Abstract
Biological redox signaling plays an important role in many diseases. Redox signaling involves reductive and oxidative mechanisms. Oxidative stress occurs when reductive mechanism overwhelms oxidative challenges. Cellular oxidative stress occurs when reactive oxygen/nitrogen species (RO/NS) exceed the cellular reductive/antioxidant capacity. Endogenously produced RO/NS from mitochondrial metabolic citric-acid-cycle coupled with electron-transport-chain or exogenous stimuli trigger cellular signaling events leading to homeostatic response or pathological damage. Recent evidence suggests that RO/NS also modulate epigenetic machinery driving gene expression. RO/NS affect DNA methylation/ demethylation, histone acetylation/deacetylation or histone methylation/demethylation. Many health beneficial phytochemicals possess redox capability that counteract RO/NS either by directly scavenging the radicals or via inductive mechanism of cellular defense antioxidant/reductive enzymes. Amazingly, these phytochemicals also possess epigenetic modifying ability. This review summarizes the latest advances on the interactions between redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals and the future challenges of integrating these events in human health.
Keywords: Redox, epigenetics, oxidative stress, metabolism, citric acid cycle, redox active phytochemicals
1. Background
Cellular reduction/oxidation (redox) state regulates diverse cellular functions ranging from homeostasis, differentiation, proliferation to apoptosis. The term and concept of “redox” for electron activity in redox reaction was first introduced by WM Clark (1922) as highlighted in DC Thorstenson’s “The concept of electron activity and its relation to redox potentials in aqueous geochemical systems” in 1984 (1). Redox is a type of basic chemical reaction in which the oxidation states of atoms are altered. Redox reactions are characterized by the transfer of electrons between chemical species, most often with one specie undergoing oxidation (loss of electrons) while another specie undergoes reduction (gain of electrons).
Following the discovery of redox for basic chemical reaction, in biological systems, oxidative stress was first introduced in 1985 by H. Sies (2). Oxidative stress occurs when the reductive mechanism is overwhelmed by the oxidative challenge. Cellular redox oxidative stress occurs when reactive oxygen/nitrogen species (RO/NS) including superoxide (O− ₂ •), hydroxyl ((OH•), hydrogen peroxide (H2O2), phenoxyl, nitric oxide (NO), nitrogen dioxide (NO−), peroxynitrite (OONO−), dinitrogen trioxide (N2O3), and nitrous acid (HNO2) radicals and others, exceed the cellular reductive/antioxidant capacity like glutathione (GSH), catalase, superoxide dismutase (SOD), glutathione peroxidase (GPX), thioredoxin, peroxiredoxin, among others (we will use RO/NS from here on to represent either ROS or RNS, unless stated otherwise). Importantly RO/NS are also secondary messengers triggering cellular signaling pathways in maintaining intracellular redox state of balance leading to normal physiologic homeostatic responses (reviewed in (3)).
Endogenous RO/NS are produced through everyday respiration in the mitochondrial citric acid cycle (CAC), also known as the TCA cycle (tricarboxylic acid cycle) or the Krebs cycle, which was first identified/discovered by Fisher and Krebs back in 1955 (4) coupled to the electron transport chain (ETC) (5) (note that we will use CAC from here on). The CAC is a series of chemical reactions used by all aerobic organisms for cellular energy adenosine triphosphate (ATP) production through the oxidation of acetyl-coenzyme A (CoA) derived from glucose, fatty acids and amino acids. The CAC cycle produces a huge flux of free radicals by being coupled to the ETC (6) with vast majority of the free radicals such as superoxide (O− ₂ •), and hydrogen peroxide (H2O2) being neutralized by cellular antioxidant enzymes like SOD, GPX and catalase with potentially 0.1% leakage (5). In addition to mitochondria, endogenous production of RO/NS occurs in other cellular compartments including peroxisomal β-oxidation of fatty acids, respiratory burst by phagocytic cells and cytochrome P450 metabolic reactions (3, 5). Exogenous sources of RO/NS include UV light, tobacco smoke, and other environmental factors including air pollution, contaminated drinking water and certain “unhealthy high-fat” dietary food, among others.
RO/NS in general can trigger cellular signal transduction events to maintain cellular redox homeostasis. In prokaryotic bacteria system, OxyR redox sensing protein which is typically in a reducing environment such as E. coli, is highly responsive to perturbation of RO/NS. When the cells are challenged by H2O2, for instance, OxyR undergoes transcriptional activation by thiol-disulfide bond formation of cysteines (Cys) 199 and 208, resulting in induction of glutaredoxin (GrxA/Grx1) and glutathione reductase (GR) to counteract H2O2 (7). The biological specificity and activity of H2O2 may result from the diverse thiol sensing molecules/proteins in the different cellular compartments, with different reactivity of the respective sulfhydryl groups towards H2O2, activated by varying concentrations and times of exposure (8). This reactivity/sensitivity or dose response such as H2O2, may range from the most sensitive/responsive with catalase, perR, peroxiredoxin 2/5, GAPDH, CDC2B, Keap1, PTP25B, thioredoxin to GSH depending on the RO/NS, triggering respective signaling responsive pathways accordingly, in a concentration-dependent fashion (8). In mammalian cells, H2O2 activates signaling molecules including AP-1, CREB, HSF1, HIF-1, NF-κB, NOTCH, NRF2-KEAP1, SP1, TP53 and SCREB-1, typically through thiol-sulfhydryl modification (8, 9), in many in vitro or cell culture settings. For instance, many redox active electrophilic compounds activate NRF2-KEAP1 anti-oxidative stress pathway via thiol-sulfhydryl modification leading to induction of cellular defense and antioxidant enzymes GST, HO-1 and NQO1 (10). Redox regulation of signaling protein kinases by phosphorylation/dephosphorylation, could be governed by the redox sensitivity of protein tyrosine phosphatases (PTPs). Oxidation by ROS via thiol-disulfide bonding would inactivate PTPs resulting in increased steady-state protein phosphorylation. Examples include CDC25 phosphatases, PTEN (phosphatase and tensin homolog) and PTP1B (protein tyrosine phosphatase 1B) (11). In this context, redox active electrophilic compounds could modulate I-κB kinase (IKK) /I-κB/NF-κB pathway via protein phosphatase 2A (PP2A) that modulates IKK (12–14).
2. Role of redox in the modulation of epigenetic machinery
Cellular metabolism plays an important role in chromatin remodeling and epigenetic reprogramming (reviewed in (15–17)). Several epigenetic marks, including DNA methylation, histone acetylation, histone methylation, long non-coding microRNA and microRNA, and ADP-ribosylation are linked to central metabolism via redox intermediates.
DNA Methylation/demethylation:
DNA methylation, catalyzed by DNA methyltransferases (DNMT1, DNMT3A, DNMT3B), is a highly dynamic process and it profoundly impacts gene transcription, development, differentiation, and cellular responsiveness to environmental cues. Among all the epigenetic mechanisms, DNA CpG methylation is probably the most studied (reviewed in (18)). DNA methylation entails the addition of methyl groups onto C5 position of cytosine (C) residue usually occur in CpG dinucleotide sequences, dispersed across the genome, leading to the formation of 5-methylcytosine (5-mC) (19). In the context of redox signaling, RO/NS can epigenetically affect DNA methylation through DNMTs activity or gene expression. After H2O2 treatment, oxidative DNA damage induces the formation of a large complex(es) containing DNMTs and members of the Polycomb Repressive Complex 4, resulting in the alteration of the methylation status of CpG regions (20). In a redox oxidizing chromatin microenvironment and in the absence of S-adenosyl methionine (SAM), DNMT3A and DNMT3B may catalyze the direct conversion of 5-mC or 5-hydroxymethylcytosine (5-hmC) to an unmethylated cysteine (21). ROS-induced DNA damage of O6-methylguanine may inhibit binding of DNMT leading to hypomethylation and inhibit methylation of adjacent cytosine nucleotides (22). Incorporation of 8-hydroxy-deoxyglunine (8-OHdG) and the oxidation by-product of 5-mC in the Methyl-CpG Binding Protein (MBP) recognition sequence may result in the significant inhibition of the binding affinity of MBP (23). Since DNA methylation is a highly active process, and it can be oxidized to 5-hmC by DNA demethylases ten-eleven translocation enzymes (TETs) (24). In general 5-mC suppresses gene transcription, however, 5-hmC appears to activate gene transcription (25). ROS may regulate DNA methylation via expression and/or activity of TETs. Knockdown of TET1 significantly increase H2O2-induced apoptosis of cerebellar granule cells, and in Tet1 knockout mice, the cerebellar granule cells are more sensitive to oxidative stress (26).
