-mTHF, and vitamin B12 [127,128]. If the correct concentrations of substrates and cofactors are ensured, methionine synthase (MS) transfers the methyl group from 5-methyltetrahydrofolate to homocysteine, leading to the formation of methionine and tetrahydrofolate [129]. Then, methionine can be converted into SAM, which will serve as a methyl-group donor [129]. In turn, the resulting THF can be converted back to 5,10-meTHF in a reaction catalyzed by SHMT [130]. There is no doubt that the entire one-carbon cycle is subject to strong dependencies resulting from the network of regulatory factors as well as the availability of substrates. The most important regulator of the folic acid-dependent side is undoubtedly vitamin B12, which is a cofactor of methionine synthase [131]. When vitamin B12 is deficient in a cell, a phenomenon called the “folate trap” occurs [126,131]. Vitamin B12 deficiency ensures the proper functioning of methionine synthase, leading to the accumulation of 5-mTHF and the inability to metabolize it to THF. Moreover, the inactivity of this part of the one-carbon cycle pathway leads to the accumulation of hCy and a significant deficiency of SAM. A small percentage of hCy can be converted to methionine in the betaine-dependent pathway, but this route is insufficient for ensuring the adequate efficiency of methionine production [127,128,129]. The accumulating excess hCy can be transsulfurated to cystathionine in a CBS-catalyzed reaction. However, it is worth paying attention to the presence of possible molecular relationships. As it turns out, SAM has the ability to allosterically activate the CBS enzyme, which drives the transsulfuration reaction with an excess of SAM and hCy [129,130,131,132]. Taking the above into account, we believe that SAM deficiency (resulting from vitamin B12 deficiency) may lead to inhibition of CBS activity, which will ultimately lead to a decrease in hCy transsulfuration. This, in turn, will cause the accumulation of homocysteine, which will be excreted into the bloodstream. Moreover, under the influence of CBS, homocysteine is transformed into cysteine, which is the precursor of glutathione [133]. Glutathione (γ-glutamyl-cysteinyl-glycine) is the main participant in redox processes in the body [133]. Excessive concentration of hCy in the bloodstream increases its susceptibility to its oxidation, which may lead to an increase in the production of reactive oxygen species (ROS) [133]. Summing up, it is plausible that vitamin B12 deficiency may result in a decrease in SAM concentration, which will translate into inhibition of CBS activity and, as a result, may lead to a reduction in glutathione synthesis, which will affect cell redox reactions. Bearing the above in mind, we believe that improper functioning of folic acid metabolism may translate not only into a reduction in the available pool of methyl moiety donors but may also exert more long-term effects in the form of impairment of cellular redox reactions.
Folic acid appears to be crucial for the well-being of pregnancy. The demand for this nutrient increases significantly during gestation [134]. Apart from the obvious contribution to DNA synthesis processes (direct participation in the synthesis of the purine ring) and amino acid metabolism, folates also contribute to epigenetic processes [6,135]. Their role in ensuring the proper course of pregnancy applies to almost all stages of gestation [6,56].
The relationship between folic acid deficiency and neural tube defects is well established. Numerous studies indicate that folic acid deficiency during embryogenesis significantly affects the occurrence of neural tube defects (NTDs) [6,133,136,137,138,139,140,141]. There are also many reasons to believe that folic acid influences the proper closing of the neural tube through epigenic processes such as DNA methylation. First, the use of methionine cycle inhibitors or DNA methyltransferase mutations in animal models has led to the occurrence of NTDs [142]. Moreover, it has been shown that polymorphisms in genes encoding enzymes involved in the one-carbon cycle may contribute to an increased rate of NTDs [143]. MTHFR polymorphisms C677T and A1298C are associated with a greater risk of NTDs [144,145]. Additionally, the C677T MTHFR polymorphism has been linked with decreased levels of plasma folate and increased levels of hCy, as well as diminished DNA methylation [145,146,147,148,149,150]. These data clearly indicate the significant involvement of the folate-dependent pathway of the one-carbon cycle in the process of proper neural tube closing (NTC). We believe that by contribution to the methyl-group donor metabolism, folates can influence DNA methylation, which translates into the silencing/expressing of specific genes that regulate the proper course of cell migration during NTC. However, we suggest that this process should not be considered only with respect to the relation between the folate component and NTC. The relationships between all pathways of the one-carbon cycle have been demonstrated—those pathways are closely interdependent. Therefore, we believe that the remaining components of this cycle (such as choline, SAM, betaine, etc.), through their regulatory influence on folate metabolism, may also significantly influence NTC processes. However, this topic requires further thorough research.
Folic acid also appears to be pivotal for numerous physiological and metabolic pathways occurring during gestation. Referring to the research, it can be seen that folic acid takes an active part in the placentation process [125]. The placentation process mainly involves the invasion of uterine tissue by fetal trophoblast cells. Trophoblast cells penetrating the uterine spiral arteries cause their thorough reconstruction—converting them into high-capacity, low-resistant vessels, and allowing them to transport large volumes of blood [151]. The entire process consists of degradation and remodeling of the extracellular matrix (ECM), and is mediated by metalloproteinases (MMPs), accompanied by increased vasculogenesis and angiogenesis [152]. Additionally, ECM remodeling by penetrating trophoblast cells often leads to the generation of oxidative stress [153]. The research clearly shows the positive effects of folates on the placenta during early gestation. It was shown that the incubation of trophoblast tissues with folic acid solution resulted in a significant increase in trophoblast invasion, with simultaneous stimulation of MMPs secretion, which actively participates in ECM remodeling during trophoblast invasion [154]. Additionally, an increased vascular density in placental tissue was observed, which clearly indicated the intensification of angiogenesis processes [154]. Moreover, it has also been shown that folic acid deficiency may be associated with placental pathologies such as placental abruption [155,156]. Finally, low folic acid concentrations have been found in patients with preeclampsia [157]. In the context of preeclampsia, recent research indicates that folic acid supplementation may help reduce the incidence of this prenatal pathology [158]. Interestingly, folic acid supplementation has also been shown to be associated with a reduction in fetal growth restriction (FGR) occurrence [159].
We refer to pathologies such as FGR, PE, and placental abruption because these pregnancy complications mainly result from an incorrect placentation process. We draw attention to this phenomenon because it appears that folic acid may not only positively affect the placentation process per se but also could be of benefit in preventing prenatal pathologies associated with inappropriate placentation processes. Due to its multifactorial action, it is possible, not only to ensure the proper course of the placenta formation process, but also to ensure balanced, uncomplicated intrauterine fetal development.
In addition to the described impacts of folates on the proper course of gestation, it is also worth paying attention to the potential neurodevelopmental benefits of their supplementation. Animal studies have shown that folic acid deficiency during neurogenesis resulted in impaired cell migration and synaptogenesis, and a general reduction in the number of cerebellar and hippocampal neurons [160,161]. Additionally, an insufficient supply of folate during the preconception and pregnancy period is associated with an increased risk of ASD and ADHD in offspring [162,163,164]. Moreover, increased folic acid intake during pregnancy reduced the occurrence of neurodevelopmental disorders [164,165,166]. A possible explanation for these effects of folate on brain development and the risk of certain neurodevelopmental diseases may lie in epigenetic modifications, especially in DNA methylation pathways. Numerous studies indicate the involvement of folates in epigenetic regulations, mainly including DNA methylation [167,168,169]. It was shown that the maternal concentration of folic acid is proportional to the global methylation of the offspring’s genome—a high concentration of folic acid in the mother’s body correlated positively with DNA hypermethylation of the offspring’s genome [125,170,171]. Moreover, conducted research showed that patients with autism spectrum disorder had lower methionine concentrations and a significant decrease in the SAM to SAH ratio, with a simultaneous increase in homocysteine concentration [172,173,174,175,176]. Taking all of the above into consideration, we believe that folic acid deficiency (which actively participates in the metabolism of methyl donors) may translate into insufficient production of methyl donors, which may lead to alterations in specific DNA methylation patterns. This, in turn, may silence some genes and may ultimately lead to some neurodevelopmental diseases [177]. However, taking into account the multifactorial and polygenic pathogenetic nature of neurodevelopmental and cognitive disorders, we believe that this topic requires further, extensive research.
Summarizing the analyzed data, we believe that folic acid (and its metabolites) can have a significant positive impact on the course of pregnancy; not only directly, but also indirectly, through the influence on DNA methylation, they can condition the proper, undisturbed development of offspring.

5. Discussion

Apart from the obvious direct influences of substances such as folic acid and choline on proper gestation, it seems that those molecules are associated with additional indirect effects that may be determinative in the future life of the offspring. The aim of this review was to present the current state of knowledge regarding the impact of crucial substances such as choline and folic acid on maternal–fetal medicine, with particular emphasis on the impact on epigenetic mechanisms.
The presented data clearly show how important it is to ensure an adequate supply of supplements during pregnancy. Providing the correct intake applies not only to folic acid and choline but also to vitamins (such as vitamins B1, B12, and D3), inositol, and docosahexaenoic acid (DHA). Currently, more and more global medical societies recommend consuming a wide range of dietary supplements throughout pregnancy [178,179,180,181,182,183] (Table 1).
Compared to recent years, patients’ awareness of the importance of ensuring the correct concentration of supplements during the preconception period and gestation has increased significantly. Several global organizations have launched extensive awareness-raising campaigns aimed at increasing dietary supplementation during pregnancy. However, despite widely used information campaigns, it seems that the use of additional supplementation and patients’ knowledge in this area are still insufficient. Referring to the research, it can be seen that, in developed countries, the current consumption of supplements, especially folic acid and choline, is still not satisfactory [6,184,185,186,187]. Therefore, we deeply believe that it is extremely important to constantly strive to increase public awareness of the principles of proper nutrition and supplementation during pregnancy. With this in mind, we believe this approach will further reduce possible maternal and fetal complications resulting from the deficiency of key factors such as folic acid or choline. Apart from the presented data on the impact of selected dietary factors on specific genetic mechanisms determining the proper development of the fetus, it seems that the use of specific dietary patterns and maintaining appropriate diet may be associated with positive pregnancy outcomes. According to research, eating a diet rich in vegetables, fruit, whole grains, nuts, legumes, and seafood, and low in red, processed meat and fried foods is associated with a reduced risk of premature births [188]. Moreover, following such a diet may be associated with higher birth weights in newborns [188]. With the above in mind, we deeply believe that a specific, broadly held understanding of following a healthy diet may be associated with achieving better obstetric outcomes and reducing potential pregnancy and neonatal complications.
