Abstract
Studies have demonstrated that epigenetic changes such as DNA methylation, histone modification, and chromatin remodeling are linked to an increased inflammatory response as well as increased risk of chronic disease development. A few studies have begun to investigate whether dietary nutrients play a beneficial role by modifying or reversing epigenetically induced inflammation. Results of these studies show that nutrients modify epigenetic pathways. However, little is known about how nutrients modulate inflammation by regulating immune cell function and/or immune cell differentiation via epigenetic pathways. This overview will provide information about the current understanding of the role of nutrients in the epigenetic control mechanisms of immune function.
Keywords: epigenetics, immunity, inflammation, nutrients, macrophages, obesity
Modes of Epigenetic Modifications
Epigenetics is the study of heritable changes that affect gene expression without DNA base pair sequence changes (1). One of the best-known examples of epigenetic alterations involves DNA methylation of carbon number 5 of nucleotide base cytosines on 5′ in CpG islands (2) by methyl transferases. In the early 1990s, it was demonstrated that inherited genes function differently depending on whether they were from the mother or the father (3–5). During the developmental period, de novo methylation of CpG islands occurs in specific genes responsible for early embryonic development (6) and the levels of CpG island methylation can vary in tissue-specific methylation patterns (7). One of the best-known examples of an imprinted gene that is expressed in a parent-specific manner is insulin-like growth factor 2 (Igf2)6. Igf2 coding for fetal growth factor is expressed from the paternal allele and highly methylated in fetal liver but is undermethylated in fetal brain (7). The expression of Igf2 is regulated by maternal nutrient intake during fetal development (8–10), potentially resulting in disease phenotypes including obesity (10). Another well-known epigenetics modification involves histone modifications that include methylation, phosphorylation, ubiquitination, and acetylation (1). Together with CpG island methylation, histone modifications affect how accessible nucleosomes are to transcription machinery and transcription activation (11). Histone modifications may also change the binding capacity of other proteins to histones via alterations of local hydrophobicity (11) and by changing activation of RNA polymerase and other transcription coactivator binding (12).
Immune Dysfunction, Inflammation, and Epigenetics
Environmental stimuli, such as toxins or infectious agents, can alter immune cell activation through epigenetic modifications. For example, immune cell activation by pathogens such as bacteria was shown to induce changes in histone modifications, DNA methylation, and chromatin structure. These changes induce inflammatory gene transcription by programming the host immune cells (13). Although the role of epigenetic modifications in inflammatory diseases has been attracting increased attention recently, the exact epigenetic mechanisms underlying the different inflammatory diseases are poorly defined. Among the recent studies addressing the immune cell epigenetic modification leading to alterations in inflammatory status, many were conducted with the use of T lymphocytes. For example, activated lymphocytes have euchromatin, which is loosely coiled, transcriptionally active, and accessible, whereas naive and resting cells contain compact nuclei with inaccessible heterochromatin (14). Upon activation, T cells recruit the SWItch/sucrose nonfermentable (SWI/SNF) complex to the chromatin to control gene expression (14). SWI/SNF complexes are chromatin-remodeling complexes that use ATP hydrolysis to remodel nucleosomes and to modulate transcription (15). Epigenetic regulation, including histone modification and DNA methylation, is important for T cell differentiation, including T helper (Th) 1 and Th2 subsets, as well as for regulatory T cells and the Th17 lineage (16, 17). For example, histone modifications result in cluster of differentiation 4 (CD4)+ T cell differentiation into the Th2 phenotype (18). Native CD4+ T cells differentiate into specific Th cell phenotypes by chromatin remodeling, allowing increased binding to the regulator regions of interferon γ (IFN-γ) or IL-4 genes (19). Increased Th1 cells that secrete high amounts of IFN-γ are critical for host defense against intracellular pathogens by activating macrophages, whereas increased Th2 cells that produce high amounts of IL-4 are essential for a humoral immune response (2). Altering chromatin structure at the promoters for the Ifn-γ or Il-4 genes can tip the balance in the case of inflammatory lung disease; administration of a gram-negative bacterium to pregnant mice prevented an asthmalike phenotype in the progeny through epigenetic modifications (20). This protection was mediated via increased histone 4 (H4) acetylation in the Ifn-γ promoter and decreased H4 acetylation at the Il-4 promoter–dependent pathway, preventing a Th2 inflammatory response (20). DNA methylation changes also play a role in Th1/Th2-specific gene expression; for example, in a model of experimental asthma, CD4+ T cells experienced an increase in DNA methylation at the Ifn-γ promoter upon allergen challenge (21). Other factors that control T cell function such as IL-2, a T cell proliferative factor, are regulated via IL-6 (Il-6) promoter demethylation in T cells (22).
Another major immune cell type that has been studied widely is the macrophage. Macrophages are key regulatory cell types in the immune response and can be polarized to relatively stable macrophage type 1 or 2 (M1 or M2) phenotypes in vitro and in vivo. Stimulation with cytokines such as IFN-γ and/or by bacterial and viral products leads to M1 polarization. This polarization results in epigenetic changes including DNA methylation and acetylation and methylation of histones of genes involved in the inflammatory process. M1 polarized macrophages are involved in the immune response during inflammation, extracellular matrix destruction, immune response to tumors, and apoptosis and mediate the proinflammatory effects attributed to adipose tissue macrophages. This polarization is dependent on many factors. Stimulation with cytokines such as IL-4, IL-13, or IL-10 and/or certain parasite-derived antigens or allergens leads to M2 polarization. This polarization is dependent on induction of demethylases that act on histones that bind to the promoter of genes associated with the M2 phenotype (23). M2 polarized macrophages are involved in the immune response during parasitic and allergic diseases as well as insulin resistance, atherosclerosis, extracellular matrix remodeling, cell proliferation, and angiogenesis. The histone lysine demethylase Jumonji domain-containing protein (JMJD) gene is expressed in macrophages (24), and decreased activity of the JMJD protein results in increased histone H3 lysine 9 (H3K9) methylation and subsequent decreases in macrophage inflammatory chemokine production (25). Interestingly, nutrient status, particularly nutrient starvation, was shown to increase expression of JMJD (26).