Histone deacetylation:
RO/NS affects histone acetylation. To date, eighteen histone deacetylases (HDACs) have been identified in humans and classified into zinc/iron-dependent deacetylases class I (HDACs 1, 2, 3, and 8), class II (HDACs 4, 5, 7, and 9), class IIB (HDACs 6 and 10), and class IV (HDAC11) (27) while class III HDACs (sirtuins 1–7) have an absolute requirement for NAD+ and are not zinc/iron-dependent for their activity (28). Several HDACs have been shown to be redox regulated. Redox-regulating protein thioredoxin 1 (TRX1) facilitates the reduction of oxidized signaling molecules and transcription factors by Cys’s thiol-disulfide exchange (29) including the nucleocytoplasmic shuttling of class II HDACs through a redox-dependent mechanism (30). TRX1 induces DnaJb5, a heat shock protein 40, and forms a multiple-protein complex with DnaJb5 and class II HDACs (30). In response to ROS/ H2O2-generating stimulus, both Cys-274/Cys-276 in DnaJb5 and Cys-667/Cys-669 in HDAC4 could be oxidized and form intramolecular disulfide bonds and both proteins could be reduced by TRX1 (30). Reduction of Cys-274/Cys-276 in DnaJb5 is essential for interaction between DnaJb5 and HDAC4, whereas reduction of Cys-667/Cys-669 in HDAC4 inhibits its nuclear export (30). Interestingly, several other Cys’s sulfhydryl (-SH) groups in HDACs appear to be sensitive to H2O2 in cardiomyocytes (30). Antioxidant protein peroxiredoxins (PRDX) serve not only as cellular defense against oxidative stress but also may act as redox relay for specific H2O2 signaling in cells (31). PRDX I and II are specific targets of HDAC6 and deacetylation of PRDXs by HDAC6 would decrease H2O2 reductive activity (32). Class I HDACs −1, −2 and −3 were postulated to be redox-sensitive with a putative redox-switch using chemical probes and covalent modification of HDAC1 at two conserved Cys residues, Cys-261 and Cys-273, coincided with attenuation of HDAC activity, changes in histone H3 and H4 acetylation patterns, de-repression of a LEF1/β-catenin model system, and transcription of HDAC-repressed genes, such as HO-1, Gadd45, and HSP70 (33). The catalytic activity of HDAC5 was found to suppress mitochondrial ROS generation and subsequent induction of NRF2-dependent antioxidant gene expression in cardiomyocytes (34). Cys-102 and Cys-153 of HDAC8, have been identified to be an integral components of the redox-switch in HDAC8, and disulfide bond formation under oxidizing conditions could be associated with a transition into a more stable protein resulting in a complete but reversible loss of enzyme activity (35). Collectively, there is strong evidence that redox RO/NS are capable of modulating chromatin accessibility by affecting HDAC/histone acetylation state in particular via multiple modifications of HDAC activity and expression. More studies would be needed to elucidate HDACs response to redox signaling.
Histone acetylation:
Histone acetylation is carried out by a group of histone acetyltransferases (HATs) including P300/CBP (CREB-binding protein) (36), enzymes that acetylate conserved lysine (K) residues on histone tails by transferring an acetyl group from acetyl-CoA to form ε-N-acetyl-lysine (37), resulting in decrease binding of histones to DNA, opening up the chromatin, and enhancing gene transcription. In E. coli, increasing the ratio of NADH to NAD+, inhibits pyruvate dehydrogenase (PDH) activity, and blocks formation of acetyl-CoA (38), since conversion of pyruvate to acetyl-CoA is catalyzed by PDH (39). In an in vitro study, increased ratio of NADH/NAD+ appears to be associated with the inhibition of PDH activity in pea (40). In maize seedlings, heat stress promotes accumulation of O2−, and induces histone hyperacetylation due to the elevated expression of genes GCN5 (General Control Nondepressible 5) and HAT-B (41). In mammalian cells, H2O2 increases the activity of HATs and increases acetylation of H3/H4 histones (42, 43).
Histone methylation/demethylation:
Histone methylation is a highly complex process, which can lead to closed or open chromatin with increased or decreased transcription of genes (44) and is maintained by two classes of enzymes: histone methyltransferases (HMTs) and histone demethylases (HDMs) (45, 46). H2O2 increases several histone methylation marks including H3K4me3 and H3K27me3 while decreases histone acetylation marks including H3K9ac and H4K8ac (47). In diabetic-induced oxidative stress retinopathy model, H3K4me1, which enhances transcriptional activation, is increased at the promoter of KEAP1, and this is accompanied by increased HMT enzyme SET7/9 (SETD7) (48). Consequently, KEAP1 expression is increased, attenuating NRF2-mediated antioxidant enzyme expression (48). In diabetic nephropathy model, 12(S)-hydroxyeicosatetraenoic acid [12(S)-HETE], which promotes oxidative stress, increases protein levels of SET7, promotes its nuclear translocation and enrichment at profibrotic gene promoters and SET7 (SETD7) gene silencing inhibited 12(S)-HETE-induced profibrotic gene expression (49). Under artery occlusion, H3K4 methyltransferase activity was decreased in astrocytes, with decreased H3K4me3 levels (50). H2O2 can oxidize C residues of protein arginine methyltransferases (PRMTs), decreases PRMT activity and methylation of histone arginine dose-dependently (51). RO/NS can affect HDMs, Lysine-specific demethylase (KDM) LSD1, generates H2O2 as a byproduct of its chromatin remodeling activity during the initial DNA damage response (52) and base excision repair proteins participate in epigenetic reprogramming and modulation of gene expression possibly via alteration of DNA methylation or histone modification (reviewed in (53)).
Overall, more studies would be needed on how different redox agents, dose response, acute versus chronic dosing, in vitro cell culture versus in vivo animal models, cell/tissue specificity and different pathological conditions can affect DNA methylation/demethylation, histone acetylation/methylation and functional gene expression.
3. Mitochondria metabolism, cellular metabolites and epigenetics
Cellular metabolism especially the mitochondrial CAC (Acetyl-CoA, NAD+/NADH, α-ketoglutarate, SAM) is tightly linked to DNA/histone modifications, chromatin remodeling (15–17) and modulating phenotypic gene expression, as shown in Figure 1. RO/NS, mitochondrial metabolites and epigenetics are highly interconnected, whether directly or indirectly. Many of the metabolites are cofactors utilized by the epigenetic enzymes that catalyze the post-translational epigenetic modification including SAM, the methyl donor for all epigenetic methylation reactions; acetyl-CoA, the acetyl (-COH) donor for histone acetylation; NAD+, the cofactor for class III HDACs sirtuins; α-ketoglutarate, the cofactor along with Fe(II) for chromatin/histones demethylases; 2-hydroxyglutarate (R-2HG), inhibitor of demethylases, among others (17), participate in the biological function of the basic epigenetic machinery. We will briefly summarize some of these metabolic co-factors in the context of oxidative stress.
Figure 1:
Interaction of mitochondrial & cellular metabolism with the epigenetic machinery. Acetyl-CoA, acetyl coenzyme A; CoA-SH, Coenzyme A with sulfhydryl (SH) functional group; NAD+/NADH, nicotinamide adenine dinucleotide oxidized (+)/reduced (H) form; JMJCs, Jumonji-C domain-containing histone lysine demethylases; SAM, S-adenosyl methionine; BOHB, β-hydroxyl-butyrate.
SAM (S-Adenosylmethionine):
As discussed above, DNMTs and HMTs add methyl group to DNA or lysine/arginine residues of histones, by transferring the methyl group of SAM to the substrate with the formation of S-Adenosyl homocysteine (SAH). SAM is synthesized from methionine catalyzed by methionine adenosyltransferase (MAT) with ATP. RO/NS reduces the level of SAM (54), and decreases the activity of DNMTs and HMTs. H2O2, via generation of hydroxy radical modifies Cys-121 of MAT resulting in inactivation of MAT’s activity and in SAM biosynthesis (55). In A549 cells, H2O2 decreased SAM levels resulting in hypomethylation of the long interspersed nuclear element-1 (LINE-1) (56). Depletion of GSH resulted in decrease of methionine and DNA hypomethylation (57). However, more work needs to be done on how different redox signals, dose response, acute versus chronic exposure of RO/NS on SAM and DNA/histone modifications.