As mentioned earlier, supplementation deficiencies associated with various substances may translate into abnormal fetal development. Among other supplements, folic acid and iron deficiencies in a pregnant patient’s diet may be related to the occurrence of congenital heart defects (CHDs) [189,190,191]. Congenital heart diseases, caused by abnormal development of the heart during pregnancy, are among the most common birth defects worldwide [191]. Without the implementation of appropriate supplementation during pregnancy and proper diagnostics, CHDs lead to disastrous consequences for the newborn child. We would like to emphasize the importance of ensuring proper supplementation during pregnancy in order to minimize the risks of these dangerous diseases. However, it appears that, when a CHD is suspected, fetal echocardiography may prove to be a helpful diagnostic tool. Referring to the research, it can be seen that fetal echocardiography is a diagnostic tool with a high percentage of effectiveness in the diagnosis of heart defects [192,193]. With the above in mind, we believe that ensuring proper supplementation during pregnancy is an extremely important factor in reducing the risk of congenital heart defects. Moreover, we also believe that if supplementation deficiencies are detected during pregnancy, it is extremely important to perform fetal echocardiography, which will enable early detection of potential congenital heart defects. Thanks to this, it will be possible to provide the patient with appropriate, specialized care and implement appropriate procedures that will help reduce possible consequences for the newborn.
We would also like to draw attention to possible complications related to excessive use of individual supplements during pregnancy. Despite the well-known positive impact of folic acid supplementation on the course of pregnancy, it is worth noting that it may also have a negative impact. Referring also to large-scale campaigns to increase patients’ awareness of folic acid supplementation, concerns have arisen about excessive folic acid concentration in the entire population. Several studies show that patients can consume too much folate, which may have adverse effects [125,194,195]. Excessive folate intake during pregnancy may be positively correlated with an increased rate of allergic outcomes in offspring. Increased folate concentration was associated, in offspring, with food allergies, atopic conditions, and an increased risk of developing bronchial asthma [196,197]. A possible explanation for this phenomenon is the impact of increased folate supply on changes in the DNA methylation pattern, which may translate into the revealing of the phenotypes responsible for the development of allergies [197,198,199]. Moreover, folate oversupply may be associated with hepatotoxic effects on the maternal liver [200,201]. Nevertheless, the positive effect of folates on the proper development of the fetus and the proper course of pregnancy leaves no doubt. It is possible that the negative effects of folate supplementation result from their incorrect dosage or insufficient knowledge of patients in this area. In many countries, folic acid supplements are sold without a prescription. Therefore, many patients use them without prior consultation with a physician and are unaware of the correct regimen for taking these supplements. Therefore, we emphasize how important it is to constantly increase patients’ awareness of developing proper supplementation habits and to develop top-down dosage regimens for specific pregnancy supplements.
There are many reasons to believe that the components of the one-carbon cycle pathways (folate and choline) responsible for generating methyl-group donors are intertwined and participate in ensuring the proper rotation of the cycle. A recent study showed that pregnant women with low plasma folate had lower plasma betaine and higher DMG and tHcy, as compared to women with higher plasma folate levels [202]. Additionally, in patients with folic acid deficiency, the plasma levels of methionine and betaine remain unchanged, and the DMG concentration increases, which suggests that betaine, acting as a methyl-group donor, is able to compensate for folic acid deficiencies and ensure the proper functioning of the one-carbon cycle [202,203]. Taking the above into account, we believe that the substances involved in the metabolic pathways of the one-carbon cycle are strongly interconnected. The deficiency of one of the factors can be compensated for by increases in the activity and concentrations of the remaining components of the pathway, leading to the sustaining of the generation of methyl residues and the utilization of hCy at an appropriate level. This may suggest the presence of regulatory mechanisms operating on the basis of negative/positive feedback.
When considering the interdependence of methyl donors, we should not forget the joint impact these substances have on cells. Large amounts of choline are delivered to the fetal brain tissue across the placenta [204]. Interestingly, the placenta is one of the few organs capable of storing large amounts of choline (and its metabolites) [204]. It is highly possible that a high concentration of choline acts as a reserve, ensuring proper choline delivery to the fetal cells. Furthermore, increased choline concentration is associated with increased S-adenosylmethionine concentrations [71]. A high concentration of choline causes increased production of SAM, suggesting that the high concentration of choline in the fetal brain tissue is intended to provide large amounts of methyl residues, which directly translates into epigenetic modifications of DNA. Increased choline concentration (especially in brain tissue) undoubtedly contributes to the neurodevelopment of the central nervous system and neurogenesis. By influencing DNA methylation, choline ensures proper brain development, which translates into a reduced rate of neurocognitive and psychiatric disorders in offspring. Folic acid has similar effects—from a positive effect on the closing of the neural tube through proper placentation, to reducing the risk of behavioral disorders. The existence of the multiple mechanisms which ensure the multifactoral availability of choline and folic acid to the fetus suggests their great role in proper neurodevelopment. Moreover, it turns out that folic acid and choline can act synergistically. It has been suggested that the entirety of the one-carbon metabolism could be strongly involved in the proper neurodevelopment process. Referring to recent studies, concurrent intake of methyl donors (such as folic acid and choline) reduces the risk of NTDs to a greater extent, compared to taking only single substances such as folic acid [71,204]. This suggests that the intake of folic acid can be supplemented with the intake of choline, which may translate into a reduced risk of NTDs. This suggests that concurrent intake of several methyl donors may have a greater positive effect in terms of epigenetic modifications. This shows a new potential direction of research which may help reduce the risk of potential maternal–fetal complications. Moreover, it seems that the mentioned substances, through cooperation and mutual synergism in relation to epigenetic modifications, create a specific “DNA protection formula”, thanks to which it is possible to implement correct DNA methylation patterns; this translates not only into the undisturbed intrauterine development of the fetus, but also the future life of the offspring. However, considering that most of the research has been conducted on animal models, we believe this interesting topic requires further exploration.
Despite our great understanding of the cellular and molecular events that occur during fetal development, the search for the factors that modulate those pathways continues. A comprehensive understanding of the impacts of specific substances supplemented during pregnancy seems to be pivotal in the context of maternal–fetal medicine. In-depth knowledge in this field will enable us to provide means that can protect the woman and her unborn child from potential complications.

Author Contributions