Maternal Infection and Epigenetics
The prenatal environment influences the risk of inflammatory disease in adulthood. For example, maternal infection or inflammation can lead to preterm birth, which is itself linked to a number of chronic diseases. Many of these adverse long-term outcomes, including cardiovascular disease, asthma, and obesity, have an inflammatory component. The mechanisms remain unclear, but inflammation may alter the appropriate maintenance of epigenetic profiles during pregnancy. Neonatal T cells, for instance, produce very little IFN-γ due to a number of factors, including low-level expression of the transcription factor T-box gene that encodes transcription factor 2 (Tbet2). However, maintaining the fine control over this Th1 cytokine in utero probably also involves epigenetic factors, including hypermethylation of the Ifn-γ promoter (27). This normal shift away from potentially dangerous Th1 responses during the neonatal period could be altered by the maternal cytokine environment. Indeed, a recent study demonstrated that the maternal cytokine response determined the corresponding cytokine responses in infants, irrespective of the mother’s atopic status or environmental factors (28). These studies offer the intriguing possibility that cytokine imprinting may be linked to epigenetic modifications. Animal studies demonstrated this supposition experimentally. Increasing histone acetylation at Ifn-γ and decreasing histone acetylation at the Il-4 promoters, for instance, were shown to protect the progeny of pregnant mice from asthmalike symptoms (discussed previously; 20). Such epigenetic modifications have been implicated in early childhood diseases ranging from autism to allergy (29–31). Maternal exposures to farm environments can increase the number of T regulatory (Treg) cells in the cord blood of their infants, which is associated with decreased Th2 cytokines and may be linked to demethylation at the forkhead box P3 (FOXP3) promoter (32). The exposure of the mother to allergens in the environment may protect her infant from subsequent atopy and asthma.
Abnormal immune responses to common childhood infections, such as parvovirus B19, are linked to epigenetic alterations that may influence cancer (33). However, a clear link between maternal infections and subsequent epigenetic alterations in the fetus, and the mechanisms involved, remains lacking. Recent studies offer some clues. For example, recurrent urinary tract infections are common in pregnant women and can cause preterm birth and low birth weight (34). Uropathogenic Escherichia coli infection of bladder epithelial cells results in hypermethylation of the tumor suppressor gene cyclin-dependent kinase 2a (CDKN2A) (35), which is also methylated in human papilloma virus 16 (HPV-16)–induced epithelial dysplasia (36).
Whether maternal infection with a urinary tract pathogen directly induces epigenetic changes in the fetus is unclear. However, one could envision a scenario in which neonatal exposures to common pathogens leads to alterations in the epigenetic landscape of the fetus, increasing later susceptibility to chronic disease or cancer. Indeed, viruses and bacteria can act as epimutagens or “biological stressors” that induce epigenetic alterations (37, 38) either directly or indirectly through the induction of inflammation.
Another example of epigenetic alterations induced by a bacterial infection comes from the bacterium Listeria monocytogenes, a food-borne pathogen that is especially dangerous for pregnant women and the fetus. L. monocytogenes activates MAPK signaling pathways, leading to specific histone modifications and transcriptional activation of MAPK-induced inflammatory genes (39). L. monocytogenes also causes a decrease in the modification of histones on another set of immunity-related genes such as chemokine (C-X-C motif) ligand 2 (Cxcl2) (40). The ability of the bacterium to modulate immune gene expression through chromatin modification offers a glimpse into how early-life infection could influence immune response and modulation in later life. Indeed, a recent study of DNA methylation patterns in preterm birth caused by infection demonstrated a significant increase in methylation at the regulatory regions for pleomorphic adenoma gene 1 (PLAG1) (41). PLAG1 is a transcription factor that stands at the nexus of growth and development, and dysregulation of PLAG1 expression has been associated with cancer (42). Clearly, further studies are needed to uncover the complex interactions between epigenetic changes induced by maternal infection and the long-term consequences for both mother and child.