Acetyl-CoA:
Histone acetylation is catalyzed by HATs including P300/CBP by transferring the acetyl group from acetyl-CoA to lysine/arginine residues of histones. Acetyl-CoA can be synthesized by oxidative metabolism/decarboxylation of pyruvate during glycolysis of glucose in mitochondrial matrix, by β-oxidation of long-chain fatty acids, or by oxidative degradation of certain amino acids. Acetyl-CoA then enters the CAC for the usual energy production. Acetyl-CoA appears to diffuse freely through the nuclear pore complex, and the fluctuation of the pool of available acetyl-CoA in the cytoplasm/nucleus (nucleocytosolic) may alter histone acetylation (17). The level of cellular acetyl-CoA may fluctuate according to cellular metabolism, under certain conditions, it may be becoming limiting factor for the acetyltransferase activity in the nucleus. The oscillation of acetyl-CoA occurs, including at high glucose levels, acetyl-CoA produced through glycolysis, may promote its utilization for lipid synthesis and histone acetylation, while under fasting or survival states, acetyl-CoA may be channeled into the mitochondria for synthesis of ATP and ketone bodies and the mitochondrial amounts of acetyl-CoA increase relative to nucleocytosolic amounts (58).
NAD+:
The cofactor NAD exists in oxidized form (NAD+) and reduced form (NADH), is central to cellular metabolism involving in redox reaction by carrying electrons from one reaction to another. The oxidized NAD+ accepts electrons, becoming reduced NADH, and the reduced NADH will become reducing agent to donate electrons, which is central during glycolysis and CAC metabolism. NAD is synthesized by de novo pathway from tryptophan/aspartic acid or through salvage pathway (59). But importantly, NAD+ is involved in cellular signaling events including ADP-ribosylation, and protein deacetylation, among others (reviewed in (17, 59, 60)). NAD+ is required for ADP-ribosylation by adding one or more ADP-ribose moieties to lysine, glutamate, or aspartate residues of proteins including histones catalyzed by poly-ADP-ribose (PAR) polymerases (PARPs) (16, 60). Under conditions when NAD+ level is altered, many cellular processes involving NAD+ such as cellular metabolism, PARPs and sirtuins would be perturbed. PARPs involve in many cellular processes including cellular stress response. For instance, PARP1 is highly involved in DNA damage detection and repair (reviewed in (60)), PARP-1 is recruited to DNA double strand breaks and mediates DNA damage repair (60). PARP recruits CHD4 (chromodomain nucleosome remodeling and histone deacetylase), part of the repressive nucleosome remodeling and deacetylase (NuRD) complex, which would repress transcription and facilitate DNA repair at the break sites (61). PARP1 inhibits the binding of KDM5B to chromatin and H3K4Me3 and its demethylase activity, leading to increase levels of H3K4me3 at the promoters of PARP1-regulated genes and enhanced expression (62). PARP1 modulates HAT P300 and arginine MT CARM1 on NF-κB to support its transcriptional activity (63, 64). Under oxidative stress and energy depletion, PARP-mediated poly(ADP-ribosylation) will be compartmentalized to the mitochondria, converted from a homeostatic process to a mechanism of cell death (65). As discussed above, the class III HDACs (sirtuins 1–7) remove acetyl marks from histone tails, and have an absolute requirement for NAD+ for their activity (28).
α-Ketoglutarate:
The Jumonji C (JmjC) domain HDMs utilize α-ketoglutarate, Fe(II), and O2 to hydroxylate histone’s methyl group with release of formaldehyde, succinate, CO2 and demethylated histone (reviewed in (66)). In yeast, treatment with H2O2, Rph1, a JmJC HDM enhances the expression of stress response genes coupled with the dissociation of Rph1 from the promoters of these genes (67). In human lung BEAS-2B cells, H2O2 increases H3K4me3 which is blocked by preincubation with ascorbate and in a cell-free system H2O2 inhibited HDM activity where increased Fe(II) rescued this inhibition (47). TET enzymes also require Fe(II) and αKG as co-factors, H2O2 exposed cells have decreased TET protein activity (47).
Butyrate and β-Hydroxybutyrate:
Butyrate and β-hydroxybutyrate (BOHB) are short chain fatty acid metabolites found in mammals. Butyrate produced via bacterial fermentation in the colon is present at millimolar concentrations and is a well-known HDAC inhibitor (68). BOHB, a ketone body, produced by the liver during fasting, calorie restriction, or ketogenic diets can reach millimolar concentrations in the blood circulation, inhibits HDACs and modify histone acetylation (69, 70). Butyrate enhances mitochondrial function during oxidative stress (71) and BOHB may protect from oxidative stress (69). Recent study shows that butyrate is a better HDAC inhibitor than BOHB (68). Further studies would be needed to better understand the interaction of redox and butyrate/BOHB and epigenetic reprogramming.
From the above discussion, clearly, cellular metabolism especially mitochondrial CAC is tightly integrated with the epigenetic machinery DNA and histone modifications, chromatin remodeling, and modulating gene expression. Question remains how different redox signals, dose response, acute versus chronic exposure, in vitro cell culture system versus in vivo animal models, and pathological conditions, would affect cellular/mitochondrial metabolism and epigenetic reprogramming.
4. Redox active phytochemicals and epigenetics
Many health beneficial dietary phytochemicals found abundantly in fruits and vegetables are redox active and also possess epigenetic effects (72). These redox active phytochemicals modify cellular redox status, interact with CAC and cellular metabolism and the epigenetic machinery (Figure 2). We will briefly summarize the most abundant classes of these redox active phytochemicals possessing epigenetic modifying capability.
Figure 2:
Interaction of Redox Active Phytochemicals with Redox Status, CAC and Epigenetics. NRF2-ARE, Nuclear factor erythroid 2-related factor 2-antioxidant response element; GSH, glutathione.
4.1. Ally sulfides
Garlic and other plants pertaining to the allium specie have valuable influence on lipid/ carbohydrate dysmetabolism (73). Diallyl sulfide, and diallyl disulfide (DADS), and diallyl trisulfide (DATS) are oil-soluble organosulfur compounds of garlic and possess redox properties including activation of NRF2-mediating pathways (74). DADS has been shown to exert anti-proliferative effects through epigenetic regulation including suppression of HDAC activity and hyperacetylation of histone H3K14, H4K12, and H4K16 in colon cancer cell lines and in rat colonocytes (75, 76). Allyl mercaptan (AM) is a metabolite derived from DADS and is reported as the most effective HDAC inhibitor among garlic-derived organosulfur compounds and their metabolites (77). At the same concentration, AM shows higher inhibition of HDAC activity than DADS in Caco-2 cells (78). AM decreases HDAC activity in HT29 cells, with increased global histone acetylation and localized hyperacetylation of histone H3 on the P21WAF1 promoter (79).
4.2. Flavonoids
Flavonoids are a very large class of phytochemicals and possess diverse biological activities including redox antioxidant activity, anti-inflammatory activity, and others.
Epigallocatechin-3-gallate (EGCG), found abundantly in green tea, possesses redox activities (80) and regulates NRF2-mediated antioxidant gene expression (81). EGCG inhibits HAT activities including P300, CBP, and PCAF (82). Combination of SFN and EGCG remodels chromatin structure via histone modification and alter methylation patterns in the ERα promoter (83). EGCG can also compact chromatin and silence cancer-related genes by influencing Polycomb-group (PcG) proteins via modulation of histone methylation and acetylation (84).
Genistein is the primary isoflavone found in soy and it activates Nrf2-mediated antioxidant response (85). In addition, genistein mediates promoter hypomethylation or histone hyperacetylation, re-activating different tumor suppressor-associated genes, p16, RARβ, and MGMT (86). However, genistein is known to bind to estrogen receptors (ER), triggering conformational changes leading to transcriptional activations of ER-regulated genes (87) and genistein has been shown to epigenetically activate BRCA1 In Vivo and triple negative breast cancer cells via antagonism toward aryl hydrocarbon receptor (AHR) (88). Further study would be needed the functional role of NRF2-redox-mediated, ER-mediated, AHR-mediated, or other signaling pathways-mediated epigenetic modifying effects of genistein.
Curcumin, a natural compound found in turmeric, possess multi-faceted redox and epigenetic mechanisms including modifying CpG methylation, modulation of DNMTs and HDACs, inhibition of HAT activities, and miRNA modulation (reviewed in (89)). In rat lymphocytes, orally-administered curcumin suppresses lipopolysaccharide (LPS)-induced Dnmt3a and Hdac2/3/4 mRNA expression (90). Pharmacokinetics and pharmacodynamics study of orally administered curcumin in healthy human volunteers shows that curcumin decreases HDAC1, HDAC2, HDAC3, and HDAC4 mRNA expression in leukocytes (91). High concentration of curcumin increases ROS production, induces DNA damage and DNA demethylation potentially via DNA repair pathway (92). DNA methylomic and transcriptomic alterations and cancer preventive effect by curcumin was reported in the azoxymethane (AOM)-dextran sulfate sodium (DSS)-induced colon cancer C57BL/6 mice (93). Analysis of the differentially expressed and differentially methylated genes in pairwise comparisons, several signaling pathways including LPS/IL-1-mediated inhibition of RXR function, Nrf2-mediated oxidative stress response, production of NO and ROS in macrophages and IL-6 signaling were identified to be modified by curcumin (93). Among all the differentially methylated genes, tumor necrosis factor (TNF) was selected for further study and its CpG promoter was hypomethylated and mRNA expression was decreased in colon epithelial cells and colon tumors of AOM-DSS mice and these events were reversed by curcumin (93). Using ex vivo lipopolysaccharide (LPS) treated peritoneal macrophages from Nrf2+/+and Nrf2−/− mice, curcumin’s anti-inflammatory and anti-oxidant effects require Nrf2 (94). CUR can both increase and decrease the accessibility of DNA and thereby influence transcriptional responses to the ligand-activated AHR (95). Question remains on the contribution of redox-mediated, AHR-mediated, or other signaling pathway-mediated epigenetic modifying effects of curcumin.