Conceptualization, W.F. and M.W.S.; methodology, W.F. and M.W.; software, M.W.; validation, W.F. and M.W.S.; formal analysis, W.F.; investigation, W.F. and M.W.S.; resources, M.W.S.; data curation, W.F. and M.W.; writing—original draft preparation, W.F.; writing—review and editing, W.F. and M.W.S.; visualization, W.F. and M.W.; supervision, W.F. and M.W.S.; project administration, M.W.S. and W.F.; funding acquisition, M.W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zaidi, A.Z.; Moore, S.E.; Okala, S.G. Impact of Maternal Nutritional Supplementation during Pregnancy and Lactation on the Infant Gut or Breastmilk Microbiota: A Systematic Review. Nutrients 2021, 13, 1137. [Google Scholar] [CrossRef]
  2. Blondin, J.H.; LoGiudice, J.A. Pregnant women’s knowledge and awareness of nutrition. Appl. Nurs. Res. 2018, 39, 167–174. [Google Scholar] [CrossRef]
  3. Brown, B.; Wright, C. Safety and efficacy of supplements in pregnancy. Nutr. Rev. 2020, 78, 813–826. [Google Scholar] [CrossRef]
  4. Fallah, F.; Pourabbas, A.; Delpisheh, A.; Veisani, Y.; Shadnoush, M. Effects of nutrition education on levels of nutritional awareness of pregnant women in Western iran. Int. J. Endocrinol. Metab. 2013, 11, 175–178. [Google Scholar] [CrossRef] [PubMed]
  5. Jun, S.; Gahche, J.J.; Potischman, N.; Dwyer, J.T.; Guenther, P.M.; Sauder, K.A.; Bailey, R.L. Dietary Supplement Use and Its Micronutrient Contribution During Pregnancy and Lactation in the United States. Obstet. Gynecol. 2020, 135, 623–633. [Google Scholar] [CrossRef]
  6. Adams, J.B.; Kirby, J.K.; Sorensen, J.C.; Pollard, E.L.; Audhya, T. Evidence based recommendations for an optimal prenatal supplement for women in the US: Vitamins and related nutrients. Matern. Health Neonatol Perinatol 2022, 8, 4. [Google Scholar] [CrossRef]
  7. Gernand, A.D.; Schulze, K.J.; Stewart, C.P.; West, K.P., Jr.; Christian, P. Micronutrient deficiencies in pregnancy worldwide: Health effects and prevention. Nat. Rev. Endocrinol. 2016, 12, 274–289. [Google Scholar] [CrossRef] [PubMed]
  8. Zablotsky, B.; Black, L.I.; Blumberg, S.J. Estimated Prevalence of Children with Diagnosed Developmental Disabilities in the United States, 2014–2016. NCHS Data Brief. 2017, 291, 1–8. [Google Scholar]
  9. Vinyard, M.; Zimmer, M.; Herrick, K.A.; Story, M.; Juan, W.; Reedy, J. Healthy Eating Index-2015 Scores Vary by Types of Food Outlets in the United States. Nutrients 2021, 13, 2717. [Google Scholar] [CrossRef]
  10. Jaiswal, A.; Dewani, D.; Reddy, L.S.; Patel, A. Choline Supplementation in Pregnancy: Current Evidence and Implications. Cureus 2023, 15, e48538. [Google Scholar] [CrossRef]
  11. Adams, J.B.; Sorenson, J.C.; Pollard, E.L.; Kirby, J.K.; Audhya, T. Evidence-Based Recommendations for an Optimal Prenatal Supplement for Women in the U.S., Part Two: Minerals. Nutrients 2021, 13, 1849. [Google Scholar] [CrossRef]
  12. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef]
  13. Feng, L.; Lou, J. DNA Methylation Analysis. Methods Mol. Biol. 2019, 1894, 181–227. [Google Scholar] [CrossRef]
  14. Sferruzzi-Perri, A.N.; Camm, E.J. The Programming Power of the Placenta. Front. Physiol. 2016, 7, 33. [Google Scholar] [CrossRef]
  15. Barker, D.J. The developmental origins of well-being. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2004, 359, 1359–1366. [Google Scholar] [CrossRef]
  16. Abdul, Q.A.; Yu, B.P.; Chung, H.Y.; Jung, H.A.; Choi, J.S. Epigenetic modifications of gene expression by lifestyle and environment. Arch. Pharm. Res. 2017, 40, 1219–1237. [Google Scholar] [CrossRef]
  17. Bollati, V.; Baccarelli, A. Environmental epigenetics. Heredity 2010, 105, 105–112. [Google Scholar] [CrossRef]
  18. Tammen, S.A.; Friso, S.; Choi, S.W. Epigenetics: The link between nature and nurture. Mol. Asp. Med. 2013, 34, 753–764. [Google Scholar] [CrossRef]
  19. Zeisel, S.H. Epigenetic mechanisms for nutrition determinants of later health outcomes. Am. J. Clin. Nutr. 2009, 89, 1488S–1493S. [Google Scholar] [CrossRef]
  20. Zhu, Z.; Cao, F.; Li, X. Epigenetic Programming and Fetal Metabolic Programming. Front. Endocrinol. 2019, 10, 764. [Google Scholar] [CrossRef]
  21. Zeisel, S. Choline, Other Methyl-Donors and Epigenetics. Nutrients 2017, 9, 445. [Google Scholar] [CrossRef]
  22. Shenker, N.; Flanagan, J.M. Intragenic DNA methylation: Implications of this epigenetic mechanism for cancer research. Br. J. Cancer 2012, 106, 248–253. [Google Scholar] [CrossRef] [PubMed]
  23. Bestor, T.H. The DNA methyltransferases of mammals. Hum. Mol. Genet. 2000, 9, 2395–2402. [Google Scholar] [CrossRef]
  24. Mihai, D. Niculescu, Nutritional Epigenetics. ILAR J. 2012, 53, 270–278. [Google Scholar] [CrossRef]
  25. Dhar, G.A.; Saha, S.; Mitra, P.; Nag Chaudhuri, R. DNA methylation and regulation of gene expression: Guardian of our health. Nucleus 2021, 64, 259–270. [Google Scholar] [CrossRef]
  26. Long, H.K.; King, H.W.; Patient, R.K.; Odom, D.T.; Klose, R.J. Protection of CpG islands from DNA methylation is DNA-encoded and evolutionarily conserved. Nucleic Acids Res. 2016, 44, 6693–6706. [Google Scholar] [CrossRef]
  27. Verdone, L.; Caserta, M.; Di Mauro, E. Role of histone acetylation in the control of gene expression. Biochem. Cell Biol. 2005, 83, 344–353. [Google Scholar] [CrossRef]
  28. Margueron, R.; Reinberg, D. Chromatin structure and the inheritance of epigenetic information. Nat. Rev. Genet. 2010, 11, 285–296. [Google Scholar] [CrossRef]
  29. Martin, B.J.E.; Brind’Amour, J.; Kuzmin, A.; Jensen, K.N.; Liu, Z.C.; Lorincz, M.; Howe, L.J. Transcription shapes genome-wide histone acetylation patterns. Nat. Commun. 2021, 12, 210. [Google Scholar] [CrossRef]
  30. Gujral, P.; Mahajan, V.; Lissaman, A.C.; Ponnampalam, A.P. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod. Biol. Endocrinol. 2020, 18, 84. [Google Scholar] [CrossRef]
  31. Toraño, E.G.; García, M.G.; Fernández-Morera, J.L.; Niño-García, P.; Fernández, A.F. The Impact of External Factors on the Epigenome: In Utero and over Lifetime. Biomed. Res. Int. 2016, 2016, 2568635. [Google Scholar] [CrossRef]
  32. Jansson, T.; Powell, T.L. Role of the placenta in fetal programming: Underlying mechanisms and potential interventional approaches. Clin. Sci. 2007, 113, 1–13. [Google Scholar] [CrossRef]
  33. Cooney, C.A. Are somatic cells inherently deficient in methylation metabolism? A proposed mechanism for DNA methylation loss, senescence and aging. Growth Dev. Aging 1993, 57, 261–273. [Google Scholar]
  34. Greer, E.L.; Shi, Y. Histone methylation: A dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 2012, 13, 343–357. [Google Scholar] [CrossRef] [PubMed]
  35. Gong, F.; Miller, K.M. Histone methylation and the DNA damage response. Mutat. Res. Rev. Mutat. Res. 2019, 780, 37–47. [Google Scholar] [CrossRef] [PubMed]
  36. Reyes, A.A.; Marcum, R.D.; He, Y. Structure and Function of Chromatin Remodelers. J. Mol. Biol. 2021, 433, 166929. [Google Scholar] [CrossRef] [PubMed]
  37. Ravi, M.; Ramanathan, S.; Krishna, K. Factors, mechanisms and implications of chromatin condensation and chromosomal structural maintenance through the cell cycle. J. Cell Physiol. 2020, 235, 758–775. [Google Scholar] [CrossRef] [PubMed]
  38. Danielsson, B.E.; Tieu, K.V.; Spagnol, S.T.; Vu, K.K.; Cabe, J.I.; Raisch, T.B.; Dahl, K.N.; Conway, D.E. Chromatin condensation regulates endothelial cell adaptation to shear stress. Mol. Biol. Cell. 2022, 33, ar101. [Google Scholar] [CrossRef]
  39. Alver, B.H.; Kim, K.H.; Lu, P.; Wang, X.; Manchester, H.E.; Wang, W.; Haswell, J.R.; Park, P.J.; Roberts, C.W. The SWI/SNF chromatin remodelling complex is required for maintenance of lineage specific enhancers. Nat. Commun. 2017, 8, 14648. [Google Scholar] [CrossRef]
  40. Blackledge, N.P.; Klose, R.J. The molecular principles of gene regulation by Polycomb repressive complexes. Nat. Rev. Mol. Cell Biol. 2021, 22, 815–833. [Google Scholar] [CrossRef] [PubMed]
  41. Piunti, A.; Shilatifard, A. The roles of Polycomb repressive complexes in mammalian development and cancer. Nat. Rev. Mol. Cell Biol. 2021, 22, 326–345. [Google Scholar] [CrossRef]
  42. Khodadadi, E.; Fahmideh, L.; Khodadadi, E.; Dao, S.; Yousefi, M.; Taghizadeh, S.; Asgharzadeh, M.; Yousefi, B.; Kafil, H.S. Current Advances in DNA Methylation Analysis Methods. Biomed. Res. Int. 2021, 2021, 8827516. [Google Scholar] [CrossRef]
  43. Yi, P.; Melnyk, S.; Pogribna, M.; Pogribny, I.P.; Hine, R.J.; James, S.J. Increase in plasma homocysteine associated with parallel increases in plasma S-adenosylhomocysteine and lymphocyte DNA hypomethylation. J. Biol. Chem. 2000, 275, 29318–29323. [Google Scholar] [CrossRef]
  44. Mentch, S.J.; Locasale, J.W. One-carbon metabolism and epigenetics: Understanding the specificity. Ann. N. Y. Acad. Sci. 2016, 1363, 91–98. [Google Scholar] [CrossRef]