Nutritional and Bioactive Factors That Regulate Inflammation via Epigenetic Mechanisms
Progress has been made in identifying the cellular and molecular signaling mechanisms underlying anti-inflammatory effects of numerous nutrients. Macronutrients (e.g., n–3 PUFAs), micronutrients (e.g., vitamins A, D, C, and E; folic acid; selenium) and other bioactive compounds (e.g., flavonoids) have all been targets of investigation. However, only a few studies addressed the regulatory role that nutrients play to modulate inflammation via epigenetic pathways. Maternal overnutrition and obesity resulting in dyslipidemia and increased systemic inflammation were shown to have profound effects on the developing embryo and the fetus in utero (43). A maternal high-saturated-fat diet was shown to induce inflammation pathways in the offspring in animal (44–46) and in human (47, 48) studies. Among the micronutrients, folic acid is a well-known one-carbon donor for methylation and synthesis of DNA. These functions of folic acid are especially critical during early postnatal development because rapid cell growth and proliferation are taking place during this period. Higher inflammation during this developmental period poses a higher risk of disease development (49) due to immune system overactivation. Anti-inflammatory nutrients such as folic acid can be beneficial in preventing inflammatory responses (50, 51). Recent in vitro studies demonstrated that folic acid–deficient conditions increased the expression of inflammatory mediators such as Il-β, Il-6, Tnf-α, and monocyte chemoattractant protein-1 (Mcp-1) in the mouse monocyte cell line RAW264.7 (52). Increased methylation of the promoter regions of tumor suppressor genes is associated with carcinogenesis, whereas decreased methylation of tumor suppressor genes is associated with a reduction in tumor development. In vivo, increased consumption of folate and folate-rich foods is associated with decreased promoter methylation of genes associated with tumor suppression (53). Promoter CpG island methylation levels in colorectal mucosa were related to RBC folate concentrations in human studies (54). In addition, high intake of maternal folic acid reduced cancer risk in offspring in a rat model by altering histone deacetylation (55). Another micronutrient that alters epigenetic pathways is vitamin D. Deficiency or low concentrations of vitamin D are associated with increased inflammation, and the anti-inflammatory actions of vitamin D may be due to changes in DNA methylation and histone modifications. Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the brain and spinal cord, and increased severity of MS is associated with decreased vitamin D status (56). Vitamin D suppresses inflammatory responses in adult humans with MS by decreasing IL-17 gene expression by blocking of nuclear factor necessary for activating T cells (NFAT) transcription factors and by recruitment of histone deacetylase (HDAC) (57). IL-12, a proinflammatory cytokine, is involved in activation and differentiation of CD4+ T cells into Th1 cells. Vitamin D suppresses Il-12 promoter activation by modulating histone modification (58). Moreover, severe vitamin D deficiency is associated with methylation changes in peripheral blood leukocyte DNA in humans (59). Among the bioactive nutrients, curcumin and curcumin-derived synthetic analogs control inflammation responses and body weight by altering DNA methylation (60) and via micro-RNA (61). In addition, curcumin inhibits high-glucose-induced NF-κB activation, proinflammatory cytokine production, and histone acetylase activity in human monocytic THP-1 cells (62). Other plant polyphenolic bioactive compounds, such as epigallocatechin gallate, inhibit inflammation via modulation of histone acetyltransferase activity (63). Few studies have addressed how phytochemical nutrients suppress the transcription activation processes via epigenetic modification (64, 65). Inflammatory cytokine gene expression may also be modulated by histone deacetylase inhibitors (HDACi) through destabilization of the DNA methylation enzyme mRNA, which then leads to decreased inflammatory gene expression (66, 67). In line with this observation, HDACi suppressed IL-1β–, TNF-α–, and Toll-like receptor ligand–induced rheumatoid arthritis synovial macrophage production of IL-6 cytokines (66). Determination of the possible nutrient control of HDACi would have important implications, because HDACi-regulated reduction in cytokine production might suppress inflammatory cytokine production in rheumatoid arthritis. Currently, however, relatively little is known regarding the potential effects of anti-inflammatory nutrients on HDACi regulation of cytokine gene expression. Interestingly, ginger, a commonly used dietary herbal product, was shown to have broad anti-inflammatory properties (68). For example, a study by Shim et al. (69) showed that bioactive compounds found in ginger extracts, such as 6-shogaol, have anti-inflammatory effects on primary rat astrocytes production of inflammatory cytokines by HDAC inhibition when these cells were stimulated with LPS. Furthermore, another recent study showed that injection of Alzheimer disease mice with 6-shogaol had beneficial effects such as improved memory by increased glial cell activation and brain neuronal survival (70). Although reduction in neuronal inflammation results in beneficial effects, whether improved memory in animals with Alzheimer disease is directly mediated via modulation of HDACi remains to be determined. Other dietary bioactive compounds such as allyl sulfur compounds found in garlic were shown to inhibit inflammation (71) and cancer cell growth (mouse erythroleukemia) via modulating acetylation of histone (72). Plant flavonoids, including apigenin (73), which is found in high concentrations in parsley (74), and chrysin (75), which is found in medicinal herbs (76), were shown to inhibit HDAC. In addition, although a better known function is its role as metabolic FA fuel for gut bacteria, the dietary fiber component butyrate was shown to induce cell growth arrest, inhibit DNA synthesis, induce cell differentiation, and regulate gene expression by inhibiting HDAC (77, 78).
Maternal Nutrition Influence on Inflammation, Immune Response to Infection, and Epigenetics
Overnutrition or nutrient deficiency during pregnancy can have devastating effects on the health of offspring at early or late stages of their life. It is becoming clear that poor maternal nutrition has a negative impact on fetal gene expression through epigenetic programming (75). These changes likely involve covalent modifications of DNA and chromatin remodeling. Inadequate fetal and early-age poor nutrition is associated with an increased risk of metabolic syndrome later in life. The Dutch Winter Hunger Study, for example, showed that nutrient deficiency during mid- and late gestation was associated with hyperglycemia in middle age (75). Furthermore, growth in utero as well as in early childhood is greatly influenced by maternal macronutrient intake, suggesting the importance of a maternal diet that contains sufficient macronutrients for adequate fetal and child development. Indeed, low circulating concentrations of vitamin B-12 and high circulating concentrations of total homocysteine predicted intrauterine growth retardation (75). Micronutrients including zinc are also essential to the epigenome. Zinc is required for methylation, and its deficiency is suggested to contribute to atherosclerosis, inflammation, and diabetes. Several studies suggest that zinc deficiency at the stages of intrauterine and infant life contributes to the pathogenesis of inflammation and metabolic diseases in adulthood (75). Furthermore, maternal zinc deficiency was shown to impair immunity in the offspring of mice (75). Methylation can be a double-edged sword, however: in utero supplementation with cofactors and methyl donors required for methyl metabolism aggravated allergic airway diseases; the transcription factor runt-related transcription factor 3 (Runx3), a negative regulator of asthma-like disease in mice, was excessively methylated in this model system (79). The importance of adequate maternal nutrition for the progeny’s health should be highlighted, because the effects of maternal zinc deficiency in offspring persist during developmental stages despite intake of appropriate amounts of zinc. Thus, macro- and micronutrient deficiencies, either in intrauterine or during infant stages, can contribute to serious human health problems throughout life.