Anthocyanidins are the sugar-free counterparts of anthocyanins. Anthocyanidins have been shown to activate Nrf2 pathway, correlating with demethylation of CpGs of the mouse Nrf2 promoter region coupled with decrease protein expression DNMT1, DNMT3a, and class I/II HDACs (96). In a high-fat diet-induced redox sensitive signaling (NF-κB and ERK1/2), anthocyanin supplementation attenuated these pathways (97). Delphinidin and pelargonidin inhibit TPA-induced transformation of mouse epidermal JB6 P+ cells coupled with promoter demethylation-mediated activation of Nrf2 (96, 98).
4.3. Isothiocyanates
Isothiocyanates, derived from glucosinolates found abundantly in crucifers, are redox active electrophiles and possess many epigenetic properties (reviewed in (99, 100)). The isothiocyanate sulforaphane was first reported to inhibit HDAC, enhanced histone acetylation, depression of P21 and BAX, and induction of cell cycle arrest/apoptosis (reviewed in (101)). Subsequently, many studies show that isothiocyanates modulate the epigenetic machinery in multiple systems (reviewed in (102)). Metanalysis of SFN shows SFN could exert its effect by restoring the ER gene expression by modulating epigenetic events in human breast cancer cell line MDA-MB-231 (103). Since the isothiocyanates are potent electrophiles, question remains on the contribution of redox electrophile-mediated, ER-mediated, or other signaling pathway-mediated epigenetic modifying effects of isothiocyanates.
4.4. Triterpenoids
Triterpenoids are structurally diverse redox active phytochemicals and possess epigenetics properties (reviewed in (104)). Ursolic acid demethylates the CpG sites in the Nrf2 promoter region, and correlates with the re-expression of Nrf2, coupled with inhibition of mRNA expression of DNMTs and HDACs, and HDAC activity (105). In rat leukocytes, orally administered ursolic acid inhibits LPS-induced DNMTs and HDACs mRNA (106) and in a UVB-induced skin carcinogenesis model, ursolic acid significantly reduced tumorigenesis coupled with modification of DNA methylome of antioxidative, anti-inflammatory, and anticancer pathways (107).
Due to space limitation, not all the redox active phytochemicals that possess capability to modify epigenetic machinery are discussed here. Some of the future challenges in integrating redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals include dose response, in vitro cell culture versus in vivo animal models versus in human, different redox stimuli, acute versus chronic dosing and in vivo absorption, distribution, and bioavailability of phytochemicals (108).
5. Gut metabolites and epigenetics
Gut microbiota-derived metabolites are small molecules that are produced as intermediate or end products of microbial metabolism. These metabolites can be derived from bacterial metabolism of dietary phytochemicals or modification of host molecules such as bile acids. Alterations by dietary phytochemicals in the composition and function of the microbiota have been implicated in patients with inflammatory conditions of the gastrointestinal tract (109–111). These alterations are likely to cause changes to gut microbial metabolite profiles, of which the main classes include short-chain fatty acids, bile acids, and tryptophan metabolites. Many phytochemicals, together with dietary fiber, have been proposed to modulate the gut microbiota. Some of the phytochemicals that positively affect the gut microbiota include polyphenols, carotenoids, phytosterols/phytostanols, lignans, alkaloids, glucosinolates and terpenes (112). Some polyphenols may act as prebiotics, while carotenoids have been shown to alter immunoglobulin A expression, an important factor for bacteria colonization (111, 113–115). Other phytochemicals may interact with the gut mucosa, another important factor for colonization, and prevent their degradation. Some polyphenols have shown to influence bacterial communication, interacting with quorum sensing. Short chain fatty acids production depends on gut microbiota composition and diet. Short chain fatty acids, mainly butyrate and acetate, reduce inflammation but increase the anti-inflammatory response of the adaptive immunity. Butyrate has also been shown to inhibit the activity of HDAC through suppressing NF-κB activation and synthesis of interferon gamma and increasing PPAR- γ (116, 117). More study would be needed on the interaction of microbiota-derived metabolites and the epigenetic machinery in the gut as well as in other tissues/organs.
6. Conclusion and perspective
Biological redox signaling and epigenetics play important roles in many diseases’ manifestation. Increasing evidence suggests that redox signaling and cellular/mitochondrial metabolism are highly integrated with reprogramming of epigenetic machinery contributing to many human diseases. Many health beneficial phytochemicals are redox active and possess epigenetic modifying capability. Future challenges include how redox bioactive phytochemicals would interact with the basic redox signaling, cellular/mitochondrial metabolism and the epigenetic machinery contributing to the overall health beneficial effects and impacting diseases.
Highlights.
Cellular oxidative stress occurs when reactive oxygen/nitrogen species (RO/NS) exceed the cellular reductive/antioxidant capacity.
Many health beneficial phytochemicals possess redox capability that counteract RO/NS either by directly scavenging the radicals or via inductive mechanism of cellular defense antioxidant/reductive enzymes.
This review summarizes the latest advances on the interactions between redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals and the future challenges of integrating these events in human health.
Acknowledgement
This work was support in part by Institutional Funds, by R01 CA200129, from the National Cancer Institute (NCI), and R01 AT009152 from National Center for Complementary and Integrative Health (NCCIH) of the National Institute of Health (NIH) awarded to Dr. Ah-Ng Tony Kong.
We apologize that due to the limitations of number of references, we are not able to cite many relevant works.
Abbreviations
- ROS
reactive oxygen species
- RNS
reactive nitrogen species
- RO/NS
reactive oxygen/nitrogen species
- CAC
citric acid cycle
- TCA
tricarboxylic acid cycle
- SOD
superoxide dismutase
- MnSOD
manganese superoxide dismutase
- NOS
nitric oxide synthase
- GST
glutathione-S-transferase
- DNMT
DNA methyltransferase
- SAM
S-adenosyl methionine
- TET
ten-eleven translocation enzyme
- HDAC
histone deacetylase
- HAT
histone acetyltransferase
- HMT
histone methyltransferase
- HDM
histone demethylase
- CVD
cardiovascular disease
- NOS
nitric oxide synthase
- AD
Alzheimer’s disease
- COPD
chronic obstructive pulmonary disease
- NAFLD
non-alcoholic fatty liver disease
- IKK
inhibitory κB kinase
- DAS
diallyl sulfide
- DADS
diallyl disulfide
- DATS
diallyl trisulfide
- GPS
glutathione peroxidase
- iNOS
inducible nitric oxide synthase
- AM
Allyl mercaptan
- ITC
isothiocyanate
- SFN
sulforaphane
- AITC
allyl-ITC
- BITC
benzyl-ITC
- PEITC
phenethyl-ITC
- UA
ursolic acid
- OA
oleanolic acid
- BA
betulinic acid
- CDDO
cyano-3,12-dioxooleana-1,9 (11)-dien-28-oic
- DMF
dimethyl fumarate
- LPS
lipopolysaccharide
- IL-6
interleukin-6
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Thorstenson DC. The concept of electron activity and its relation to redox potentials in aqueous geochemical systems: U.S. Geological Survey Open-File Report; 1984. [Google Scholar]
- 2.Sies H. Oxidative Stress. 1 ed. London: Academic Press; 1985. 1/28/1985. 507 p. [Google Scholar]