  45. Obeid, R. The metabolic burden of methyl donor deficiency with focus on the betaine homocysteine methyltransferase pathway. Nutrients 2013, 5, 3481–3495. [Google Scholar] [CrossRef]
  46. Finkelstein, J.D. Methionine metabolism in mammals. J. Nutr. Biochem. 1990, 1, 228–237. [Google Scholar] [CrossRef] [PubMed]
  47. Martínez, Y.; Li, X.; Liu, G.; Bin, P.; Yan, W.; Más, D.; Valdivié, M.; Hu, C.A.; Ren, W.; Yin, Y. The role of methionine on metabolism, oxidative stress, and diseases. Amino Acids 2017, 49, 2091–2098. [Google Scholar] [CrossRef] [PubMed]
  48. Mota-Martorell, N.; Jové, M.; Borrás, C.; Berdún, R.; Obis, È.; Sol, J.; Cabré, R.; Pradas, I.; Galo-Licona, J.D.; Puig, J.; et al.; et al. Methionine transsulfuration pathway is upregulated in long-lived humans. Free Radic. Biol. Med. 2021, 162, 38–52. [Google Scholar] [CrossRef] [PubMed]
  49. Detich, N.; Hamm, S.; Just, G.; Knox, J.D.; Szyf, M. The methyl donor S-Adenosylmethionine inhibits active demethylation of DNA: A candidate novel mechanism for the pharmacological effects of S-Adenosylmethionine. J. Biol. Chem. 2003, 278, 20812–20820. [Google Scholar] [CrossRef] [PubMed]
  50. Zaric, B.L.; Obradovic, M.; Bajic, V.; Haidara, M.A.; Jovanovic, M.; Isenovic, E.R. Homocysteine and Hyperhomocysteinaemia. Curr. Med. Chem. 2019, 26, 2948–2961. [Google Scholar] [CrossRef] [PubMed]
  51. D’Souza, S.W.; Glazier, J.D. Homocysteine Metabolism in Pregnancy and Developmental Impacts. Front. Cell Dev. Biol. 2022, 10, 802285. [Google Scholar] [CrossRef]
  52. Pérez-Miguelsanz, J.; Vallecillo, N.; Garrido, F.; Reytor, E.; Pérez-Sala, D.; Pajares, M.A. Betaine homocysteine S-methyltransferase emerges as a new player of the nuclear methionine cycle. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1165–1182. [Google Scholar] [CrossRef] [PubMed]
  53. Shulpekova, Y.; Nechaev, V.; Kardasheva, S.; Sedova, A.; Kurbatova, A.; Bueverova, E.; Kopylov, A.; Malsagova, K.; Dlamini, J.C.; Ivashkin, V. The Concept of Folic Acid in Health and Disease. Molecules 2021, 26, 3731. [Google Scholar] [CrossRef] [PubMed]
  54. Ferrazzi, E.; Tiso, G.; Di Martino, D. Folic acid versus 5- methyl tetrahydrofolate supplementation in pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 253, 312–319. [Google Scholar] [CrossRef] [PubMed]
  55. Lan, X.; Field, M.S.; Stover, P.J. Cell cycle regulation of folate-mediated one-carbon metabolism. Wiley Interdiscip. Rev. Syst. Biol. Med. 2018, 10, e1426. [Google Scholar] [CrossRef] [PubMed]
  56. Lionaki, E.; Ploumi, C.; Tavernarakis, N. One-Carbon Metabolism: Pulling the Strings behind Aging and Neurodegeneration. Cells 2022, 11, 214. [Google Scholar] [CrossRef] [PubMed]
  57. Hajkova, P.; Erhardt, S.; Lane, N.; Haaf, T.; El-Maarri, O.; Reik, W.; Walter, J.; Surani, M.A. Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 2002, 117, 15–23. [Google Scholar] [CrossRef] [PubMed]
  58. Basu, A.; Tiwari, V.K. Epigenetic reprogramming of cell identity: Lessons from development for regenerative medicine. Clin. Epigenetics 2021, 13, 144. [Google Scholar] [CrossRef] [PubMed]
  59. Leermakers, E.T.; Moreira, E.M.; Kiefte-de Jong, J.C.; Darweesh, S.K.; Visser, T.; Voortman, T.; Bautista, P.K.; Chowdhury, R.; Gorman, D.; Bramer, W.M.; et al. Effects of choline on health across the life course: A systematic review. Nutr. Rev. 2015, 73, 500–522. [Google Scholar] [CrossRef]
  60. Caudill, M.A. Pre- and postnatal health: Evidence of increased choline needs. J. Am. Diet. Assoc. 2010, 110, 1198–1206. [Google Scholar] [CrossRef]
  61. Jiang, X.; West, A.A.; Caudill, M.A. Maternal choline supplementation: A nutritional approach for improving offspring health? Trends Endocrinol. Metab. 2014, 25, 263–273. [Google Scholar] [CrossRef] [PubMed]
  62. Wiedeman, A.M.; Barr, S.I.; Green, T.J.; Xu, Z.; Innis, S.M.; Kitts, D.D. Dietary Choline Intake: Current State of Knowledge Across the Life Cycle. Nutrients 2018, 10, 1513. [Google Scholar] [CrossRef] [PubMed]
  63. Bekdash, R.A. Neuroprotective Effects of Choline and Other Methyl Donors. Nutrients 2019, 11, 2995. [Google Scholar] [CrossRef] [PubMed]
  64. Jacobs, R.L.; Stead, L.M.; Devlin, C.; Tabas, I.; Brosnan, M.E.; Brosnan, J.T.; Vance, D.E. Physiological regulation of phospholipid methylation alters plasma homocysteine in mice. J. Biol. Chem. 2005, 280, 28299–28305. [Google Scholar] [CrossRef] [PubMed]
  65. Resseguie, M.; Song, J.; Niculescu, M.D.; da Costa, K.A.; Randall, T.A.; Zeisel, S.H. Phosphatidylethanolamine N-methyltransferase (PEMT) gene expression is induced by estrogen in human and mouse primary hepatocytes. FASEB J. 2007, 21, 2622–2632. [Google Scholar] [CrossRef] [PubMed]
  66. Wortmann, S.B.; Mayr, J.A. Choline-related-inherited metabolic diseases-A mini review. J. Inherit. Metab. Dis. 2019, 42, 237–242. [Google Scholar] [CrossRef]
  67. Tayebati, S.K. Phospholipid and Lipid Derivatives as Potential Neuroprotective Compounds. Molecules 2018, 23, 2257. [Google Scholar] [CrossRef]
  68. Skripuletz, T.; Manzel, A.; Gropengießer, K.; Schäfer, N.; Gudi, V.; Singh, V.; Salinas Tejedor, L.; Jörg, S.; Hammer, A.; Voss, E.; et al. Pivotal role of choline metabolites in remyelination. Brain 2015, 138 Pt 2, 398–413. [Google Scholar] [CrossRef]
  69. Mellott, T.J.; Huleatt, O.M.; Shade, B.N.; Pender, S.M.; Liu, Y.B.; Slack, B.E.; Blusztajn, J.K. Perinatal Choline Supplementation Reduces Amyloidosis and Increases Choline Acetyltransferase Expression in the Hippocampus of the APPswePS1dE9 Alzheimer’s Disease Model Mice. PLoS ONE 2017, 12, e0170450. [Google Scholar] [CrossRef]
  70. Blusztajn, J.K.; Zeisel, S.H.; Wurtman, R.J. Developmental changes in the activity of phosphatidylethanolamine N-methyltransferases in rat brain. Biochem. J. 1985, 232, 505–511. [Google Scholar] [CrossRef]
  71. Zeisel, S.H. Choline: Critical role during fetal development and dietary requirements in adults. Annu. Rev. Nutr. 2006, 26, 229–250. [Google Scholar] [CrossRef]
  72. Hayakawa, K.; Hirosawa, M.; Tabei, Y.; Arai, D.; Tanaka, S.; Murakami, N.; Yagi, S.; Shiota, K. Epigenetic switching by the metabolism-sensing factors in the generation of orexin neurons from mouse embryonic stem cells. J. Biol. Chem. 2013, 288, 17099–17110. [Google Scholar] [CrossRef] [PubMed]
  73. Michel, V.; Yuan, Z.; Ramsubir, S.; Bakovic, M. Choline transport for phospholipid synthesis. Exp. Biol. Med. 2006, 231, 490–504. [Google Scholar] [CrossRef] [PubMed]
  74. Morita, S.Y.; Ikeda, Y. Regulation of membrane phospholipid biosynthesis in mammalian cells. Biochem. Pharmacol. 2022, 206, 115296. [Google Scholar] [CrossRef]
  75. Lykidis, A.; Jackowski, S. Regulation of mammalian cell membrane biosynthesis. Prog. Nucleic Acid Res. Mol. Biol. 2001, 65, 361–393. [Google Scholar] [CrossRef] [PubMed]
  76. Wallace, J.M.; McCormack, J.M.; McNulty, H.; Walsh, P.M.; Robson, P.J.; Bonham, M.P.; Duffy, M.E.; Ward, M.; Molloy, A.M.; Scott, J.M.; et al. Choline supplementation and measures of choline and betaine status: A randomised, controlled trial in postmenopausal women. Br. J. Nutr. 2012, 108, 1264–1271. [Google Scholar] [CrossRef] [PubMed]
  77. Arumugam, M.K.; Paal, M.C.; Donohue TMJr Ganesan, M.; Osna, N.A.; Kharbanda, K.K. Beneficial Effects of Betaine: A Comprehensive Review. Biology 2021, 10, 456. [Google Scholar] [CrossRef] [PubMed]
  78. Cho, E.; Zeisel, S.H.; Jacques, P.; Selhub, J.; Dougherty, L.; Colditz, G.A.; Willett, W.C. Dietary choline and betaine assessed by food-frequency questionnaire in relation to plasma total homocysteine concentration in the Framingham Offspring Study. Am. J. Clin. Nutr. 2006, 83, 905–911. [Google Scholar] [CrossRef] [PubMed]
  79. Chiuve, S.E.; Giovannucci, E.L.; Hankinson, S.E.; Zeisel, S.H.; Dougherty, L.W.; Willett, W.C.; Rimm, E.B. The association between betaine and choline intakes and the plasma concentrations of homocysteine in women. Am. J. Clin. Nutr. 2007, 86, 1073–1081. [Google Scholar] [CrossRef]
  80. Finkelstein, J.D.; Harris, B.J.; Kyle, W.E. Methionine metabolism in mammals: Kinetic study of betaine-homocysteine methyltransferase. Arch. Biochem. Biophys. 1972, 153, 320–324. [Google Scholar] [CrossRef]
  81. Meck, W.H.; Williams, C.L. Metabolic imprinting of choline by its availability during gestation: Implications for memory and attentional processing across the lifespan. Neurosci. Biobehav. Rev. 2003, 27, 385–399. [Google Scholar] [CrossRef] [PubMed]
  82. Meck, W.H.; Williams, C.L.; Cermak, J.M.; Blusztajn, J.K. Developmental periods of choline sensitivity provide an ontogenetic mechanism for regulating memory capacity and age-related dementia. Front. Integr. Neurosci. 2008, 1, 7. [Google Scholar] [CrossRef]