Another example of maternal nutrition deficiency that can affect the progeny’s health is the intake of a diet restricted in proteins during conception through pregnancy and lactation. Several reports showed that a maternal diet low in protein caused metabolic abnormalities, impaired immunity, increased sensitivity to oxidative stress, and glucose dyshomeostasis (75). Further studies from our laboratory showed that a low-protein prenatal and high-fat postnatal diet in Sprague-Dawley rats increased the number of small adipocytes in adipose tissue and decreased insulin sensitivity (75). Such data are suggestive of alterations in the DNA methylation within adipocytes and a subsequent increased risk of developing type 2 diabetes.
FAs are essential components of the human diet because adequate amounts of these fats are required for health and normal growth. In general, high intake of diets rich in n–3 FAs such as EPA, DHA, and α-linolenic acid (18:3n−3) confer protective effects. A recent study examined whether dietary n–3 PUFAs influence leptin and leptin receptor promoter DNA methylation. Results showed no effects of n–3 PUFAs on promoter DNA methylation in leptin and leptin receptor genes (75). Another study examined effects of fish oil supplementation on global DNA methylation. Although the exact underlying mechanisms involved in the modulation of insulin sensitivity by DNA methylation remain to be investigated, fish oil supplementation for 2 generations increased insulin-stimulated glucose uptake and insulin sensitivity, lowered blood lipid concentrations, and decreased global DNA methylation in the liver (75). Interestingly, the anticarcinogenic effects of n–3 PUFAs were tested in U937 leukemia cells in vitro. n–3 PUFA treatment induced the myeloid lineage-specific transcription factors CCAAT/enhancer-binding protein binding to the macrophage colony-stimulating factor (M-CSF) receptor promoter, resulting in enhanced M-CSF receptor gene expression via methylation alterations of the CpG islands in the M-CSF gene promoter region (75). These data suggest that maternal dietary FAs, especially n–3 PUFAs, modulate inflammatory functions in various immune cells by epigenetic mechanisms. High-fat diets alone (75) or when combined with high fructose intake by female rats during prenatal and throughout postnatal life were shown to increase the risk of renal and metabolic diseases later in life (75). Maternal obesity and/or high-fat diet intake predispose offspring to various vascular diseases involving hormonal dysregulation and inflammatory cytokine release. Phytochemical bioactive compounds called indoles are found in commonly consumed cruciferous vegetables such as broccoli, cabbage, and Brussels sprouts (80). It was shown that maternal indole supplementation reduces bisphenol A–induced prostatic tissue lesions and inflammation in a rat model study (81). Flavonoid quercetins are found in red onions, citrus fruits, and red wine (82). Although quercetins are known to have beneficial effects such as lipid (83) and blood pressure (84)–lowering effects, whether maternal diets supplemented with quercetins have anti-inflammatory effects is not clear. For example, although maternal quercetin intake reduces endoplasmic reticulum stress–associated stress and inflammation (85), maternal flavonoid supplementation also was shown to increase hepatic inflammatory cytokine expression in the liver (86) and increase the risk of carcinogenesis (87). Interestingly, intake of specific supplements such as green tea was shown to mitigate high-fat–induced metabolic abnormalities such as insulin resistance in rat offspring (75). The addition of green tea extract, which contains high amounts of flavonoid catechins compared with maternal high-fat diet alone, also resulted in increased serum adiponectin concentrations (88).
Summary and Conclusions
In summary, maternal nutrition has a considerable impact on fetal metabolic programming. Specifically, high-fat diets, micro- and macronutrient deficiencies, and low-protein diet intake during conception through lactation are all deleterious to fetal and infant development. The deleterious effects involve alterations in gene expression and epigenetic programming that converge to induce inflammation, impair the immune system, and cause a series of pathologic conditions at both early and late stages of life. Maternal nutrient intake, maternal infection, obesity, environmental pollution and toxicants, and psychological stress may all influence immune function and inflammation in utero (Figure 1). Of these, maternal nutrition may be a key factor. Both micro- and macronutrients modulate immune cell proinflammatory cytokine and chemokine secretory functions, immune cell differentiation, and immune cell proliferation by altering promoter region DNA methylation and histone modification. Because increased chronic inflammation causes metabolic complications and further contributes to the development of diabetes, cancer, and cardiovascular disease, increased anti-inflammatory nutrient and bioactive compounds may help reduce in utero programming of immune cells that control inflammatory response in the offspring. Future controlled human supplementation studies are needed to make nutrient supplementation recommendations. Both nutritional influences and infections likely induce global epigenetic changes, making it difficult to link specific changes in gene expression to any particular epigenetic modification. The rapidly expanding literature on maternal influences on fetal development and later disease states must therefore be interpreted carefully. However, new advances in next-generation sequencing and other technologies will enable researchers to unravel these complex networks and link specific gene expression changes to specific epigenetic modifications induced by infections and nutritional influences.
FIGURE 1.
Epigenetic regulators of immune and inflammatory functions in offspring. Maternal nutrient intake, infection, obesity, environmental pollution, and stress contribute to epigenetic changes in the fetus by DNA methylation and histone modifications. These epigenetic changes, in turn, alter immune function and inflammatory responses by activating inflammatory cytokines and chemokines, resulting in increased risk of acute and chronic diseases in offspring.
Acknowledgments
KJC, CAB, and OG wrote the manuscript; and KJC had primary responsibility for final content. All authors read and approved the final manuscript.