- 3.Sies H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017;11:613–9. Epub 2017/01/23. doi: 10.1016/j.redox.2016.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fischer EH, Krebs EG. Conversion of phosphorylase b to phosphorylase a in muscle extracts. J Biol Chem. 1955;216(1):121–32. Epub 1955/09/01. [PubMed] [Google Scholar]
- 5.Beckman KB, Ames BN. The free radical theory of aging matures. Physiol Rev. 1998;78(2):547–81. Epub 1998/04/30. doi: 10.1152/physrev.1998.78.2.547. [DOI] [PubMed] [Google Scholar]
- 6.Cadenas E, Davies KJ. Mitochondrial free radical generation, oxidative stress, and aging. Free Radic Biol Med. 2000;29(3–4):222–30. Epub 2000/10/18. doi: 10.1016/s0891-5849(00)00317-8. [DOI] [PubMed] [Google Scholar]
- 7.Zheng M, Åslund F, Storz G. Activation of the OxyR Transcription Factor by Reversible Disulfide Bond Formation. Science. 1998;279(5357):1718–22. doi: 10.1126/science.279.5357.1718. [DOI] [PubMed] [Google Scholar]
- 8.Marinho HS, Real C, Cyrne L, Soares H, Antunes F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014;2:535–62. Epub 2014/03/19. doi: 10.1016/j.redox.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sies H. Oxidative stress: a concept in redox biology and medicine. Redox Biol. 2015;4:180–3. Epub 2015/01/16. doi: 10.1016/j.redox.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Baird L, Yamamoto M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol Cell Biol. 2020;40(13). Epub 2020/04/15. doi: 10.1128/mcb.00099-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cho SH, Lee CH, Ahn Y, Kim H, Kim H, Ahn CY, Yang KS, Lee SR. Redox regulation of PTEN and protein tyrosine phosphatases in H(2)O(2) mediated cell signaling. FEBS Lett. 2004;560(1–3):7–13. Epub 2004/03/17. doi: 10.1016/s0014-5793(04)00112-7. [DOI] [PubMed] [Google Scholar]
- 12.Kray AE, Carter RS, Pennington KN, Gomez RJ, Sanders LE, Llanes JM, Khan WN, Ballard DW, Wadzinski BE. Positive regulation of IkappaB kinase signaling by protein serine/threonine phosphatase 2A. J Biol Chem. 2005;280(43):35974–82. Epub 2005/08/30. doi: 10.1074/jbc.M506093200. [DOI] [PubMed] [Google Scholar]
- 13.Codreanu SG, Adams DG, Dawson ES, Wadzinski BE, Liebler DC. Inhibition of protein phosphatase 2A activity by selective electrophile alkylation damage. Biochemistry. 2006;45(33):10020–9. Epub 2006/08/16. doi: 10.1021/bi060551n. [DOI] [PubMed] [Google Scholar]
- 14.Xu C, Shen G, Chen C, Gelinas C, Kong AN. Suppression of NF-kappaB and NF-kappaB-regulated gene expression by sulforaphane and PEITC through IkappaBalpha, IKK pathway in human prostate cancer PC-3 cells. Oncogene. 2005;24(28):4486–95. doi: 10.1038/sj.onc.1208656. [DOI] [PubMed] [Google Scholar]
- 15.Wallace DC, Fan W. Energetics, epigenetics, mitochondrial genetics. Mitochondrion. 2010;10(1):12–31. Epub 2009/10/03. doi: 10.1016/j.mito.2009.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cyr AR, Domann FE. The redox basis of epigenetic modifications: from mechanisms to functional consequences. Antioxid Redox Signal. 2011;15(2):551–89. Epub 2010/10/06. doi: 10.1089/ars.2010.3492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Janke R, Dodson AE, Rine J. Metabolism and epigenetics. Annu Rev Cell Dev Biol. 2015;31:473–96. Epub 2015/09/12. doi: 10.1146/annurev-cellbio-100814-125544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Feinberg AP. The Key Role of Epigenetics in Human Disease Prevention and Mitigation. N Engl J Med. 2018;378(14):1323–34. Epub 2018/04/05. doi: 10.1056/NEJMra1402513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kulis M, Esteller M. DNA methylation and cancer. Adv Genet. 2010;70:27–56. Epub 2010/10/06. doi: 10.1016/b978-0-12-380866-0.60002-2. [DOI] [PubMed] [Google Scholar]
- 20.O’Hagan HM, Wang W, Sen S, Destefano Shields C, Lee SS, Zhang YW, Clements EG, Cai Y, Van Neste L, Easwaran H, Casero RA, Sears CL, Baylin SB. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell. 2011;20(5):606–19. Epub 2011/11/19. doi: 10.1016/j.ccr.2011.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.van der Wijst MG, Venkiteswaran M, Chen H, Xu G-L, Plösch T, Rots MG. Local chromatin microenvironment determines DNMT activity: from DNA methyltransferase to DNA demethylase or DNA dehydroxymethylase. Epigenetics. 2015;10(8):671–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hepburn PA, Margison GP, Tisdale MJ. Enzymatic methylation of cytosine in DNA is prevented by adjacent O6-methylguanine residues. J Biol Chem. 1991;266(13):7985–7. Epub 1991/05/05. [PubMed] [Google Scholar]
- 23.Valinluck V, Tsai HH, Rogstad DK, Burdzy A, Bird A, Sowers LC. Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic Acids Res. 2004;32(14):4100–8. Epub 2004/08/11. doi: 10.1093/nar/gkh739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature. 2013;502(7472):472–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Branco MR, Ficz G, Reik W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat Rev Genet. 2011;13(1):7–13. Epub 2011/11/16. doi: 10.1038/nrg3080. [DOI] [PubMed] [Google Scholar]
- 26.Xin Y-J, Yuan B, Yu B, Wang Y-Q, Wu J-J, Zhou W-H, Qiu Z. Tet1-mediated DNA demethylation regulates neuronal cell death induced by oxidative stress. Scientific reports. 2015;5:7645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hassell KN. Histone Deacetylases and their Inhibitors in Cancer Epigenetics. Diseases. 2019;7(4). Epub 2019/11/07. doi: 10.3390/diseases7040057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sauve AA, Wolberger C, Schramm VL, Boeke JD. The Biochemistry of Sirtuins. Annual Review of Biochemistry. 2006;75(1):435–65. doi: 10.1146/annurev.biochem.74.082803.133500. [DOI] [PubMed] [Google Scholar]
- 29.Nagarajan N, Oka S, Sadoshima J. Modulation of signaling mechanisms in the heart by thioredoxin 1. Free Radic Biol Med. 2017;109:125–31. Epub 2016/12/21. doi: 10.1016/j.freeradbiomed.2016.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ago T, Liu T, Zhai P, Chen W, Li H, Molkentin JD, Vatner SF, Sadoshima J. A redox-dependent pathway for regulating class II HDACs and cardiac hypertrophy. Cell. 2008;133(6):978–93. Epub 2008/06/17. doi: 10.1016/j.cell.2008.04.041. [DOI] [PubMed] [Google Scholar]
- 31.Sobotta MC, Liou W, Stöcker S, Talwar D, Oehler M, Ruppert T, Scharf AN, Dick TP. Peroxiredoxin-2 and STAT3 form a redox relay for H2O2 signaling. Nat Chem Biol. 2015;11(1):64–70. Epub 2014/11/18. doi: 10.1038/nchembio.1695. [DOI] [PubMed] [Google Scholar]
- 32.Parmigiani R, Xu W, Venta-Perez G, Erdjument-Bromage H, Yaneva M, Tempst P, Marks P. HDAC6 is a specific deacetylase of peroxiredoxins and is involved in redox regulation. Proceedings of the National Academy of Sciences. 2008;105(28):9633–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Doyle K, Fitzpatrick FA. Redox signaling, alkylation (carbonylation) of conserved cysteines inactivates class I histone deacetylases 1, 2, and 3 and antagonizes their transcriptional repressor function. Journal of Biological Chemistry. 2010;285(23):17417–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hu T, Schreiter FC, Bagchi RA, Tatman PD, Hannink M, McKinsey TA. HDAC5 catalytic activity suppresses cardiomyocyte oxidative stress and NRF2 target gene expression. J Biol Chem. 2019;294(21):8640–52. Epub 2019/04/10. doi: 10.1074/jbc.RA118.007006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jänsch N, Meyners C, Muth M, Kopranovic A, Witt O, Oehme I, Meyer-Almes FJ. The enzyme activity of histone deacetylase 8 is modulated by a redox-switch. Redox Biol. 2019;20:60–7. Epub 2018/10/08. doi: 10.1016/j.redox.2018.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang Z, Zang C, Cui K, Schones DE, Barski A, Peng W, Zhao K. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell. 2009;138(5):1019–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Allfrey VG, Faulkner R, Mirsky AE. ACETYLATION AND METHYLATION OF HISTONES AND THEIR POSSIBLE ROLE IN THE REGULATION OF RNA SYNTHESIS. Proc Natl Acad Sci U S A. 1964;51(5):786–94. Epub 1964/05/01. doi: 10.1073/pnas.51.5.786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ojima Y, Suryadarma P, Tsuchida K, Taya M. Accumulation of pyruvate by changing the redox status in Escherichia coli. Biotechnol Lett. 2012;34(5):889–93. Epub 2012/01/05. doi: 10.1007/s10529-011-0842-y. [DOI] [PubMed] [Google Scholar]