  83. Meck, W.H.; Williams, C.L. Characterization of the facilitative effects of perinatal choline supplementation on timing and temporal memory. Neuroreport 1997, 8, 2831–2835. [Google Scholar] [CrossRef]
  84. Tomizawa, H.; Matsuzawa, D.; Ishii, D.; Matsuda, S.; Kawai, K.; Mashimo, Y.; Sutoh, C.; Shimizu, E. Methyl-donor deficiency in adolescence affects memory and epigenetic status in the mouse hippocampus. Genes Brain Behav. 2015, 14, 301–309. [Google Scholar] [CrossRef]
  85. Meck, W.H.; Williams, C.L. Choline supplementation during prenatal development reduces proactive interference in spatial memory. Brain Res. Dev. Brain Res. 1999, 118, 51–59. [Google Scholar] [CrossRef]
  86. Albright, C.D.; Friedrich, C.B.; Brown, E.C.; Mar, M.H.; Zeisel, S.H. Maternal dietary choline availability alters mitosis, apoptosis and the localization of TOAD-64 protein in the developing fetal rat septum. Brain Res. Dev Brain Res. 1999, 115, 123–129. [Google Scholar] [CrossRef] [PubMed]
  87. Ernst, A.M.; Gimbel, B.A.; de Water, E.; Eckerle, J.K.; Radke, J.P.; Georgieff, M.K.; Wozniak, J.R. Prenatal and Postnatal Choline Supplementation in Fetal Alcohol Spectrum Disorder. Nutrients 2022, 14, 688. [Google Scholar] [CrossRef] [PubMed]
  88. Guo-Ross, S.X.; Clark, S.; Montoya, D.A.; Jones, K.H.; Obernier, J.; Shetty, A.K.; White, A.M.; Blusztajn, J.K.; Wilson, W.A.; Swartzwelder, H.S. Prenatal choline supplementation protects against postnatal neurotoxicity. J. Neurosci. 2002, 22, RC195. [Google Scholar] [CrossRef]
  89. Yang, Y.; Liu, Z.; Cermak, J.M.; Tandon, P.; Sarkisian, M.R.; Stafstrom, C.E.; Neill, J.C.; Blusztajn, J.K.; Holmes, G.L. Protective effects of prenatal choline supplementation on seizure-induced memory impairment. J. Neurosci. 2000, 20, RC109. [Google Scholar] [CrossRef]
  90. Boeke, C.E.; Gillman, M.W.; Hughes, M.D.; Rifas-Shiman, S.L.; Villamor, E.; Oken, E. Choline intake during pregnancy and child cognition at age 7 years. Am. J. Epidemiol. 2013, 177, 1338–1347. [Google Scholar] [CrossRef]
  91. Li, K.; Wahlqvist, M.L.; Li, D. Nutrition, One-Carbon Metabolism and Neural Tube Defects: A Review. Nutrients 2016, 8, 741. [Google Scholar] [CrossRef] [PubMed]
  92. Albright, C.D.; Tsai, A.Y.; Friedrich, C.B.; Mar, M.H.; Zeisel, S.H. Choline availability alters embryonic development of the hippocampus and septum in the rat. Brain Res. Dev. Brain Res. 1999, 113, 13–20. [Google Scholar] [CrossRef]
  93. Shaw, G.M.; Finnell, R.H.; Blom, H.J.; Carmichael, S.L.; Vollset, S.E.; Yang, W.; Ueland, P.M. Choline and risk of neural tube defects in a folate-fortified population. Epidemiology 2009, 20, 714–719. [Google Scholar] [CrossRef] [PubMed]
  94. Shaw, G.M.; Carmichael, S.L.; Yang, W.; Selvin, S.; Schaffer, D.M. Periconceptional dietary intake of choline and betaine and neural tube defects in offspring. Am. J. Epidemiol. 2004, 160, 102–109. [Google Scholar] [CrossRef]
  95. Lee, H.S.; Barraza-Villarreal, A.; Biessy, C.; Duarte-Salles, T.; Sly, P.D.; Ramakrishnan, U.; Rivera, J.; Herceg, Z.; Romieu, I. Dietary supplementation with polyunsaturated fatty acid during pregnancy modulates DNA methylation at IGF2/H19 imprinted genes and growth of infants. Physiol. Genom. 2014, 46, 851–857. [Google Scholar] [CrossRef]
  96. Popova, S.; Charness, M.E.; Burd, L.; Crawford, A.; Hoyme, H.E.; Mukherjee, R.A.S.; Riley, E.P.; Elliott, E.J. Fetal alcohol spectrum disorders. Nat. Rev. Dis. Primers. 2023, 9, 11. [Google Scholar] [CrossRef]
  97. Thomas, J.D.; Abou, E.J.; Dominguez, H.D. Prenatal choline supplementation mitigates the adverse effects of prenatal alcohol exposure on development in rats. Neurotoxicol. Teratol. 2009, 31, 303–311. [Google Scholar] [CrossRef] [PubMed]
  98. Wozniak, J.R.; Fink, B.A.; Fuglestad, A.J.; Eckerle, J.K.; Boys, C.J.; Sandness, K.E.; Radke, J.P.; Miller, N.C.; Lindgren, C.; Brearley, A.M.; et al. Four-year follow-up of a randomized controlled trial of choline for neurodevelopment in fetal alcohol spectrum disorder. J. Neurodev. Disord. 2020, 12, 9. [Google Scholar] [CrossRef]
  99. Jennings, L.; Basiri, R. Amino Acids, B Vitamins, and Choline May Independently and Collaboratively Influence the Incidence and Core Symptoms of Autism Spectrum Disorder. Nutrients 2022, 14, 2896. [Google Scholar] [CrossRef]
  100. Doyle-Thomas, K.A.; Card, D.; Soorya, L.V.; Wang, A.T.; Fan, J.; Anagnostou, E. Metabolic mapping of deep brain structures and associations with symptomatology in autism spectrum disorders. Res. Autism. Spectr. Disord. 2014, 8, 44–51. [Google Scholar] [CrossRef]
  101. Corrigan, N.M.; Shaw, D.W.; Estes, A.M.; Richards, T.L.; Munson, J.; Friedman, S.D.; Dawson, G.; Artru, A.A.; Dager, S.R. Atypical developmental patterns of brain chemistry in children with autism spectrum disorder. JAMA Psychiatry 2013, 70, 964–974. [Google Scholar] [CrossRef]
  102. Wang, L.; Zheng, R.; Xu, Y.; Zhou, Z.; Guan, P.; Wu, Y.; Zhou, J.; Cheng, Z.; Zhang, L. Altered Metabolic Characteristics in Plasma of Young Boys with Autism Spectrum Disorder. J. Autism. Dev. Disord. 2022, 52, 4897–4907. [Google Scholar] [CrossRef] [PubMed]
  103. Ross, R.G.; Hunter, S.K.; McCarthy, L.; Beuler, J.; Hutchison, A.K.; Wagner, B.D.; Leonard, S.; Stevens, K.E.; Freedman, R. Perinatal choline effects on neonatal pathophysiology related to later schizophrenia risk. Am. J. Psychiatry 2013, 170, 290–298. [Google Scholar] [CrossRef]
  104. Ross, R.G.; Hunter, S.K.; Hoffman, M.C.; McCarthy, L.; Chambers, B.M.; Law, A.J.; Leonard, S.; Zerbe, G.O.; Freedman, R. Perinatal Phosphatidylcholine Supplementation and Early Childhood Behavior Problems: Evidence for CHRNA7 Moderation. Am. J. Psychiatry 2016, 173, 509–516. [Google Scholar] [CrossRef] [PubMed]
  105. Craciunescu, C.N.; Albright, C.D.; Mar, M.H.; Song, J.; Zeisel, S.H. Choline availability during embryonic development alters progenitor cell mitosis in developing mouse hippocampus. J. Nutr. 2003, 133, 3614–3618. [Google Scholar] [CrossRef]
  106. Powers, B.E.; Velazquez, R.; Strawderman, M.S.; Ginsberg, S.D.; Mufson, E.J.; Strupp, B.J. Maternal Choline Supplementation as a Potential Therapy for Down Syndrome: Assessment of Effects Throughout the Lifespan. Front. Aging Neurosci. 2021, 13, 723046. [Google Scholar] [CrossRef] [PubMed]
  107. Moon, J.; Chen, M.; Gandhy, S.U.; Strawderman, M.; Levitsky, D.A.; Maclean, K.N.; Strupp, B.J. Perinatal choline supplementation improves cognitive functioning and emotion regulation in the Ts65Dn mouse model of Down syndrome. Behav. Neurosci. 2010, 124, 346–361. [Google Scholar] [CrossRef]
  108. Jaiswal, S.K.; Sukla, K.K.; Chauhan, A.; Lakhotia, A.R.; Kumar, A.; Rai, A.K. Choline metabolic pathway gene polymorphisms and risk for Down syndrome: An association study in a population with folate-homocysteine metabolic impairment. Eur. J. Clin. Nutr. 2017, 71, 45–50. [Google Scholar] [CrossRef]
  109. Scelfo, A.; Fachinetti, D. Keeping the Centromere under Control: A Promising Role for DNA Methylation. Cells 2019, 8, 912. [Google Scholar] [CrossRef]
  110. Jiang, X.; Bar, H.Y.; Yan, J.; Jones, S.; Brannon, P.M.; West, A.A.; Perry, C.A.; Ganti, A.; Pressman, E.; Devapatla, S.; et al. A higher maternal choline intake among third-trimester pregnant women lowers placental and circulating concentrations of the antiangiogenic factor fms-like tyrosine kinase-1 (sFLT1). FASEB J. 2013, 27, 1245–1253. [Google Scholar] [CrossRef]
  111. Jiang, X.; Jones, S.; Andrew, B.Y.; Ganti, A.; Malysheva, O.V.; Giallourou, N.; Brannon, P.M.; Roberson, M.S.; Caudill, M.A. Choline inadequacy impairs trophoblast function and vascularization in cultured human placental trophoblasts. J. Cell Physiol. 2014, 229, 1016–1027. [Google Scholar] [CrossRef] [PubMed]
  112. Kwan, S.T.C.; King, J.H.; Yan, J.; Jiang, X.; Wei, E.; Fomin, V.G.; Roberson, M.S.; Caudill, M.A. Maternal choline supplementation during murine pregnancy modulates placental markers of inflammation, apoptosis and vascularization in a fetal sex-dependent manner. Placenta 2017, 53, 57–65. [Google Scholar] [CrossRef]
  113. King, J.H.; Kwan, S.T.C.; Yan, J.; Klatt, K.C.; Jiang, X.; Roberson, M.S.; Caudill, M.A. Maternal Choline Supplementation Alters Fetal Growth Patterns in a Mouse Model of Placental Insufficiency. Nutrients 2017, 9, 765. [Google Scholar] [CrossRef] [PubMed]
  114. Morasso, M.I.; Grinberg, A.; Robinson, G.; Sargent, T.D.; Mahon, K.A. Placental failure in mice lacking the homeobox gene Dlx3. Proc. Natl. Acad. Sci. USA 1999, 96, 162–167. [Google Scholar] [CrossRef]