Footnotes
Abbreviations used: CDKN2A, cyclin-dependent kinase 2a; CD4, cluster of differentiation 4; Cxcl2, chemokine C-X-C motif ligand 2; FOXP3, forkhead box P3; H3K9, histone H3 lysine 9; H4, histone 4; HDAC, histone deacetylase; HDACi, histone deacetylase inhibitor; HPV-16, human papilloma virus 16; IFN-γ, interferon γ; Igf2, insulin-like growth factor 2; JMJD, Jumonji domain-containing protein; Mcp-1, monocyte chemoattractant protein-1; M-CSF, macrophage colony-stimulating factor; MS, multiple sclerosis; M1, macrophage type 1; M2, macrophage type 2; NFAT, nuclear factor necessary for activating T cells; PLAG1, pleomorphic adenoma gene 1; Runx3, runt-related transcription factor 3; SWI/SNF, SWItch/sucrose nonfermentable; Tbet2, T-box gene that encodes transcription factor 2; Th, T helper; Tnf, tumor necrosis factor.
References
- 1.Choi SW, Friso S. Epigenetics: a new bridge between nutrition and health. Adv Nutr 2010;1:8–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ibáñez de Cáceres I, Cairns P. Methylated DNA sequences for early cancer detection, molecular classification and chemotherapy response prediction. Clin Transl Oncol 2007;9:429–37. [DOI] [PubMed] [Google Scholar]
- 3.Reik W, Walter J. Genomic imprinting: parental influence on the genome. Nat Rev Genet 2001;2:21–32. [DOI] [PubMed] [Google Scholar]
- 4.Reik W, Collick A, Norris ML, Barton SC, Surani MA. Genomic imprinting determines methylation of parental alleles in transgenic mice. Nature 1987;328:248–51. [DOI] [PubMed] [Google Scholar]
- 5.Swain JL, Stewart TA, Leder P. Parental legacy determines methylation and expression of an autosomal transgene: a molecular mechanism for parental imprinting. Cell 1987;50:719–27. [DOI] [PubMed] [Google Scholar]
- 6.Schlesinger Y, Straussman R, Keshet I, Farkash S, Hecht M, Zimmerman J, Eden E, Yakhini Z, Ben-Shushan E, Reubinoff BE, et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet 2007;39:232–6. [DOI] [PubMed] [Google Scholar]
- 7.Feil R, Walter J, Allen ND, Reik W. Developmental control of allelic methylation in the imprinted mouse Igf2 and H19 genes. Development 1994;120:2933–43. [DOI] [PubMed] [Google Scholar]
- 8.Kovacheva VP, Mellott TJ, Davison JM, Wagner N, Lopez-Coviella I, Schnitzler AC, Blusztajn JK. Gestational choline deficiency causes global and Igf2 gene DNA hypermethylation by up-regulation of Dnmt1 expression. J Biol Chem 2007;282:31777–88. [DOI] [PubMed] [Google Scholar]
- 9.Napoli I, Blusztajn JK, Mellott TJ. 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;1237:124–35. [DOI] [PubMed] [Google Scholar]
- 10.Rehfeldt C, Lefaucheur L, Block J, Stabenow B, Pfuhl R, Otten W, Metges CC, Kalbe C. Limited and excess protein intake of pregnant gilts differently affects body composition and cellularity of skeletal muscle and subcutaneous adipose tissue of newborn and weanling piglets. Eur J Nutr 2012;51:151–65. [DOI] [PubMed] [Google Scholar]
- 11.Migliori V, Phalke S, Bezzi M, Guccione E. Arginine/lysine-methyl/methyl switches: biochemical role of histone arginine methylation in transcriptional regulation. Epigenomics 2010;2:119–37. [DOI] [PubMed] [Google Scholar]
- 12.Mahajan K, Fang B, Koomen JM, Mahajan NP. H2B Tyr37 phosphorylation suppresses expression of replication-dependent core histone genes. Nat Struct Mol Biol 2012;19:930–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bierne H, Hamon M, Cossart P. Epigenetics and bacterial infections. Cold Spring Harb Perspect Med 2012;2:a010272. [DOI] [PMC free article] [PubMed]
- 14.Jeong SM, Lee C, Lee SK, Kim J, Seong RH. The SWI/SNF chromatin-remodeling complex modulates peripheral T cell activation and proliferation by controlling AP-1 expression. J Biol Chem 2010;285:2340–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wilson BG, Roberts CW. SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer 2011;11:481–92. [DOI] [PubMed] [Google Scholar]
- 16.Janson PC, Winerdal ME, Winqvist O. At the crossroads of T helper lineage commitment—epigenetics points the way. Biochim Biophys Acta 2009;1790:906–19. [DOI] [PubMed] [Google Scholar]
- 17.Kanno Y, Vahedi G, Hirahara K, Singleton K, O'Shea JJ. Transcriptional and epigenetic control of T helper cell specification: molecular mechanisms underlying commitment and plasticity. Annu Rev Immunol 2012;30:707–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hamalainen-Laanaya HK, Kobie JJ, Chang C, Zeng WP. Temporal and spatial changes of histone 3 K4 dimethylation at the IFN-gamma gene during Th1 and Th2 cell differentiation. J Immunol 2007;179:6410–5. [DOI] [PubMed] [Google Scholar]
- 19.Morinobu A, Kanno Y, O'Shea JJ. Discrete roles for histone acetylation in human T helper 1 cell-specific gene expression. J Biol Chem 2004;279:40640–6. [DOI] [PubMed] [Google Scholar]
- 20.Brand S, Teich R, Dicke T, Harb H, Yildirim AO, Tost J, Schneider-Stock R, Waterland RA, Bauer UM, von Mutius E, et al. Epigenetic regulation in murine offspring as a novel mechanism for transmaternal asthma protection induced by microbes. J Allergy Clin Immunol 2011;128:618–25, e611–7. [DOI] [PubMed]