- 39.R MS, Wang Y, Zhang X, Cheng H, Sun L, He S, Hao F. Redox Components: Key Regulators of Epigenetic Modifications in Plants. Int J Mol Sci. 2020;21(4). Epub 2020/02/26. doi: 10.3390/ijms21041419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Miernyk JA, Randall DD. Some kinetic and regulatory properties of the pea mitochondrial pyruvate dehydrogenase complex. Plant Physiol. 1987;83(2):306–10. Epub 1987/02/01. doi: 10.1104/pp.83.2.306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang P, Zhao L, Hou H, Zhang H, Huang Y, Wang Y, Li H, Gao F, Yan S, Li L. Epigenetic Changes are Associated with Programmed Cell Death Induced by Heat Stress in Seedling Leaves of Zea mays. Plant Cell Physiol. 2015;56(5):965–76. Epub 2015/02/12. doi: 10.1093/pcp/pcv023. [DOI] [PubMed] [Google Scholar]
- 42.Rahman I, Gilmour PS, Jimenez LA, MacNee W. Oxidative stress and TNF-alpha induce histone acetylation and NF-kappaB/AP-1 activation in alveolar epithelial cells: potential mechanism in gene transcription in lung inflammation. Mol Cell Biochem. 2002;234–235(1–2):239–48. Epub 2002/08/07. [PubMed] [Google Scholar]
- 43.Tomita K, Barnes PJ, Adcock IM. The effect of oxidative stress on histone acetylation and IL-8 release. Biochem Biophys Res Commun. 2003;301(2):572–7. Epub 2003/02/05. doi: 10.1016/s0006-291x(02)03029-2. [DOI] [PubMed] [Google Scholar]
- 44.Kietzmann T, Petry A, Shvetsova A, Gerhold JM, Görlach A. The epigenetic landscape related to reactive oxygen species formation in the cardiovascular system. Br J Pharmacol. 2017;174(12):1533–54. Epub 2017/03/24. doi: 10.1111/bph.13792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhang P, Torres K, Liu X, Liu C-g, E Pollock R. An overview of chromatin-regulating proteins in cells. Current Protein and Peptide Science. 2016;17(5):401–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nature Reviews Genetics. 2016;17(8):487–500. doi: 10.1038/nrg.2016.59. [DOI] [PubMed] [Google Scholar]
- 47.Niu Y, DesMarais TL, Tong Z, Yao Y, Costa M. Oxidative stress alters global histone modification and DNA methylation. Free Radic Biol Med. 2015;82:22–8. doi: 10.1016/j.freeradbiomed.2015.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mishra M, Zhong Q, Kowluru RA. Epigenetic modifications of Keap1 regulate its interaction with the protective factor Nrf2 in the development of diabetic retinopathy. Invest Ophthalmol Vis Sci. 2014;55(11):7256–65. Epub 2014/10/11. doi: 10.1167/iovs.14-15193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yuan H, Reddy MA, Deshpande S, Jia Y, Park JT, Lanting LL, Jin W, Kato M, Xu ZG, Das S, Natarajan R. Epigenetic Histone Modifications Involved in Profibrotic Gene Regulation by 12/15-Lipoxygenase and Its Oxidized Lipid Products in Diabetic Nephropathy. Antioxid Redox Signal. 2016;24(7):361–75. Epub 2015/10/24. doi: 10.1089/ars.2015.6372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chisholm NC, Henderson ML, Selvamani A, Park MJ, Dindot S, Miranda RC, Sohrabji F. Histone methylation patterns in astrocytes are influenced by age following ischemia. Epigenetics. 2015;10(2):142–52. Epub 2015/01/08. doi: 10.1080/15592294.2014.1001219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Morales Y, Nitzel DV, Price OM, Gui S, Li J, Qu J, Hevel JM. Redox Control of Protein Arginine Methyltransferase 1 (PRMT1) Activity. J Biol Chem. 2015;290(24):14915–26. Epub 2015/04/26. doi: 10.1074/jbc.M115.651380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Duquette ML, Kim J, Shi LZ, Berns MW. LSD1 mediated changes in the local redox environment during the DNA damage response. PLoS One. 2018;13(8):e0201907. Epub 2018/08/11. doi: 10.1371/journal.pone.0201907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Li J, Braganza A, Sobol RW. Base excision repair facilitates a functional relationship between Guanine oxidation and histone demethylation. Antioxid Redox Signal. 2013;18(18):2429–43. Epub 2013/01/15. doi: 10.1089/ars.2012.5107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Panayiotidis MI, Stabler SP, Allen RH, Pappa A, White CW. Oxidative stress-induced regulation of the methionine metabolic pathway in human lung epithelial-like (A549) cells. Mutat Res. 2009;674(1–2):23–30. Epub 2008/11/18. doi: 10.1016/j.mrgentox.2008.10.006. [DOI] [PubMed] [Google Scholar]
- 55.Sánchez-Góngora E, Ruiz F, Mingorance J, An W, Corrales FJ, Mato JM. Interaction of liver methionine adenosyltransferase with hydroxyl radical. The FASEB Journal. 1997;11(12):1013–9. doi: 10.1096/fasebj.11.12.9337154. [DOI] [PubMed] [Google Scholar]
- 56.Kloypan C, Srisa-art M, Mutirangura A, Boonla C. LINE-1 hypomethylation induced by reactive oxygen species is mediated via depletion of S-adenosylmethionine. Cell Biochem Funct. 2015;33(6):375–85. Epub 2015/07/17. doi: 10.1002/cbf.3124. [DOI] [PubMed] [Google Scholar]
- 57.Lertratanangkoon K, Wu CJ, Savaraj N, Thomas ML. Alterations of DNA methylation by glutathione depletion. Cancer Letters. 1997;120(2):149–56. doi: 10.1016/S0304-3835(97)00300-5. [DOI] [PubMed] [Google Scholar]
- 58.Shi L, Tu BP. Acetyl-CoA and the regulation of metabolism: mechanisms and consequences. Current Opinion in Cell Biology. 2015;33:125–31. doi: 10.1016/j.ceb.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Xiao W, Wang RS, Handy DE, Loscalzo J. NAD(H) and NADP(H) Redox Couples and Cellular Energy Metabolism. Antioxid Redox Signal. 2018;28(3):251–72. Epub 2017/06/27. doi: 10.1089/ars.2017.7216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gupte R, Liu Z, Kraus WL. PARPs and ADP-ribosylation: recent advances linking molecular functions to biological outcomes. Genes & development. 2017;31(2):101–26. doi: 10.1101/gad.291518.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chou DM, Adamson B, Dephoure NE, Tan X, Nottke AC, Hurov KE, Gygi SP, Colaiácovo MP, Elledge SJ. A chromatin localization screen reveals poly (ADP ribose)-regulated recruitment of the repressive polycomb and NuRD complexes to sites of DNA damage. Proc Natl Acad Sci U S A. 2010;107(43):18475–80. Epub 2010/10/13. doi: 10.1073/pnas.1012946107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Krishnakumar R, Kraus WL. PARP-1 regulates chromatin structure and transcription through a KDM5B-dependent pathway. Mol Cell. 2010;39(5):736–49. Epub 2010/09/14. doi: 10.1016/j.molcel.2010.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hassa PO, Buerki C, Lombardi C, Imhof R, Hottiger MO. Transcriptional coactivation of nuclear factor-kappaB-dependent gene expression by p300 is regulated by poly(ADP)-ribose polymerase-1. J Biol Chem. 2003;278(46):45145–53. Epub 2003/09/10. doi: 10.1074/jbc.M307957200. [DOI] [PubMed] [Google Scholar]
- 64.Hassa PO, Covic M, Bedford MT, Hottiger MO. Protein arginine methyltransferase 1 coactivates NF-kappaB-dependent gene expression synergistically with CARM1 and PARP1. J Mol Biol. 2008;377(3):668–78. Epub 2008/02/19. doi: 10.1016/j.jmb.2008.01.044. [DOI] [PubMed] [Google Scholar]
- 65.Du L, Zhang X, Han YY, Burke NA, Kochanek PM, Watkins SC, Graham SH, Carcillo JA, Szabó C, Clark RSB. Intra-mitochondrial Poly(ADP-ribosylation) Contributes to NAD+ Depletion and Cell Death Induced by Oxidative Stress. Journal of Biological Chemistry. 2003;278(20):18426–33. doi: 10.1074/jbc.M301295200. [DOI] [PubMed] [Google Scholar]