  115. Napoli, I.; Blusztajn, J.K.; Mellott, T.J. Prenatal choline supplementation in rats increases the expression of IGF2 and its receptor IGF2R and enhances IGF2-induced acetylcholine release in hippocampus and frontal cortex. Brain Res. 2008, 1237, 124–135. [Google Scholar] [CrossRef]
  116. Kovacheva, V.P.; Mellott, T.J.; Davison, J.M.; Wagner, N.; Lopez-Coviella, I.; Schnitzler, A.C.; Blusztajn, J.K. Gestational choline deficiency causes global and Igf2 gene DNA hypermethylation by up-regulation of Dnmt1 expression. J. Biol. Chem. 2007, 282, 31777–31788. [Google Scholar] [CrossRef] [PubMed]
  117. Xiong, F.; Zhang, L. Role of the hypothalamic-pituitary-adrenal axis in developmental programming of health and disease. Front. Neuroendocrinol. 2013, 34, 27–46. [Google Scholar] [CrossRef] [PubMed]
  118. Coussons-Read, M.E. Effects of prenatal stress on pregnancy and human development: Mechanisms and pathways. Obstet. Med. 2013, 6, 52–57. [Google Scholar] [CrossRef]
  119. Schatzberg, A.F.; Keller, J.; Tennakoon, L.; Lembke, A.; Williams, G.; Kraemer, F.B.; Sarginson, J.E.; Lazzeroni, L.C.; Murphy, G.M. HPA axis genetic variation, cortisol and psychosis in major depression. Mol. Psychiatry 2014, 19, 220–227. [Google Scholar] [CrossRef]
  120. Porter, R.J.; Gallagher, P. Abnormalities of the HPA axis in affective disorders: Clinical subtypes and potential treatments. Acta Neuropsychiatr. 2006, 18, 193–209. [Google Scholar] [CrossRef]
  121. Gluckman, P.D.; Hanson, M.A.; Beedle, A.S. Early life events and their consequences for later disease: A life history and evolutionary perspective. Am. J. Hum. Biol. 2007, 19, 1–19. [Google Scholar] [CrossRef] [PubMed]
  122. Jiang, X.; Yan, J.; West, A.A.; Perry, C.A.; Malysheva, O.V.; Devapatla, S.; Pressman, E.; Vermeylen, F.; Caudill, M.A. Maternal choline intake alters the epigenetic state of fetal cortisol-regulating genes in humans. FASEB J. 2012, 26, 3563–3574. [Google Scholar] [CrossRef] [PubMed]
  123. Thöny, B.; Auerbach, G.; Blau, N. Tetrahydrobiopterin biosynthesis, regeneration and functions. Biochem J. 2000, 347 Pt 1, 1–16. [Google Scholar] [CrossRef]
  124. Caudill, M.A. Folate bioavailability: Implications for establishing dietary recommendations and optimizing status. Am. J. Clin. Nutr. 2010, 91, 1455S–1460S. [Google Scholar] [CrossRef] [PubMed]
  125. McGee, M.; Bainbridge, S.; Fontaine-Bisson, B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr. Rev. 2018, 76, 469–478. [Google Scholar] [CrossRef] [PubMed]
  126. Zheng, Y.; Cantley, L.C. Toward a better understanding of folate metabolism in health and disease. J. Exp. Med. 2019, 216, 253–266. [Google Scholar] [CrossRef]
  127. Obeid, R.; Fedosov, S.N.; Nexo, E. Cobalamin coenzyme forms are not likely to be superior to cyano- and hydroxyl-cobalamin in prevention or treatment of cobalamin deficiency. Mol. Nutr. Food Res. 2015, 59, 1364–1372. [Google Scholar] [CrossRef] [PubMed]
  128. Castaño, E.; Piñuñuri, R.; Hirsch, S.; Ronco, A.M. Folatos y Embarazo, conceptos actuales: ¿Es necesaria una suplementación con Acido Fólico? [Folate and Pregnancy, current concepts: It is required folic acid supplementation?]. Rev. Chil. Pediatr. 2017, 88, 199–206. [Google Scholar] [CrossRef]
  129. Menezo, Y.; Elder, K.; Clement, A.; Clement, P. Folic Acid, Folinic Acid, 5 Methyl TetraHydroFolate Supplementation for Mutations That Affect Epigenesis through the Folate and One-Carbon Cycles. Biomolecules 2022, 12, 197. [Google Scholar] [CrossRef]
  130. Annibal, A.; Tharyan, R.G.; Schonewolff, M.F.; Tam, H.; Latza, C.; Auler, M.M.K.; Grönke, S.; Partridge, L.; Antebi, A. Regulation of the one carbon folate cycle as a shared metabolic signature of longevity. Nat. Commun. 2021, 12, 3486. [Google Scholar] [CrossRef]
  131. Schaevitz, L.; Berger-Sweeney, J.; Ricceri, L. One-carbon metabolism in neurodevelopmental disorders: Using broad-based nutraceutics to treat cognitive deficits in complex spectrum disorders. Neurosci. Biobehav. Rev. 2014, 46 Pt 2, 270–284. [Google Scholar] [CrossRef]
  132. Couto, N.; Wood, J.; Barber, J. The role of glutathione reductase and related enzymes on cellular redox homoeostasis network. Free Radic. Biol. Med. 2016, 95, 27–42. [Google Scholar] [CrossRef] [PubMed]
  133. van Gool, J.D.; Hirche, H.; Lax, H.; De Schaepdrijver, L. Folic acid and primary prevention of neural tube defects: A review. Reprod. Toxicol. 2018, 80, 73–84. [Google Scholar] [CrossRef] [PubMed]
  134. Bruinse, H.W.; van den Berg, H. Changes of some vitamin levels during and after normal pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 1995, 61, 31–37. [Google Scholar] [CrossRef]
  135. Fang, J.Y.; Xiao, S.D. Folic acid, polymorphism of methyl-group metabolism genes, and DNA methylation in relation to GI carcinogenesis. J. Gastroenterol. 2003, 38, 821–829. [Google Scholar] [CrossRef]
  136. Barchitta, M.; Maugeri, A.; Magnano San Lio, R.; Favara, G.; La Mastra, C.; La Rosa, M.C.; Agodi, A. Dietary Folate Intake and Folic Acid Supplements among Pregnant Women from Southern Italy: Evidence from the “Mamma & Bambino” Cohort. Int. J. Environ. Res. Public Health 2020, 17, 638. [Google Scholar] [CrossRef]
  137. Wald, N.J. Folic acid and neural tube defects: Discovery, debate and the need for policy change. J. Med. Screen 2022, 29, 138–146. [Google Scholar] [CrossRef]
  138. Jonker, H.; Capelle, N.; Lanes, A.; Wen, S.W.; Walker, M.; Corsi, D.J. Maternal folic acid supplementation and infant birthweight in low- and middle-income countries: A systematic review. Matern. Child. Nutr. 2020, 16, e12895. [Google Scholar] [CrossRef]
  139. Willoughby, M.L.; Jewell, F.J. Investigation of folic acid requirements in pregnancy. Br. Med. J. 1966, 2, 1568–1571. [Google Scholar] [CrossRef]
  140. Engelhardt, D.M.; Martyr, C.A.; Niswander, L. Pathogenesis of neural tube defects: The regulation and disruption of cellular processes underlying neural tube closure. WIREs Mech. Dis. 2022, 14, e1559. [Google Scholar] [CrossRef]
  141. Berry, R.J.; Li, Z.; Erickson, J.D.; Li, S.; Moore, C.A.; Wang, H.; Mulinare, J.; Zhao, P.; Wong, L.Y.; Gindler, J.; et al. Prevention of neural-tube defects with folic acid in China. China-U.S. Collaborative Project for Neural Tube Defect Prevention. N. Engl. J. Med. 1999, 341, 1485–1490. [Google Scholar] [CrossRef]
  142. Dunlevy, L.P.; Burren, K.A.; Chitty, L.S.; Copp, A.J.; Greene, N.D. Excess methionine suppresses the methylation cycle and inhibits neural tube closure in mouse embryos. FEBS Lett. 2006, 580, 2803–2807. [Google Scholar] [CrossRef]
  143. Friso, S.; Choi, S.W.; Girelli, D.; Mason, J.B.; Dolnikowski, G.G.; Bagley, P.J.; Olivieri, O.; Jacques, P.F.; Rosenberg, I.H.; Corrocher, R.; et al. A common mutation in the 5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation through an interaction with folate status. Proc. Natl. Acad. Sci. USA 2002, 99, 5606–5611. [Google Scholar] [CrossRef]
  144. Cai, C.Q.; Fang, Y.L.; Shu, J.B.; Zhao, L.S.; Zhang, R.P.; Cao, L.R.; Wang, Y.Z.; Zhi, X.F.; Cui, H.L.; Shi, O.Y.; et al. Association of neural tube defects with maternal alterations and genetic polymorphisms in one-carbon metabolic pathway. Ital. J. Pediatr. 2019, 45, 37. [Google Scholar] [CrossRef]
  145. Ouyang, S.; Li, Y.; Liu, Z.; Chang, H.; Wu, J. Association between MTR A2756G and MTRR A66G polymorphisms and maternal risk for neural tube defects: A meta-analysis. Gene 2013, 515, 308–312. [Google Scholar] [CrossRef]
  146. Gaughan, D.J.; Kluijtmans, L.A.; Barbaux, S.; McMaster, D.; Young, I.S.; Yarnell, J.W.; Evans, A.; Whitehead, A.S. The methionine synthase reductase (MTRR) A66G polymorphism is a novel genetic determinant of plasma homocysteine concentrations. Atherosclerosis 2001, 157, 451–456. [Google Scholar] [CrossRef]
  147. Liew, S.C.; Gupta, E.D. Methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism: Epidemiology, metabolism and the associated diseases. Eur. J. Med. Genet. 2015, 58, 1–10. [Google Scholar] [CrossRef]
  148. 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, 488–500. [Google Scholar] [CrossRef]
  149. Wang, Y.; Liu, Y.; Ji, W.; Qin, H.; Wu, H.; Xu, D.; Turtuohut, T.; Wang, Z. Variants in MTHFR gene and neural tube defects susceptibility in China. Metab. Brain Dis. 2015, 30, 1017–1026. [Google Scholar] [CrossRef]