- 21. Brand S, Kesper DA, Teich R, Kilic-Niebergall E, Pinkenburg O, Bothur E, Lohoff M, Garn H, Pfefferle PI, Renz H. DNA methylation of TH1/TH2 cytokine genes affects sensitization and progress of experimental asthma. J Allergy Clin Immunol 2012;129:602–10, e1606. [DOI] [PubMed]
- 22.Bruniquel D, Schwartz RH. Selective, stable demethylation of the interleukin-2 gene enhances transcription by an active process. Nat Immunol 2003;4:235–40. [DOI] [PubMed] [Google Scholar]
- 23.Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, Miyake T, Matsushita K, Okazaki T, Saitoh T, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol 2010;11:936–44. [DOI] [PubMed] [Google Scholar]
- 24.Osawa T, Tsuchida R, Muramatsu M, Shimamura T, Wang F, Suehiro J, Kanki Y, Wada Y, Yuasa Y, Aburatani H, et al. Inhibition of histone demethylase JMJD1A improves anti-angiogenic therapy and reduces tumor-associated macrophages. Cancer Res 2013;73:3019–28. [DOI] [PubMed] [Google Scholar]
- 25.Tausendschön M, Dehne N, Brune B. Hypoxia causes epigenetic gene regulation in macrophages by attenuating Jumonji histone demethylase activity. Cytokine 2011;53:256–62. [DOI] [PubMed] [Google Scholar]
- 26.Osawa T, Muramatsu M, Wang F, Tsuchida R, Kodama T, Minami T, Shibuya M. Increased expression of histone demethylase JHDM1D under nutrient starvation suppresses tumor growth via down-regulating angiogenesis. Proc Natl Acad Sci USA 2011;108:20725–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.White GP, Watt PM, Holt BJ, Holt PG. Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol 2002;168:2820–7. [DOI] [PubMed] [Google Scholar]
- 28.Djuardi Y, Wibowo H, Supali T, Ariawan I, Bredius RG, Yazdanbakhsh M, Rodrigues LC, Sartono E. Determinants of the relationship between cytokine production in pregnant women and their infants. PLoS ONE 2009;4:e7711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lillycrop KA, Burdge GC. Epigenetic mechanisms linking early nutrition to long term health. Best Pract Res Clin Endocrinol Metab 2012;26:667–76. [DOI] [PubMed] [Google Scholar]
- 30.Miyake K, Hirasawa T, Koide T, Kubota T. Epigenetics in autism and other neurodevelopmental diseases. Adv Exp Med Biol 2012;724:91–8. [DOI] [PubMed] [Google Scholar]
- 31.Salam MT, Zhang Y, Begum K. Epigenetics and childhood asthma: current evidence and future research directions. Epigenomics 2012;4:415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Schaub B, Liu J, Hoppler S, Schleich I, Huehn J, Olek S, Wieczorek G, Illi S, von Mutius E. Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J Allergy Clin Immunol 2009;123:774–82, e775. [DOI] [PubMed]
- 33.Vasconcelos GM, Christensen BC, Houseman EA, Xiao J, Marsit CJ, Wiencke JK, Zheng S, Karagas MR, Nelson HH, Wrensch MR, et al. History of parvovirus B19 infection is associated with a DNA methylation signature in childhood acute lymphoblastic leukemia. Epigenetics 2011;6:1436–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schneeberger C, Geerlings SE, Middleton P, Crowther CA. Interventions for preventing recurrent urinary tract infection during pregnancy. Cochrane Database Syst Rev 2012;11:CD009279. [DOI] [PubMed] [Google Scholar]
- 35.Tolg C, Bagli DJ. Uropathogenic Escherichia coli infection: potential importance of epigenetics. Epigenomics 2012;4:229–35. [DOI] [PubMed] [Google Scholar]
- 36.Fonseca-Silva T, Farias LC, Cardoso CM, Souza LR, Carvalho Fraga CA, Oliveira MV, Barros LO, Alves LR, De-Paula AM, Marques-Silva L, et al. Analysis of p16(CDKN2A) methylation and HPV-16 infection in oral mucosal dysplasia. Pathobiology 2012;79:94–100. [DOI] [PubMed] [Google Scholar]
- 37.Barros SP, Offenbacher S. Modifiable risk factors in periodontal disease: epigenetic regulation of gene expression in the inflammatory response. Periodontol 2000 2014;64:95–110. [DOI] [PubMed] [Google Scholar]
- 38.Paschos K, Allday MJ. Epigenetic reprogramming of host genes in viral and microbial pathogenesis. Trends Microbiol 2010;18:439–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Schmeck B, Beermann W, van Laak V, Zahlten J, Opitz B, Witzenrath M, Hocke AC, Chakraborty T, Kracht M, Rosseau S, et al. Intracellular bacteria differentially regulated endothelial cytokine release by MAPK-dependent histone modification. J Immunol 2005;175:2843–50. [DOI] [PubMed] [Google Scholar]
- 40.Hamon MA, Batsche E, Regnault B, Tham TN, Seveau S, Muchardt C, Cossart P. Histone modifications induced by a family of bacterial toxins. Proc Natl Acad Sci USA 2007;104:13467–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Liu Y, Hoyo C, Murphy S, Huang Z, Overcash F, Thompson J, Brown H, Murtha AP. DNA methylation at imprint regulatory regions in preterm birth and infection. Am J Obstet Gynecol 2013;208:395e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Engel JR, Smallwood A, Harper A, Higgins MJ, Oshimura M, Reik W, Schofield PN, Maher ER. Epigenotype-phenotype correlations in Beckwith-Wiedemann syndrome. J Med Genet 2000;37:921–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Heerwagen MJ, Miller MR, Barbour LA, Friedman JE. Maternal obesity and fetal metabolic programming: a fertile epigenetic soil. Am J Physiol Regul Integr Comp Physiol 2010;299:R711–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sasaki A, de Vega W, Sivanathan S, St-Cyr S, McGowan PO. Maternal high-fat diet alters anxiety behavior and glucocorticoid signaling in adolescent offspring. Neuroscience 2014;272:92–101. [DOI] [PubMed] [Google Scholar]
- 45.Xue Y, Wang H, Du M, Zhu MJ. Maternal obesity induces gut inflammation and impairs gut epithelial barrier function in nonobese diabetic mice. J Nutr Biochem 2014;25:758–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yan X, Huang Y, Wang H, Du M, Hess BW, Ford SP, Nathanielsz PW, Zhu MJ. Maternal obesity induces sustained inflammation in both fetal and offspring large intestine of sheep. Inflamm Bowel Dis 2011;17:1513–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Catalano PM, Presley L, Minium J, Hauguel-de Mouzon S. Fetuses of obese mothers develop insulin resistance in utero. Diabetes Care 2009;32:1076–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Leibowitz KL, Moore RH, Ahima RS, Stunkard AJ, Stallings VA, Berkowitz RI, Chittams JL, Faith MS, Stettler N. Maternal obesity associated with inflammation in their children. World J Pediatr 2012;8:76–9. [DOI] [PubMed] [Google Scholar]
- 49.Meyer U, Feldon J. Neural basis of psychosis-related behaviour in the infection model of schizophrenia. Behav Brain Res 2009;204:322–34. [DOI] [PubMed] [Google Scholar]
- 50.Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, Sakaguchi S, Takeda K. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol 2012;189:2869–78. [DOI] [PubMed] [Google Scholar]
- 51.Thomas TP, Goonewardena SN, Majoros IJ, Kotlyar A, Cao Z, Leroueil PR, Baker JR Jr. Folate-targeted nanoparticles show efficacy in the treatment of inflammatory arthritis. Arthritis Rheum 2011;63:2671–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kolb AF, Petrie L. Folate deficiency enhances the inflammatory response of macrophages. Mol Immunol 2013;54:164–72. [DOI] [PubMed] [Google Scholar]
- 53.Jacobs DI, Hansen J, Fu A, Stevens RG, Tjonneland A, Vogel UB, Zheng T, Zhu Y. Methylation alterations at imprinted genes detected among long-term shiftworkers. Environ Mol Mutagen 2013;54:141–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wallace K, Grau MV, Levine AJ, Shen L, Hamdan R, Chen X, Gui J, Haile RW, Barry EL, Ahnen D, et al. Association between folate levels and CpG Island hypermethylation in normal colorectal mucosa. Cancer Prev Res (Phila) 2010;3:1552–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cho K, Mabasa L, Bae S, Walters MW, Park CS. Maternal high-methyl diet suppresses mammary carcinogenesis in female rat offspring. Carcinogenesis 2012;33:1106–12. [DOI] [PubMed] [Google Scholar]
- 56.Smolders J, Menheere P, Kessels A, Damoiseaux J, Hupperts R. Association of vitamin D metabolite levels with relapse rate and disability in multiple sclerosis. Mult Scler 2008;14:1220–4. [DOI] [PubMed] [Google Scholar]
- 57.Joshi S, Pantalena LC, Liu XK, Gaffen SL, Liu H, Rohowsky-Kochan C, Ichiyama K, Yoshimura A, Steinman L, Christakos S, et al. 1,25-Dihydroxyvitamin D(3) ameliorates Th17 autoimmunity via transcriptional modulation of interleukin-17A. Mol Cell Biol 2011;31:3653–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gynther P, Toropainen S, Matilainen JM, Seuter S, Carlberg C, Vaisanen S. Mechanism of 1alpha,25-dihydroxyvitamin D(3)-dependent repression of interleukin-12B. Biochim Biophys Acta 2011;1813:810–8. [DOI] [PubMed] [Google Scholar]
- 59.Wang X, Falkner B, Zhu H, Shi H, Su S, Xu X, Sharma AK, Dong Y, Treiber F, Gutin B, et al. A genome-wide methylation study on essential hypertension in young African American males. PLoS One 2013;8:e53938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vanden Berghe W. Epigenetic impact of dietary polyphenols in cancer chemoprevention: lifelong remodeling of our epigenomes. Pharmacol Res 2012;65:565–76. [DOI] [PubMed] [Google Scholar]
- 61.Bao B, Ahmad A, Kong D, Ali S, Azmi AS, Li Y, Banerjee S, Padhye S, Sarkar FH. Hypoxia induced aggressiveness of prostate cancer cells is linked with deregulated expression of VEGF, IL-6 and miRNAs that are attenuated by CDF. PLoS ONE 2012;7:e43726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Yun JM, Jialal I, Devaraj S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem 2011;22:450–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Choi KC, Jung MG, Lee YH, Yoon JC, Kwon SH, Kang HB, Kim MJ, Cha JH, Kim YJ, Jun WJ, et al. Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation. Cancer Res 2009;69:583–92. [DOI] [PubMed] [Google Scholar]
- 64.Vanden Berghe W, Haegeman G. Epigenetic remedies by dietary phytochemicals against inflammatory skin disorders: myth or reality? Curr Drug Metab 2010;11:436–50. [DOI] [PubMed] [Google Scholar]
- 65.Rajendran P, Ho E, Williams DE, Dashwood RH. Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin Epigenetics 2011;3:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Grabiec AM, Korchynskyi O, Tak PP, Reedquist KA. Histone deacetylase inhibitors suppress rheumatoid arthritis fibroblast-like synoviocyte and macrophage IL-6 production by accelerating mRNA decay. Ann Rheum Dis 2012;71:424–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Matsushita K, Takeuchi O, Standley DM, Kumagai Y, Kawagoe T, Miyake T, Satoh T, Kato H, Tsujimura T, Nakamura H, et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature 2009;458:1185–90. [DOI] [PubMed] [Google Scholar]
- 68.Ali BH, Blunden G, Tanira MO, Nemmar A. Some phytochemical, pharmacological and toxicological properties of ginger (Zingiber officinale Roscoe): a review of recent research. Food Chem Toxicol 2008;46:409–20. [DOI] [PubMed]
- 69. Shim S, Kim S, Choi DS, Kwon YB, Kwon J. Anti-inflammatory effects of [6]-shogaol: potential roles of HDAC inhibition and HSP70 induction. Food Chem Toxicol 2011;49:2734–40. [DOI] [PubMed]
- 70.Moon M, Kim HG, Choi JG, Oh H, Lee PK, Ha SK, Kim SY, Park Y, Huh Y, Oh MS. 6-Shogaol, an active constituent of ginger, attenuates neuroinflammation and cognitive deficits in animal models of dementia. Biochem Biophys Res Commun 2014;449:8–13. [DOI] [PubMed] [Google Scholar]
- 71.Hui C, Like W, Yan F, Tian X, Qiuyan W, Lifeng H. S-allyl-L-cysteine sulfoxide inhibits tumor necrosis factor-alpha induced monocyte adhesion and intercellular cell adhesion molecule-1 expression in human umbilical vein endothelial cells. Anat Rec (Hoboken) 2010;293:421–30. [DOI] [PubMed] [Google Scholar]
- 72.Lea MA, Rasheed M, Randolph VM, Khan F, Shareef A, desBordes C. Induction of histone acetylation and inhibition of growth of mouse erythroleukemia cells by S-allylmercaptocysteine. Nutr Cancer 2002;43:90–102. [DOI] [PubMed] [Google Scholar]
- 73.Pandey M, Kaur P, Shukla S, Abbas A, Fu P, Gupta S. Plant flavone apigenin inhibits HDAC and remodels chromatin to induce growth arrest and apoptosis in human prostate cancer cells: in vitro and in vivo study. Mol Carcinog 2012;51:952–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lefort ÉC, Blay J. Apigenin and its impact on gastrointestinal cancers. Mol Nutr Food Res 2013;57:126–44. [DOI] [PubMed] [Google Scholar]
- 75.Pal-Bhadra M, Ramaiah MJ, Reddy TL, Krishnan A, Pushpavalli SN, Babu KS, Tiwari AK, Rao JM, Yadav JS, Bhadra U. Plant HDAC inhibitor chrysin arrest cell growth and induce p21WAF1 by altering chromatin of STAT response element in A375 cells. BMC Cancer 2012;12:180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pushpavalli G, Veeramani C, Pugalendi KV. Influence of chrysin on hepatic marker enzymes and lipid profile against D-galactosamine-induced hepatotoxicity rats. Food Chem Toxicol 2010;48:1654–9. [DOI] [PubMed]
- 77.Sekhavat A, Sun JM, Davie JR. Competitive inhibition of histone deacetylase activity by trichostatin A and butyrate. Biochem Cell Biol 2007;85:751–8. [DOI] [PubMed]
- 78.Steliou K, Boosalis MS, Perrine SP, Sangerman J, Faller DV. Butyrate histone deacetylase inhibitors. Biores Open Access 2012;1:192–8. [DOI] [PMC free article] [PubMed]
- 79.Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, Bailey N, Potts EN, Whitehead G, Brass DM, et al. In utero supplementation with methyl donors enhances allergic airway disease in mice. J Clin Invest 2008;118:3462–9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 80.Shorey LE, Madeen EP, Atwell LL, Ho E, Lohr CV, Pereira CB, Dashwood RH, Williams DE. Differential modulation of dibenzo[def,p]chrysene transplacental carcinogenesis: maternal diets rich in indole-3-carbinol versus sulforaphane. Toxicol Appl Pharmacol 2013;270:60–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Brandt JZ, Silveira LT, Grassi TF, Anselmo-Franci JA, Favaro WJ, Felisbino SL, Barbisan LF, Scarano WR. Indole-3-carbinol attenuates the deleterious gestational effects of bisphenol A exposure on the prostate gland of male F1 rats. Reprod Toxicol 2014;43:56–66. [DOI] [PubMed] [Google Scholar]
- 82.Panchal SK, Poudyal H, Brown L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J Nutr 2012;142:1026–32. [DOI] [PubMed] [Google Scholar]
- 83. Juzwiak S, Wojcicki J, Mokrzycki K, Marchlewicz M, Bialecka M, Wenda-Rozewicka L, Gawronska-Szklarz B, Drozdzik M. Effect of quercetin on experimental hyperlipidemia and atherosclerosis in rabbits. Pharmacol Rep 2005;57:604–9. [PubMed]
- 84.Yamamoto Y, Oue E. Antihypertensive effect of quercetin in rats fed with a high-fat high-sucrose diet. Biosci Biotechnol Biochem 2006;70:933–9. [DOI] [PubMed] [Google Scholar]
- 85.Wu Z, Zhao J, Xu H, Lyv Y, Feng X, Fang Y, Xu Y. Maternal quercetin administration during gestation and lactation decrease endoplasmic reticulum stress and related inflammation in the adult offspring of obese female rats. Eur J Nutr 2014;53:1669–83. [DOI] [PubMed] [Google Scholar]
- 86.Vanhees K, Godschalk RW, Sanders A, van Waalwijk van Doorn-Khosrovani SB, van Schooten FJ. Maternal quercetin intake during pregnancy results in an adapted iron homeostasis at adulthood. Toxicology 2011;290:350–8. [DOI] [PubMed] [Google Scholar]
- 87. Vanhees K, de Bock L, Godschalk RW, van Schooten FJ, van Waalwijk van Doorn-Khosrovani SB. Prenatal exposure to flavonoids: implication for cancer risk. Toxicol Sci 2011;120:59–67. [DOI] [PubMed]
- 88.Li S, Tse IM, Li ET. Maternal green tea extract supplementation to rats fed a high-fat diet ameliorates insulin resistance in adult male offspring. J Nutr Biochem 2012;23:1655–60. [DOI] [PubMed] [Google Scholar]