- 66.Leon KE, Aird KM. Jumonji C Demethylases in Cellular Senescence. Genes (Basel). 2019;10(1). Epub 2019/01/13. doi: 10.3390/genes10010033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Liang CY, Wang LC, Lo WS. Dissociation of the H3K36 demethylase Rph1 from chromatin mediates derepression of environmental stress-response genes under genotoxic stress in Saccharomyces cerevisiae. Mol Biol Cell. 2013;24(20):3251–62. Epub 2013/08/30. doi: 10.1091/mbc.E12-11-0820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Steliou K, Boosalis MS, Perrine SP, Sangerman J, Faller DV. Butyrate histone deacetylase inhibitors. Biores Open Access. 2012;1(4):192–8. Epub 2013/03/22. doi: 10.1089/biores.2012.0223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Newman JC, Verdin E. β-hydroxybutyrate: Much more than a metabolite. Diabetes Research and Clinical Practice. 2014;106(2):173–81. doi: 10.1016/j.diabres.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Chriett S, Dąbek A, Wojtala M, Vidal H, Balcerczyk A, Pirola L. Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Scientific Reports. 2019;9(1):742. doi: 10.1038/s41598-018-36941-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rose S, Bennuri SC, Davis JE, Wynne R, Slattery JC, Tippett M, Delhey L, Melnyk S, Kahler SG, MacFabe DF, Frye RE. Butyrate enhances mitochondrial function during oxidative stress in cell lines from boys with autism. Translational Psychiatry. 2018;8(1):42. doi: 10.1038/s41398-017-0089-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lee JH, Khor TO, Shu L, Su ZY, Fuentes F, Kong AN. Dietary phytochemicals and cancer prevention: Nrf2 signaling, epigenetics, and cell death mechanisms in blocking cancer initiation and progression. Pharmacol Ther. 2013;137(2):153–71. Epub 2012/10/09. doi: 10.1016/j.pharmthera.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Milner JA. A historical perspective on garlic and cancer. J Nutr. 2001;131(3s):1027S–31S. Epub 2001/03/10. doi: 10.1093/jn/131.3.1027S. [DOI] [PubMed] [Google Scholar]
- 74.Chen C, Pung D, Leong V, Hebbar V, Shen G, Nair S, Li W, Kong AN. Induction of detoxifying enzymes by garlic organosulfur compounds through transcription factor Nrf2: effect of chemical structure and stress signals. Free Radic Biol Med. 2004;37(10):1578–90. doi: 10.1016/j.freeradbiomed.2004.07.021. [DOI] [PubMed] [Google Scholar]
- 75.Druesne N, Pagniez A, Mayeur C, Thomas M, Cherbuy C, Duée PH, Martel P, Chaumontet C. Diallyl disulfide (DADS) increases histone acetylation and p21(waf1/cip1) expression in human colon tumor cell lines. Carcinogenesis. 2004;25(7):1227–36. Epub 2004/02/21. doi: 10.1093/carcin/bgh123. [DOI] [PubMed] [Google Scholar]
- 76.Druesne-Pecollo N, Chaumontet C, Pagniez A, Vaugelade P, Bruneau A, Thomas M, Cherbuy C, Duée PH, Martel P. In vivo treatment by diallyl disulfide increases histone acetylation in rat colonocytes. Biochem Biophys Res Commun. 2007;354(1):140–7. Epub 2007/01/11. doi: 10.1016/j.bbrc.2006.12.158. [DOI] [PubMed] [Google Scholar]
- 77.Rajendran P, Ho E, Williams DE, Dashwood RH. Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin Epigenetics. 2011;3(1):4. Epub 2012/01/17. doi: 10.1186/1868-7083-3-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Druesne N, Pagniez A, Mayeur C, Thomas M, Cherbuy C, Duée P-H, Martel P, Chaumontet C. Diallyl disulfide (DADS) increases histone acetylation and p21 waf1/cip1 expression in human colon tumor cell lines. Carcinogenesis. 2004;25(7):1227–36. doi: 10.1093/carcin/bgh123. [DOI] [PubMed] [Google Scholar]
- 79.Nian H, Delage B, Pinto JT, Dashwood RH. Allyl mercaptan, a garlic-derived organosulfur compound, inhibits histone deacetylase and enhances Sp3 binding on the P21WAF1 promoter. Carcinogenesis. 2008;29(9):1816–24. Epub 2008/07/17. doi: 10.1093/carcin/bgn165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kim HS, Quon MJ, Kim JA. New insights into the mechanisms of polyphenols beyond antioxidant properties; lessons from the green tea polyphenol, epigallocatechin 3-gallate. Redox Biol. 2014;2:187–95. Epub 2014/02/05. doi: 10.1016/j.redox.2013.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Shen G, Xu C, Hu R, Jain MR, Nair S, Lin W, Yang CS, Chan JY, Kong AN. Comparison of (−)-epigallocatechin-3-gallate elicited liver and small intestine gene expression profiles between C57BL/6J mice and C57BL/6J/Nrf2 (−/−) mice. Pharm Res. 2005;22(11):1805–20. Epub 2005/09/01. doi: 10.1007/s11095-005-7546-8. [DOI] [PubMed] [Google Scholar]
- 82.Choi KC, Jung MG, Lee YH, Yoon JC, Kwon SH, Kang HB, Kim MJ, Cha JH, Kim YJ, Jun WJ, Lee JM, Yoon HG. Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation. Cancer Res. 2009;69(2):583–92. Epub 2009/01/17. doi: 10.1158/0008-5472.CAN-08-2442. [DOI] [PubMed] [Google Scholar]
- 83.Li Y, Meeran SM, Tollefsbol TO. Combinatorial bioactive botanicals re-sensitize tamoxifen treatment in ER-negative breast cancer via epigenetic reactivation of ERalpha expression. Sci Rep. 2017;7(1):9345. Epub 2017/08/26. doi: 10.1038/s41598-017-09764-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Balasubramanian S, Adhikary G, Eckert RL. The Bmi-1 polycomb protein antagonizes the (−)-epigallocatechin-3-gallate-dependent suppression of skin cancer cell survival. Carcinogenesis. 2010;31(3):496–503. Epub 2009/12/18. doi: 10.1093/carcin/bgp314. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 85.Barve A, Khor TO, Nair S, Lin W, Yu S, Jain MR, Chan JY, Kong AN. Pharmacogenomic profile of soy isoflavone concentrate in the prostate of Nrf2 deficient and wild-type mice. Journal of pharmaceutical sciences. 2008;97(10):4528–45. doi: 10.1002/jps.21311. [DOI] [PubMed] [Google Scholar]
- 86.Fang MZ, Chen D, Sun Y, Jin Z, Christman JK, Yang CS. Reversal of hypermethylation and reactivation of p16INK4a, RARbeta, and MGMT genes by genistein and other isoflavones from soy. Clin Cancer Res. 2005;11(19 Pt 1):7033–41. doi: 10.1158/1078-0432.CCR-05-0406. [DOI] [PubMed] [Google Scholar]
- 87.Vaya J, Tamir S. The relation between the chemical structure of flavonoids and their estrogen-like activities. Curr Med Chem. 2004;11(10):1333–43. Epub 2004/05/12. doi: 10.2174/0929867043365251. [DOI] [PubMed] [Google Scholar]
- 88.Donovan MG, Selmin OI, Doetschman TC, Romagnolo DF. Epigenetic Activation of BRCA1 by Genistein In Vivo and Triple Negative Breast Cancer Cells Linked to Antagonism toward Aryl Hydrocarbon Receptor. Nutrients. 2019;11(11). Epub 2019/10/28. doi: 10.3390/nu11112559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wu R, Wang L, Yin R, Hudlikar R, Li S, Kuo HD, Peter R, Sargsyan D, Guo Y, Liu X, Kong AN. Epigenetics/epigenomics and prevention by curcumin of early stages of inflammatory-driven colon cancer. Molecular carcinogenesis. 2020;59(2):227–36. Epub 2019/12/11. doi: 10.1002/mc.23146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Boyanapalli SSS, Huang Y, Su Z, Cheng D, Zhang C, Guo Y, Rao R, Androulakis IP, Kong AN. Pharmacokinetics and Pharmacodynamics of Curcumin in regulating anti-inflammatory and epigenetic gene expression. Biopharmaceutics & drug disposition. 2018. Epub 2018/06/06. doi: 10.1002/bdd.2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Cheng D, Li W, Wang L, Lin T, Poiani G, Wassef A, Hudlikar R, Ondar P, Brunetti L, Kong AN. Pharmacokinetics, Pharmacodynamics, and PKPD Modeling of Curcumin in Regulating Antioxidant and Epigenetic Gene Expression in Healthy Human Volunteers. Mol Pharm. 2019;16(5):1881–9. Epub 2019/03/13. doi: 10.1021/acs.molpharmaceut.8b01246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tong R, Wu X, Liu Y, Liu Y, Zhou J, Jiang X, Zhang L, He X, Ma L. Curcumin-Induced DNA Demethylation in Human Gastric Cancer Cells Is Mediated by the DNA-Damage Response Pathway. Oxidative Medicine and Cellular Longevity. 2020;2020:2543504. doi: 10.1155/2020/2543504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Guo Y, Wu R, Gaspar JM, Sargsyan D, Su ZY, Zhang C, Gao L, Cheng D, Li W, Wang C, Yin R, Fang M, Verzi MP, Hart RP, Kong AN. DNA methylome and transcriptome alterations and cancer prevention by curcumin in colitis-accelerated colon cancer in mice. Carcinogenesis. 2018;39(5):669–80. Epub 2018/03/17. doi: 10.1093/carcin/bgy043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Boyanapalli SS, Paredes-Gonzalez X, Fuentes F, Zhang C, Guo Y, Pung D, Saw CL, Kong AN. Nrf2 knockout attenuates the anti-inflammatory effects of phenethyl isothiocyanate and curcumin. Chem Res Toxicol. 2014;27(12):2036–43. Epub 2014/11/12. doi: 10.1021/tx500234h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Mohammadi-Bardbori A, Akbarizadeh AR, Delju F, Rannug A. Chromatin remodeling by curcumin alters endogenous aryl hydrocarbon receptor signaling. Chem Biol Interact. 2016;252:19–27. Epub 2016/04/05. doi: 10.1016/j.cbi.2016.03.037. [DOI] [PubMed] [Google Scholar]
- 96.Kuo HD, Wu R, Li S, Yang AY, Kong AN. Anthocyanin Delphinidin Prevents Neoplastic Transformation of Mouse Skin JB6 P+ Cells: Epigenetic Re-activation of Nrf2-ARE Pathway. Aaps j. 2019;21(5):83. Epub 2019/06/30. doi: 10.1208/s12248-019-0355-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cremonini E, Daveri E, Mastaloudis A, Adamo AM, Mills D, Kalanetra K, Hester SN, Wood SM, Fraga CG, Oteiza PI. Anthocyanins protect the gastrointestinal tract from high fat diet-induced alterations in redox signaling, barrier integrity and dysbiosis. Redox Biol. 2019;26:101269. Epub 2019/07/23. doi: 10.1016/j.redox.2019.101269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Li S, Li W, Wang C, Wu R, Yin R, Kuo HC, Wang L, Kong AN. Pelargonidin reduces the TPA induced transformation of mouse epidermal cells -potential involvement of Nrf2 promoter demethylation. Chem Biol Interact. 2019;309:108701. Epub 2019/06/11. doi: 10.1016/j.cbi.2019.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fuentes F, Paredes-Gonzalez X, Kong AN. Dietary Glucosinolates Sulforaphane, Phenethyl Isothiocyanate, Indole-3-Carbinol/3,3’-Diindolylmethane: Anti-Oxidative Stress/Inflammation, Nrf2, Epigenetics/Epigenomics and In Vivo Cancer Chemopreventive Efficacy. Curr Pharmacol Rep. 2015;1(3):179–96. Epub 2015/10/13. doi: 10.1007/s40495-015-0017-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hudlikar R, Wang L, Wu R, Li S, Peter R, Shannar A, Chou PJ, Liu X, Liu Z, Kuo HD, Kong AN. Epigenetics/epigenomics and prevention of early stages of cancer by isothiocyanates. Cancer Prev Res (Phila). 2020. Epub 2020/10/16. doi: 10.1158/1940-6207.Capr-20-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ho E, Clarke JD, Dashwood RH. Dietary sulforaphane, a histone deacetylase inhibitor for cancer prevention. J Nutr. 2009;139(12):2393–6. Epub 2009/10/09. doi: 10.3945/jn.109.113332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Li S, Chen M, Li Y, Tollefsbol TO. Prenatal epigenetics diets play protective roles against environmental pollution. Clin Epigenetics. 2019;11(1):82. Epub 2019/05/18. doi: 10.1186/s13148-019-0659-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gianfredi V, Vannini S, Moretti M, Villarini M, Bragazzi NL, Izzotti A, Nucci D. Sulforaphane and Epigallocatechin Gallate Restore Estrogen Receptor Expression by Modulating Epigenetic Events in the Breast Cancer Cell Line MDA-MB-231: A Systematic Review and Meta-Analysis. J Nutrigenet Nutrigenomics. 2017;10(3–4):126–35. Epub 2017/10/19. doi: 10.1159/000480636. [DOI] [PubMed] [Google Scholar]
- 104.Li S, Kuo H-C, Yin R, Wu R, Liu X, Wang L, Hudlikar R, Peter R, Kong A-N. Epigenetics/Epigenomics of Triterpenoids in Cancer Prevention and in Health. Biochemical Pharmacology. 2020;175:113890. doi: 10.1016/j.bcp.2020.113890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kim H, Ramirez CN, Su ZY, Kong AN. Epigenetic modifications of triterpenoid ursolic acid in activating Nrf2 and blocking cellular transformation of mouse epidermal cells. J Nutr Biochem. 2016;33:54–62. Epub 2016/06/05. doi: 10.1016/j.jnutbio.2015.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zhang C, Wang C, Li W, Wu R, Guo Y, Cheng D, Yang Y, Androulakis IP, Kong AN. Pharmacokinetics and Pharmacodynamics of the Triterpenoid Ursolic Acid in Regulating the Antioxidant, Anti-inflammatory, and Epigenetic Gene Responses in Rat Leukocytes. Mol Pharm. 2017;14(11):3709–17. Epub 2017/10/17. doi: 10.1021/acs.molpharmaceut.7b00469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yang Y, Yin R, Wu R, Ramirez CN, Sargsyan D, Li S, Wang L, Cheng D, Wang C, Hudlikar R, Kuo HC, Lu Y, Kong AN. DNA methylome and transcriptome alterations and cancer prevention by triterpenoid ursolic acid in UVB-induced skin tumor in mice. Molecular carcinogenesis. 2019;58(10):1738–53. Epub 2019/06/27. doi: 10.1002/mc.23046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ramirez CN, Li W, Zhang C, Wu R, Su S, Wang C, Gao L, Yin R, Kong AN. In Vitro-In Vivo Dose Response of Ursolic Acid, Sulforaphane, PEITC, and Curcumin in Cancer Prevention. The AAPS journal. 2017;20(1):19. Epub 2017/12/22. doi: 10.1208/s12248-017-0177-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Man SM. Inflammasomes in the gastrointestinal tract: infection, cancer and gut microbiota homeostasis. Nat Rev Gastroenterol Hepatol. 2018;15(12):721–37. Epub 2018/09/07. doi: 10.1038/s41575-018-0054-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human Gut Microbiota and Gastrointestinal Cancer. Genomics Proteomics Bioinformatics. 2018;16(1):33–49. Epub 2018/02/24. doi: 10.1016/j.gpb.2017.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Brennan CA, Garrett WS. Gut Microbiota, Inflammation, and Colorectal Cancer. Annu Rev Microbiol. 2016;70:395–411. Epub 2016/09/09. doi: 10.1146/annurev-micro-102215-095513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Dingeo G, Brito A, Samouda H, Iddir M, La Frano MR, Bohn T. Phytochemicals as modifiers of gut microbial communities. Food Funct. 2020;11(10):8444–71. Epub 2020/10/01. doi: 10.1039/d0fo01483d. [DOI] [PubMed] [Google Scholar]
- 113.Castro-Barquero S, Lamuela-Raventos RM, Domenech M, Estruch R. Relationship between Mediterranean Dietary Polyphenol Intake and Obesity. Nutrients. 2018;10(10). Epub 2018/10/20. doi: 10.3390/nu10101523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Lyu Y, Wu L, Wang F, Shen X, Lin D. Carotenoid supplementation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis. Exp Biol Med (Maywood). 2018;243(7):613–20. Epub 2018/03/15. doi: 10.1177/1535370218763760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jyonouchi H, Sun S, Gross M. Effect of carotenoids on in vitro immunoglobulin production by human peripheral blood mononuclear cells: astaxanthin, a carotenoid without vitamin A activity, enhances in vitro immunoglobulin production in response to a T-dependent stimulant and antigen. Nutr Cancer. 1995;23(2):171–83. Epub 1995/01/01. doi: 10.1080/01635589509514373. [DOI] [PubMed] [Google Scholar]
- 116.Davis-Richardson AG, Triplett EW. A model for the role of gut bacteria in the development of autoimmunity for type 1 diabetes. Diabetologia. 2015;58(7):1386–93. Epub 2015/05/10. doi: 10.1007/s00125-015-3614-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Al Theyab A, Almutairi T, Al-Suwaidi AM, Bendriss G, McVeigh C, Chaari A. Epigenetic Effects of Gut Metabolites: Exploring the Path of Dietary Prevention of Type 1 Diabetes. Front Nutr. 2020;7:563605. Epub 2020/10/20. doi: 10.3389/fnut.2020.563605. [DOI] [PMC free article] [PubMed] [Google Scholar]