  150. Wilson, R.D.; Genetics Committee; Wilson, R.D.; Audibert, F.; Brock, J.A.; Carroll, J.; Cartier, L.; Gagnon, A.; Johnson, J.A.; Langlois, S.; et al. Pre-conception Folic Acid and Multivitamin Supplementation for the Primary and Secondary Prevention of Neural Tube Defects and Other Folic Acid-Sensitive Congenital Anomalies. J. Obstet. Gynaecol. Can. 2015, 37, 534–552. [Google Scholar] [CrossRef]
  151. Burton, G.J.; Jauniaux, E. What is the placenta? Am. J. Obstet. Gynecol. 2015, 213 (Suppl. S4), S6.e1–S6.e4. [Google Scholar] [CrossRef]
  152. Pijnenborg, R.; Vercruysse, L.; Hanssens, M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta 2006, 27, 939–958. [Google Scholar] [CrossRef]
  153. Raijmakers, M.T.; Burton, G.J.; Jauniaux, E.; Seed, P.T.; Peters, W.H.; Steegers, E.A.; Poston, L. Placental NAD(P)H oxidase mediated superoxide generation in early pregnancy. Placenta 2006, 27, 158–163. [Google Scholar] [CrossRef]
  154. Williams, P.J.; Bulmer, J.N.; Innes, B.A.; Broughton Pipkin, F. Possible roles for folic acid in the regulation of trophoblast invasion and placental development in normal early human pregnancy. Biol. Reprod. 2011, 84, 1148–1153. [Google Scholar] [CrossRef]
  155. Pickell, L.; Li, D.; Brown, K.; Mikael, L.G.; Wang, X.L.; Wu, Q.; Luo, L.; Jerome-Majewska, L.; Rozen, R. Methylenetetrahydrofolate reductase deficiency and low dietary folate increase embryonic delay and placental abnormalities in mice. Birth Defects Res. A Clin. Mol. Teratol. 2009, 85, 531–541. [Google Scholar] [CrossRef] [PubMed]
  156. Wen, S.W.; Zhou, J.; Yang, Q.; Fraser, W.; Olatunbosun, O.; Walker, M. Maternal exposure to folic acid antagonists and placenta-mediated adverse pregnancy outcomes. CMAJ 2008, 179, 1263–1268. [Google Scholar] [CrossRef] [PubMed]
  157. Mistry, H.D.; Mather, J.; Ramsay, M.M.; Kurlak, L.O.; Symonds, M.E.; Broughton Pipkin, F. Homocysteine and folate plasma concentrations in mother and baby at delivery after pre-eclamptic or normotensive pregnancy: Influence of parity. Pregnancy Hypertens. 2011, 1, 150–155. [Google Scholar] [CrossRef] [PubMed]
  158. Wen, S.W.; Chen, X.K.; Rodger, M.; White, R.R.; Yang, Q.; Smith, G.N.; Sigal, R.J.; Perkins, S.L.; Walker, M.C. Folic acid supplementation in early second trimester and the risk of preeclampsia. Am. J. Obstet. Gynecol. 2008, 198, 45.e1–45.e7. [Google Scholar] [CrossRef]
  159. Zhao, M.; Chen, Y.H.; Dong, X.T.; Zhou, J.; Chen, X.; Wang, H.; Wu, S.X.; Xia, M.Z.; Zhang, C.; Xu, D.X. Folic acid protects against lipopolysaccharide-induced preterm delivery and intrauterine growth restriction through its anti-inflammatory effect in mice. PLoS ONE 2013, 8, e82713. [Google Scholar] [CrossRef]
  160. Balashova, O.A.; Visina, O.; Borodinsky, L.N. Folate action in nervous system development and disease. Dev. Neurobiol. 2018, 78, 391–402. [Google Scholar] [CrossRef]
  161. Yang, Y.; Li, X.; Sun, Q.; He, B.; Jia, Y.; Cai, D.; Zhao, R. Folate deprivation induces cell cycle arrest at G0/G1 phase and apoptosis in hippocampal neuron cells through down-regulation of IGF-1 signaling pathway. Int. J. Biochem. Cell Biol. 2016, 79, 222–230. [Google Scholar] [CrossRef] [PubMed]
  162. Li, M.; Francis, E.; Hinkle, S.N.; Ajjarapu, A.S.; Zhang, C. Preconception and Prenatal Nutrition and Neurodevelopmental Disorders: A Systematic Review and Meta-Analysis. Nutrients 2019, 11, 1628. [Google Scholar] [CrossRef] [PubMed]
  163. Raghavan, R.; Selhub, J.; Paul, L.; Ji, Y.; Wang, G.; Hong, X.; Zuckerman, B.; Fallin, M.D.; Wang, X. A prospective birth cohort study on cord blood folate subtypes and risk of autism spectrum disorder. Am. J. Clin. Nutr. 2020, 112, 1304–1317. [Google Scholar] [CrossRef]
  164. DeVilbiss, E.A.; Gardner, R.M.; Newschaffer, C.J.; Lee, B.K. Maternal folate status as a risk factor for autism spectrum disorders: A review of existing evidence. Br. J. Nutr. 2015, 114, 663–672. [Google Scholar] [CrossRef]
  165. Riahi, F.; Tashakori, A.; Vanani, G.S. Effects of Folic Acid on Appetite in Children with Attention Deficit Hyperactivity Disorder (ADHD) Treated with Methylphenidate: A Randomized Double-Blind Clinical Trial. Iran J. Med. Sci. 2018, 43, 9–17. [Google Scholar]
  166. Surén, P.; Roth, C.; Bresnahan, M.; Haugen, M.; Hornig, M.; Hirtz, D.; Lie, K.K.; Lipkin, W.I.; Magnus, P.; Reichborn-Kjennerud, T.; et al. Association between maternal use of folic acid supplements and risk of autism spectrum disorders in children. JAMA 2013, 309, 570–577. [Google Scholar] [CrossRef]
  167. Michels, K.B.; Binder, A.M. Impact of folic acid supplementation on the epigenetic profile in healthy unfortified individuals—A randomized intervention trial. Epigenetics 2024, 19, 2293410. [Google Scholar] [CrossRef]
  168. Zsigrai, S.; Kalmár, A.; Barták, B.K.; Nagy, Z.B.; Szigeti, K.A.; Valcz, G.; Kothalawala, W.; Dankó, T.; Sebestyén, A.; Barna, G.; et al. Folic Acid Treatment Directly Influences the Genetic and Epigenetic Regulation along with the Associated Cellular Maintenance Processes of HT-29 and SW480 Colorectal Cancer Cell Lines. Cancers 2022, 14, 1820. [Google Scholar] [CrossRef]
  169. Kruman, I.I.; Mouton, P.R.; Emokpae, R., Jr.; Cutler, R.G.; Mattson, M.P. Folate deficiency inhibits proliferation of adult hippocampal progenitors. Neuroreport 2005, 16, 1055–1059. [Google Scholar] [CrossRef]
  170. Liu, H.Y.; Liu, S.M.; Zhang, Y.Z. Maternal Folic Acid Supplementation Mediates Offspring Health via DNA Methylation. Reprod. Sci. 2020, 27, 963–976. [Google Scholar] [CrossRef]
  171. Richmond, R.C.; Sharp, G.C.; Herbert, G.; Atkinson, C.; Taylor, C.; Bhattacharya, S.; Campbell, D.; Hall, M.; Kazmi, N.; Gaunt, T.; et al. The long-term impact of folic acid in pregnancy on offspring DNA methylation: Follow-up of the Aberdeen Folic Acid Supplementation Trial (AFAST). Int. J. Epidemiol. 2018, 47, 928–937. [Google Scholar] [CrossRef]
  172. Dutta, S.; Chatterjee, A.; Sinha, S.; Chattopadhyay, A.; Mukhopadhyay, K. Correlation between cystathionine beta synthase gene polymorphisms, plasma homocysteine and idiopathic mental retardation in Indian individuals from Kolkata. Neurosci. Lett. 2009, 453, 214–218. [Google Scholar] [CrossRef]
  173. Saha, T.; Chatterjee, M.; Sinha, S.; Rajamma, U.; Mukhopadhyay, K. Components of the folate metabolic pathway and ADHD core traits: An exploration in eastern Indian probands. J. Hum. Genet. 2017, 62, 687–695. [Google Scholar] [CrossRef]
  174. Ornoy, A.; Weinstein-Fudim, L.; Tfilin, M.; Ergaz, Z.; Yanai, J.; Szyf, M.; Turgeman, G. S-adenosyl methionine prevents ASD like behaviors triggered by early postnatal valproic acid exposure in very young mice. Neurotoxicol. Teratol. 2019, 71, 64–74. [Google Scholar] [CrossRef]
  175. Guo, B.Q.; Ding, S.B.; Li, H.B. Blood biomarker levels of methylation capacity in autism spectrum disorder: A systematic review and meta-analysis. Acta Psychiatr. Scand. 2020, 141, 492–509. [Google Scholar] [CrossRef]
  176. Lintas, C. Linking genetics to epigenetics: The role of folate and folate-related pathways in neurodevelopmental disorders. Clin. Genet. 2019, 95, 241–252. [Google Scholar] [CrossRef]
  177. Xu, Y.; Chen, X.T.; Luo, M.; Tang, Y.; Zhang, G.; Wu, D.; Yang, B.; Ruan, D.Y.; Wang, H.L. Multiple epigenetic factors predict the attention deficit/hyperactivity disorder among the Chinese Han children. J. Psychiatr. Res. 2015, 64, 40–50. [Google Scholar] [CrossRef]
  178. Skrypnik, D.; Moszak, M.; Wender-Ozegowska, E.; Bogdanski, P. Comparison of Polish and international guidelines on diet supplements in pregnancy—Review. Ginekol. Pol. 2021, 92, 322–330. [Google Scholar] [CrossRef]
  179. Wender-Ożegowska, E.; Bomba-Opoń, D.; Brązert, J.; Celewicz, Z.; Czajkowski, K.; Gutaj, P.; Malinowska-Polubiec, A.; Zawiejska, A.; Wielgoś, M. Standards of Polish Society of Gynecologists and Obstetricians in management of women with diabetes. Ginekol. Pol. 2018, 89, 341–350. [Google Scholar] [CrossRef] [PubMed]
  180. Bomba-Opoń, D.; Hirnle, L.; Kalinka, J.; Seremak-Mrozikiewicz, A. Folate supplementation during the preconception period, pregnancy and puerperium. Polish Society of Gynecologists and Obstetricians Guidelines. Ginekol. Pol. 2017, 88, 633–636. [Google Scholar] [CrossRef] [PubMed]
  181. ACOG Committee Opinion. No. 495. Vitamin D: Screening and supplementation during pregnancy. Obstet. Gynecol. 2011, 118, 197–198. [Google Scholar] [CrossRef]
  182. Toriello, H.V.; Policy and Practice Guideline Committee of the American College of Medical Genetics. Policy statement on folic acid and neural tube defects. Genet. Med. 2011, 13, 593–596. [Google Scholar] [CrossRef]
  183. Antenatal Care for Uncomplicated Pregnancies; National Institute for Health and Care Excellence (NICE): London, UK, 2019.
  184. Ren, A.; Zhang, L.; Li, Z.; Hao, L.; Tian, Y.; Li, Z. Awareness and use of folic acid, and blood folate concentrations among pregnant women in northern China-an area with a high prevalence of neural tube defects. Reprod. Toxicol. 2006, 22, 431–436. [Google Scholar] [CrossRef]
  185. Hlushko, K.; Boyarchuk, O.; Kinash, M.; Burbela, E.; Rohalska, Y.; Dobrovolska, L. Awareness of folic acid use and its effects among medical students in Ukraine. Wiad. Lek. 2021, 74, 2033–2038. [Google Scholar] [CrossRef]
  186. Derbyshire, E.; Obeid, R.; Schön, C. Habitual Choline Intakes across the Childbearing Years: A Review. Nutrients 2021, 13, 4390. [Google Scholar] [CrossRef]
  187. Gómez, M.F.; Field, C.J.; Olstad, D.L.; Loehr, S.; Ramage, S.; McCargar, L.J.; APrON Study Team. Use of micronutrient supplements among pregnant women in Alberta: Results from the Alberta Pregnancy Outcomes and Nutrition (APrON) cohort. Matern. Child. Nutr. 2015, 11, 497–510. [Google Scholar] [CrossRef]
  188. Raghavan, R.; Dreibelbis, C.; Kingshipp, B.L.; Wong, Y.P.; Abrams, B.; Gernand, A.D.; Rasmussen, K.M.; Siega-Riz, A.M.; Stang, J.; Casavale, K.O.; et al. Dietary patterns before and during pregnancy and birth outcomes: A systematic review. Am. J. Clin. Nutr. 2019, 109 (Suppl. S7), 729S–756S. [Google Scholar] [CrossRef]
  189. Obeid, R.; Holzgreve, W.; Pietrzik, K. Folate supplementation for prevention of congenital heart defects and low birth weight: An update. Cardiovasc. Diagn Ther. 2019, 9 (Suppl. S2), S424–S433. [Google Scholar] [CrossRef]
  190. Cheng, Z.; Gu, R.; Lian, Z.; Gu, H.F. Evaluation of the association between maternal folic acid supplementation and the risk of congenital heart disease: A systematic review and meta-analysis. Nutr. J. 2022, 21, 20. [Google Scholar] [CrossRef]
  191. Yan, M.X.; Zhao, Y.; Zhao, D.D.; Dang, S.N.; Zhang, R.; Duan, X.Y.; Rong, P.X.; Dang, Y.S.; Pei, L.L.; Qu, P.F. The Association of Folic Acid, Iron Nutrition during Pregnancy and Congenital Heart Disease in Northwestern China: A Matched Case-Control Study. Nutrients 2022, 14, 4541. [Google Scholar] [CrossRef]
  192. Liu, H.; Zhou, J.; Feng, Q.L.; Gu, H.T.; Wan, G.; Zhang, H.M.; Xie, Y.J.; Li, X.S. Fetal echocardiography for congenital heart disease diagnosis: A meta-analysis, power analysis and missing data analysis. Eur. J. Prev. Cardiol. 2015, 22, 1531–1547. [Google Scholar] [CrossRef]
  193. Mozumdar, N.; Rowland, J.; Pan, S.; Rajagopal, H.; Geiger, M.K.; Srivastava, S.; Stern, K.W.D. Diagnostic Accuracy of Fetal Echocardiography in Congenital Heart Disease. J. Am. Soc. Echocardiogr. 2020, 33, 1384–1390. [Google Scholar] [CrossRef]
  194. Masih, S.P.; Plumptre, L.; Ly, A.; Berger, H.; Lausman, A.Y.; Croxford, R.; Kim, Y.I.; O’Connor, D.L. Pregnant Canadian Women Achieve Recommended Intakes of One-Carbon Nutrients through Prenatal Supplementation but the Supplement Composition, Including Choline, Requires Reconsideration. J. Nutr. 2015, 145, 1824–1834. [Google Scholar] [CrossRef]
  195. McGowan, E.C.; Hong, X.; Selhub, J.; Paul, L.; Wood, R.A.; Matsui, E.C.; Keet, C.A.; Wang, X. Association Between Folate Metabolites and the Development of Food Allergy in Children. J. Allergy Clin. Immunol. Pract. 2020, 8, 132–140.e5. [Google Scholar] [CrossRef]
  196. Whitrow, M.J.; Moore, V.M.; Rumbold, A.R.; Davies, M.J. Effect of supplemental folic acid in pregnancy on childhood asthma: A prospective birth cohort study. Am. J. Epidemiol. 2009, 170, 1486–1493. [Google Scholar] [CrossRef]
  197. Yang, F.; Zhu, J.; Wang, Z.; Wang, L.; Tan, T.; Sun, L. Relationship between maternal folic acid supplementation during pregnancy and risk of childhood asthma: Systematic review and dose-response meta-analysis. Front. Pediatr. 2022, 10, 1000532. [Google Scholar] [CrossRef]
  198. Patel, K.R.; Sobczyńska-Malefora, A. The adverse effects of an excessive folic acid intake. Eur. J. Clin. Nutr. 2017, 71, 159–163. [Google Scholar] [CrossRef]
  199. Christensen, K.E.; Mikael, L.G.; Leung, K.Y.; Lévesque, N.; Deng, L.; Wu, Q.; Malysheva, O.V.; Best, A.; Caudill, M.A.; Greene, N.D.; et al. High folic acid consumption leads to pseudo-MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am. J. Clin. Nutr. 2015, 101, 646–658. [Google Scholar] [CrossRef]
  200. Fernàndez-Roig, S.; Cavallé-Busquets, P.; Fernandez-Ballart, J.D.; Ballesteros, M.; Berrocal-Zaragoza, M.I.; Salat-Batlle, J.; Ueland, P.M.; Murphy, M.M. Low folate status enhances pregnancy changes in plasma betaine and dimethylglycine concentrations and the association between betaine and homocysteine. Am. J. Clin. Nutr. 2013, 97, 1252–1259. [Google Scholar] [CrossRef]
  201. McGregor, D.O.; Dellow, W.J.; Robson, R.A.; Lever, M.; George, P.M.; Chambers, S.T. Betaine supplementation decreases post-methionine hyperhomocysteinemia in chronic renal failure. Kidney Int. 2002, 61, 1040–1046. [Google Scholar] [CrossRef]
  202. Allen, R.H.; Stabler, S.P.; Savage, D.G.; Lindenbaum, J. Metabolic abnormalities in cobalamin (vitamin B12) and folate deficiency. FASEB J. 1993, 7, 1344–1353. [Google Scholar] [CrossRef] [PubMed]
  203. Sweiry, J.H.; Page, K.R.; Dacke, C.G.; Abramovich, D.R.; Yudilevich, D.L. Evidence of saturable uptake mechanisms at maternal and fetal sides of the perfused human placenta by rapid paired-tracer dilution: Studies with calcium and choline. J. Dev. Physiol. 1986, 8, 435–445. [Google Scholar]
  204. Petersen, J.M.; Smith-Webb, R.S.; Shaw, G.M.; Carmichael, S.L.; Desrosiers, T.A.; Nestoridi, E.; Darling, A.M.; Parker, S.E.; Politis, M.D.; Yazdy, M.M.; et al. Periconceptional intakes of methyl donors and other micronutrients involved in one-carbon metabolism may further reduce the risk of neural tube defects in offspring: A United States population-based case-control study of women meeting the folic acid recommendations. Am. J. Clin. Nutr. 2023, 118, 720–728. [Google Scholar] [CrossRef]
Figure 1. Diagram showing an overview of epigenetic mechanisms.
Figure 1. Diagram showing an overview of epigenetic mechanisms.
Nutrients 16 00678 g001
Figure 2. Diagram showing the course of the one-carbon cycle. MTHFR—methylenetetrahydrofolate reductase; DHFR—dihydrofolate reductase; MS—methionine synthase; SHMT—serine hydroxymethyltransferase; CBS—cystathionine β-synthase; 5-MTHF—5-methyltetrahydrofolate.
Figure 2. Diagram showing the course of the one-carbon cycle. MTHFR—methylenetetrahydrofolate reductase; DHFR—dihydrofolate reductase; MS—methionine synthase; SHMT—serine hydroxymethyltransferase; CBS—cystathionine β-synthase; 5-MTHF—5-methyltetrahydrofolate.
Nutrients 16 00678 g002
Table 1. Table summarizing the recommendations of various medical societies regarding the supplementation of selected nutrients during pregnancy. DHA: docosahexaenoic acid.
Table 1. Table summarizing the recommendations of various medical societies regarding the supplementation of selected nutrients during pregnancy. DHA: docosahexaenoic acid.
Dietary SupplementAmerican College of Obstetricians and GynecologistsNational Institute for Health and Care Excellence/Royal College of Obstetricians and Gynecologists (NICE/RJOG)The Polish Society of Gynecologists and Obstetricians
Folic acid0.4 mg–0.6 mg/day0.4 mg0.4 mg–5 mg (depending on pregnancy complications)
Iodine200 μg/dayNot applicable150–200 μg/day
Vitamin D1000–2000 IU/day1000 IU/day1500–2000 IU/day
DHANot applicableNot applicable600 mg/day
Choline450 mg/day400 mg/dayNot applicable
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Socha, M.W.; Flis, W.; Wartęga, M. Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation. Nutrients 2024, 16, 678. https://doi.org/10.3390/nu16050678

AMA Style

Socha MW, Flis W, Wartęga M. Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation. Nutrients. 2024; 16(5):678. https://doi.org/10.3390/nu16050678

Chicago/Turabian Style

Socha, Maciej W., Wojciech Flis, and Mateusz Wartęga. 2024. "Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation" Nutrients 16, no. 5: 678. https://doi.org/10.3390/nu16050678

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop