Next Article in Journal
Cloning and Characterization of a Thermostable Endolysin of Bacteriophage TP-84 as a Potential Disinfectant and Biofilm-Removing Biological Agent
Next Article in Special Issue
Intercontinental Gut Microbiome Variances in IBD
Previous Article in Journal
Biodistribution and Cellular Internalization of Inactivated SARS-CoV-2 in Wild-Type Mice
Previous Article in Special Issue
The P2X7 Receptor Promotes Colorectal Inflammation and Tumorigenesis by Modulating Gut Microbiota and the Inflammasome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models

1
Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, 4000 Liege, Belgium
2
Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France
3
CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France
4
NGERE (Nutrition-Genetics and Exposure to Environmental Risks), National Institute of Health and Medical Research, University of Lorraine, F-54000 Nancy, France
5
Lung Cancer Epigenetics, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
6
International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Universidad de la Salud del Estado de Puebla, Puebla 72000, Mexico
7
Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, 20132 Milan, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(14), 7611; https://doi.org/10.3390/ijms23147611
Submission received: 31 May 2022 / Revised: 5 July 2022 / Accepted: 6 July 2022 / Published: 9 July 2022
(This article belongs to the Special Issue Molecular Advances in Inflammatory Bowel Diseases)

Abstract

:
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that encompass two main phenotypes, namely Crohn’s disease and ulcerative colitis. These conditions occur in genetically predisposed individuals in response to environmental factors. Epigenetics, acting by DNA methylation, post-translational histones modifications or by non-coding RNAs, could explain how the exposome (or all environmental influences over the life course, from conception to death) could influence the gene expression to contribute to intestinal inflammation. We performed a scoping search using Medline to identify all the elements of the exposome that may play a role in intestinal inflammation through epigenetic modifications, as well as the underlying mechanisms. The environmental factors epigenetically influencing the occurrence of intestinal inflammation are the maternal lifestyle (mainly diet, the occurrence of infection during pregnancy and smoking); breastfeeding; microbiota; diet (including a low-fiber diet, high-fat diet and deficiency in micronutrients); smoking habits, vitamin D and drugs (e.g., IBD treatments, antibiotics and probiotics). Influenced by both microbiota and diet, short-chain fatty acids are gut microbiota-derived metabolites resulting from the anaerobic fermentation of non-digestible dietary fibers, playing an epigenetically mediated role in the integrity of the epithelial barrier and in the defense against invading microorganisms. Although the impact of some environmental factors has been identified, the exposome-induced epimutations in IBD remain a largely underexplored field. How these environmental exposures induce epigenetic modifications (in terms of duration, frequency and the timing at which they occur) and how other environmental factors associated with IBD modulate epigenetics deserve to be further investigated.

1. Introduction

Inflammatory bowel diseases (IBD) are chronic relapsing-remitting inflammatory disorders of the gastrointestinal tract encompassing two main phenotypes: Crohn’s disease (CD) and ulcerative colitis (UC). The pathogenesis of IBD is not fully understood to date, but the most commonly accepted hypothesis is an inappropriate gut mucosal immune response towards the constituents of the gut microbiota, which cross an impaired epithelial barrier, in genetically predisposed individuals and under the influence of environmental factors [1]. Epidemiological studies (such as those carried out on monozygotic twins [2] and immigrants [3]), as well as the increase over time of the CD and UC incidence and prevalence (while the human gene pool is the same as before) [4], are all arguments that emphasize the importance of environmental factors in the occurrence of these inflammatory diseases. Epigenetics is a branch of life science that studies mechanisms regulating DNA-dependent processes (e.g., transcription, replication, recombination, repair, etc.) without primarily involving the nucleotide sequence of the DNA but, rather, the structure of how DNA is packed in the cell nucleus (chromatin structure), which can be inherited by daughter cells after cell division. Epigenetic mechanisms, including DNA methylation, post-translational histones modifications and non-coding ribonucleic acids (ncRNAs) [5], [6], regulating gene expression provide plausible explanations for the influence of the environment on gene expression profiles that favor intestinal inflammation [7]. Supporting this line of ideas, DNA methylation profiles observed in older monozygous twins with different environmental histories shows that epigenetic imprinting occurs mainly during crucial periods of development, whereas epigenomic changes can also occur day after day and accumulate over time in response to the exposome [8,9,10].
The term exposome has been proposed to encompass all environmental influences over the life course, from conception to death, that may influence disease emergence and clinical outcomes [11,12]. External environmental factors influencing the occurrence of IBD include the maternal lifestyle and in utero events [13], breastfeeding [14], diet [7], smoking habits [15,16], drugs [16,17,18], physical activity [16], stress [19], appendicectomy [16], vitamin D/UV exposure [16], infections [20] and hygiene [21]. While it is possible that these different factors directly induce epigenetic changes in the host, it is also possible that they influence the microbiome, an internal component of the exposome, and contribute to the occurrence of IBD through the exposome–microbiome–epigenome axis [22].
The impact of exposomes on the epigenetics in IBD has been poorly studied and is probably underestimated. This review aims to identify all the elements of the exposome that may play a role in intestinal inflammation through epigenetic modifications, as well as the underlying mechanisms that may contribute to IBD pathophysiology.

2. Epigenetics in IBD

Epigenetic mechanisms of gene expressions are involved in the intestinal epithelium homeostasis and in the development and differentiation of the immune cells, as well as in the modulation of responses generated by the immune system to defend against potential pathogens [23]. These epigenetic changes are reversible [24]. The genomic DNA in the eukaryotic cell nucleus is organized into chromatin. Chromatin consists of nucleic acids (genomic DNA and different types of RNAs); histone proteins (H2A, H2B, H3, H4 and H1) and non-histone chromatin-associated proteins [5,25,26]. Nucleosomes constitute the functional and structural units of chromatin. A nucleosome is built by around 146 bp of genomic DNA surrounding a histone octamer, which consists of two H2A–H2B dimers and one (H3–H4)2 tetramer [27,28]. In other words, chromatin is the physiological template for all DNA-dependent biological processes, including transcription. This fact increases the complexity of transcription regulation, since it implies that the chromatin structure has to be dynamic to grant or block access of transcription regulators to their respective binding elements on the DNA and to the transcription machinery to the genomic information in the nucleotide sequence. The epigenetic mechanisms of transcriptional regulation involve DNA methylation, histone modifications, nucleosome remodeling, interaction with the nuclear matrix and regulation via long non-coding RNAs (lncRNAs) and micro RNAs (miR) [26,29,30,31]. These mechanisms of transcription regulation establish cell-specific, heritable patterns of differential gene expression and silencing from the same genome and allow the cells to change these gene expression signatures in response to stimuli, such as changing conditions due to their environment [32,33].
DNA methylation in eukaryotes refers to the covalent transfer of a methyl group (-CH3) to the carbon atom at position 5 of cytosine forming 5-methylcytosine (5mC), most frequently at the dinucleotide sequence CG (mCG) [31,34,35,36,37]. DNA regions that are ≥200 bp long and show a CG:GC ratio ≥ 0.6 are defined as a CpG island [38]. The presence of DNA methylation prevents transcription factors from reaching gene promoters and generally leads to gene silencing [39,40]. DNA methylation in eukaryotes is catalyzed by DNA methyltransferases (DNMTs): DNMT1, which maintains DNA methylation patterns (during DNA replication and cell division), and DNMT3A/3B, which are responsible for de novo methylating DNMTs (during development or differentiation) [41,42,43]. These enzymes transfer methyl groups from S-adenosyl-L-methionine (SAM) to the cytosine residues in DNA [44]. On the contrary, DNA demethylation is mediated by the ten-eleven translocation (TET) enzymes, which add a hydroxyl group onto the methyl group of 5mC to form 5hmC (5-hydroxymethyl cytosine) [45]. Compared to healthy subjects, IBD patients show DNA methylation changes both at the cell (mainly immune) level and at the tissue level [46,47,48,49,50,51,52]. These changes also differ between UC and CD patients and involve several loci responsible for the regulation of immune responses [46,47,48,49,50,51,52].
Another epigenetic mechanism of transcriptional regulation involves post-translational modifications of histone proteins (further referred to as histone modifications). Histone proteins (H1, H2A, H2B H3 and H4) are relatively small and basic proteins that are abundant in the cell nucleus and are an essential part of the nucleosome, as described above. Due to structural characteristics of the nucleosome, histone proteins can undergo post-translational modifications at their N-terminal tails, which include acetylation, methylation, phosphorylation, ubiquitination and sumoylation, among others [53,54,55,56]. While DNA methylation is relatively stable in somatic cells, histone modifications are more diverse and dynamic, changing rapidly during the course of the cell cycle [6,30,53,54]. Acetylation at specific amino acids of histones (e.g., histone 3 lysine 9 acetylation; H3K9Ac) is generally associated with active chromatin and is mediated by histone acetyltransferases (HAT) and removed by histone deacetylases (HDAC). Histone methylation also occurs at specific amino acids of histone proteins and can be associated with both the repression (e.g., H3 lysine 27 trimethylation; H3K27me3) and activation (e.g., H3 lysine 4 trimethylation; H3K4me3) of gene expressions. There is a variety of enzymes mediating histone methylation (histone methyltransferases; HMT) and histone demethylation [57,58]. Similarly, the reactions leading to other histone modifications are catalyzed by a broad spectrum of enzymes in a regulated manner. Several environmental agents induce changes in histone modifications, thereby leading to changes in gene expression signatures.
In addition to these mechanisms, epigenetic regulation can also involve ncRNA, which are RNAs not translated into proteins, including miRs and lncRNAs. If miRs have a length of 18–25 nucleotides, lncRNAs are over 200 bases long [59]. These nucleic acid molecules can regulate gene expressions by interfering with messenger RNA (mRNA) translations by degrading them or through interactions with protein complexes involved in the regulation of gene expression [59,60]. The ncRNAs are differentially expressed between the control and IBD subjects, and there is also a difference in expression between CD and UC patients [61,62,63]. In IBD, miRs are involved in the regulation of the intestinal mucosal barrier, T-cell differentiation, the Th17 signaling pathway and autophagy [63]. In UC patients, miR-21, miR-16 and let-7 expressions are significantly increased in inflamed mucosa, while miR-192, miR-375 and miR-422b expressions are significantly reduced [61]. In CD patients, miR-23b, miR-106 and miR-191 are significantly increased in the inflamed mucosa, while miR-19b and miR-629 expressions are significantly decreased [61].
All these epigenetic mechanisms contribute to the development, progression and maintenance of IBD. They are usually triggered by a range of environmental factors. Some authors have mentioned three critical periods during which the environment can favor the onset of the disease: (1) during the prenatal period (in response to the maternal lifestyle), (2) in the early postnatal period (during gut microbiota colonization) and (3) just before the disease onset [64]. This review aims to study the impact of the exposome on the epigenome in IBD.

3. Methods

To identify exposome elements that could impact the epigenetics of IBD, we performed a scoping search using Medline. We used the following Medical Subject Heading (MeSH) terms (‘epigenetics’ OR ‘epigenomics’ OR ‘DNA methylation’ OR ‘histone(s)’ OR ‘short noncoding RNA’ OR ‘long noncoding RNAs’ OR ‘microRNA’ OR ‘miR’ OR “miRNA”) AND (“Inflammatory bowel disease” OR “IBD” OR “intestinal inflammation” OR “Crohn’s disease” OR “ulcerative colitis” OR “colitis”). Secondary references of the retrieved articles were reviewed to identify publications not captured by the electronic search. We excluded articles not written in English and those related to colitis-associated cancer.

4. Results

4.1. Parental Exposition

Accumulating evidence has pointed out that in utero environmental exposure can influence the epigenetic programming of the offspring and have an impact on its fate, conditioning its health status or, on the contrary, its lifelong risk of inflammatory conditions [65,66,67,68]. This is explained by the fact that the occurrence of an epimutation in a stem cell during embryonic development is transmitted to all their daughter cells and affects many more cells than those occurring in adult stem and/or somatic cells during postnatal development [69]. These epigenetic changes can not only be transmitted during successive division but also are passed on from generation to generation, some authors mentioning a real transgenerational epigenetic inheritance [70,71,72,73,74,75,76,77,78,79,80]. These prenatal environmental induced-epigenetic modifications could therefore contribute to the IBD epidemic not only by contributing to this condition but also by passing on modifications to subsequent generations, contributing to familial IBD predisposition, as illustrated by immigration studies [64,81,82,83,84,85].
There are few data on prenatal epigenetic plasticity in response to the environment in intestinal inflammation [64]. Some data suggest that this epigenomic reprogramming occurs in response to maternal diet modifications, and an excess of prenatal micronutrients (i.e., methyl donors routinely incorporated into prenatal supplements, such as folate, methionine, betaine and vitamin B12) in the maternal diet could confer an increased risk of colitis in the offspring [73]. The occurrence of maternal infection during pregnancy could also lead to the production of IL-6, known to induce epigenetic changes in fetal intestinal epithelial stem cells, which could induce long-lasting impacts on intestinal immune homeostasis and a predisposition toward inflammatory disorders [86]. In addition to diet and infections, maternal smoking during pregnancy could also have an impact on the risk of developing IBD [87]. A study of the impact of prenatal maternal smoking on the offspring’s DNA methylation has made it possible to highlight 69 differentially methylated CpGs in 36 genomic regions, among which four CpG sites were associated with an increased risk of IBD [87]. Maternal smoking induced persistent alterations in DNA methylation (rather, global hypomethylation [88,89,90,91,92]) but also miR dysregulation in the exposed offspring, changes that can be transmitted to the next generation [90,93,94,95,96,97,98]. Taken together, these data suggest that these maternal influences during prenatal development can induce epigenetic changes in the offspring, sometimes considered by some authors as the first step towards IBD development (by introducing a permanent change in the disease-relevant cell types) [64,99,100].

4.2. Microbiota

Occurring in this predisposing environment, a microorganism’s gut colonization during the first hours of life can be considered as the second step toward the occurrence of IBD [64,99,100]. Influenced by the mode of delivery, the presence or absence of breastfeeding and early environmental exposure, the early-life gut microbiota sets trajectories for health or IBD [101,102]. This newly formed microbiome will modulate until the age of 3 years to reach a globally largely similar taxonomic composition as in adults and will act as an epigenetic modulator, modifying the epimutations induced in the prenatal period. Breastfeeding and early bacterial colonization appear to play an important role in DNA methylation in intestinal epithelial stem cells and to condition the lifelong gut health [103].
The microbiome can induce epigenetic changes both in the intestinal epithelium and in immune cells (Table 1). Comparing the epigenomes of germ-free mice or antibiotic-treated mice to conventional mice, it appears that this microbiome can influence the host epigenetics through changes in DNA methylation, histone modifications and, also, through ncRNAs [104,105,106,107,108]. Species belonging to Firmicutes (especially Faecalibacterium prausnitzii and Roseburia species [109]) and Bacteroides genera, known to be reduced in IBD [110], have an epigenetically mediated anti-inflammatory action (HDAC inhibition) via the production of short-chain fatty acids (SCFAs) (the role of these will be discussed in more detail below) [111]. The commensal flora can also affect the bioavailability of methyl groups through their production of folate and affects the host DNA methylation [112,113].
Some germs may also contribute to the occurrence of IBD through their epigenetic mechanisms. Adherent-invasive Escherichia coli (AIEC), commonly associated with CD [114], upregulates the levels of miR-30c and miR-130a in intestinal epithelial cells (IECs), which reduces the levels of ATG5 and ATG16L1 and inhibits autophagy, leading to increased numbers of intracellular AIEC and the inflammatory response [115]. In turn, AIEC-infected IECs secrete exosomes that can transfer these same miR to recipient IECs with the same consequences, promoting the invasion and proliferation of infected tissues [116]. In addition, AIEC triggers an excessive mucosal immune response against the gut microbiota via the let-7b/TLR4 miR signaling pathway [117]. Mycobacterium avium subspecies paratuberculosis (MAP), also known to be associated with IBD [118], induces miR-21 expression in infected macrophages and decreases their ability to eliminate the bacteria, thus contributing to intestinal inflammation [119].
Microbial components such as lipopolysaccharides (LPS) and flagellin may also induce host epigenetic changes. LPS (a major component of the Gram-negative bacteria outer membrane) contributes to the development of intestinal inflammation by promoting the activation of NF-κB (nuclear factor-kappa B) pathways and the cytokines released by the downregulation of miR-19b, miR-497 and miR-215 in IECs [120], monocyte/macrophage cells [121] and fibroblast cells [122], respectively. LPS can also increase the level of H19 lncRNA in IECs that bind to miR (miR-34a and let-7), inhibiting cell proliferation and, thus, impairing the intestinal epithelial barrier [20]. In contrast, the flagellins of some bacteria—in particular, Roseburia intestinalis (found in a reduced abundance in IBD patients)—have rather epigenetically mediated anti-inflammatory actions [123,124]. Flagellin inhibits the activation of the NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome and proptosis in macrophages via miR-223-3p [123] and induces a lncRNA (HIF1A-AS2) that inactivates the NF-κB/Jnk (c-Jun N-terminal kinase) pathway [124].
Table 1. Impact of the microbiota on the epigenome in intestinal inflammation. AIEC, adherent-invasive Escherichia coli; CpG, cytosine–phosphate–guanine; DCs, dendritic cells; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; ETBF, Enterotoxigenic Bacteroides fragilis; HDAC, histone deacetylases; IECs, intestinal epithelial cells; IL, interleukin; KO, knockout; LPS, lipopolysaccharide; lncRNAs, long non-coding RNAs; MAP, Mycobacterium avium subspecies paratuberculosis; miR, micro-RNA; NF-κB, nuclear factor-kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; NOD2, nucleotide-binding oligomerization domain 2; PSC, primary sclerosing cholangitis; STAT, signal transducer and activator of transcription; TLR, toll-like receptor; TNBS, 2,4,6-trinitrobenzenesulfonic acid; UC, ulcerative colitis; WT, wild-type; ↑, increase; ↓ decrease.
Table 1. Impact of the microbiota on the epigenome in intestinal inflammation. AIEC, adherent-invasive Escherichia coli; CpG, cytosine–phosphate–guanine; DCs, dendritic cells; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; ETBF, Enterotoxigenic Bacteroides fragilis; HDAC, histone deacetylases; IECs, intestinal epithelial cells; IL, interleukin; KO, knockout; LPS, lipopolysaccharide; lncRNAs, long non-coding RNAs; MAP, Mycobacterium avium subspecies paratuberculosis; miR, micro-RNA; NF-κB, nuclear factor-kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; NOD2, nucleotide-binding oligomerization domain 2; PSC, primary sclerosing cholangitis; STAT, signal transducer and activator of transcription; TLR, toll-like receptor; TNBS, 2,4,6-trinitrobenzenesulfonic acid; UC, ulcerative colitis; WT, wild-type; ↑, increase; ↓ decrease.
GermActivityEpigenetic MechanismTissue/CellsMechanismModelAuthor
Commensal bacteria
Commensal bacteriaAnti-inflammatory activitymiR-10aDCsNegatively regulates host miR-10a expression, which contribute to the intestinal homeostasis maintenance by targeting IL-12/IL-23p40 expressionC57BL/6 (B6) miceXue X, et al. (2011) [125]
Commensal floraProinflammatory activitymiR-107DCs and macrophagesDownregulates miR-107 expression, known to represses the expression of IL-23p19, thereby favouring IL-23 expressionIECs, lamina propria CD11c+ myeloid cells including dendritic cells and macrophages, and T cells; DSS-induced colitis in miceXue X, et al. (2014) [126]
Commensal bacteriaAnti-inflammatory activitymiR-10aDCsInhibits human DCs miR-10a expression, which downregulates mucosal inflammatory response through inhibition of IL-12/IL-23p40 and NOD2 expression, and blockade of Th1/Th17 cell immune responsesHuman monocyte-derived dendritic cellsWu W, et al. (2015) [127]
Commensal microbiome-dependent (Bacteroides acidifaciens and Lactobacillus johnsoniiAnti-inflammatory activitymiR-21-5pIECsCommensal microbiome-dependent miR-21-5p expression in IECs regulates intestinal epithelial permeability via ADP Ribosylation Factor 4 (ARF4)HT-29 and Caco-2 cellsNakata K., et al. (2017) [128]
Cluster(s)
of Bacteroidetes and of protective Firmicutes and ClostridiaAnti-inflammatory activitymiR-21Colonic mucosaeLeads to miR-21 reduction, known to influence the pathogenesis of intestinal inflammation by causing propagation of a disrupted gut microbiotaWT and miR-21−/− miceJohnston DGW, et al. (2018) [129]
Cluster enriched in Bacteroides fragilis-DNA methylationIntestinal mucosaInduces 33 and 19 significantly hyper-methylated or hypomethylated sites, including hyper-methylated signals in the gene body of Notch Receptor 4 (NOTCH4)50 CD; 80 UC; 31 controlsRyan FJ, et al. (2020) [130]
Cluster enriched in Escherichia/Shigella/Klebsiella and Ruminococcus gnavusProinflammatory activityDNA methylationIntestinal mucosaLarger number of differentially methylated CpG sites (131 hyper- and 475 hypomethylated), including hypomethylation in CCDC88B (recently correlated with risk of CD) and Transporter 2 (TAP2), involved in genetic heterogeneity of CD
Cluster enriched in B. vulgatus-DNA methylationIntestinal mucosaInduces 23 hyper- and 18 hypomethylated sites, significant hyper-methylation was observed in the gene body of DNA Damage Regulated Autophagy Modulator 1 (DRAM1)
Specific germ
Adherent-invasive Escherichia coli (AIEC)Proinflammatory activitymiR-30c and miR-130aIECsUpregulates levels of miR-30c and miR-130a in IECs (by activating NF-κB), reducing the levels of ATG5 and ATG16L1 and inhibiting autophagy, leading to increased numbers of intracellular AIEC and an increased inflammatory responseCultured IECs and mouse enterocytesNguyen HT, et al. (2014) [115]
AIECProinflammatory activitylet-7bIECsInstigates excessive mucosal immune response against gut microbiota via miR let-7b/TLR4 signaling pathwayWT and IL-10 KO mice; T84 cellsGuo Z, et al. (2018) [117]
AIECProinflammatory activitymiR-30c and miR-130aIECsAIEC-infected IECs secretes exosomes that can transfer specific miRs (miR-30c and miR-130a) to recipient IECs, inhibiting autophagy-mediated clearance of intracellular AIECT84 cellsLarabi A, et al. (2020) [116]
Mycobacterium avium subspecies paratuberculosis (MAP)Proinflammatory activitymiR-21MacrophagesMAP upregulates miR-21 in macrophages, a change that results in diminished macrophages clearance ability and favours pathogens survival within the cellsTHP-1 cellsMostoufi-Afshar S, et al. (2018) [119]
Lactobacillus rhamnosus GGAnti-inflammatory activitymiR-146a and miR-155DCsInduces a significant downregulation of miR-146a expression, a negative regulator of immune response, and upupregulation of on miR-155Cultured DCsGiahi L., et al. (2012)
Lactobacillus acidophilusAnti-inflammatory activitymiRsColonic mucosaeL. acidophilus induce miRs expressionDSS-induced colitis in miceKim WK, et al. (2021) [131]
Faecalibacterium prausnitziiAnti-inflammatory activityHDAC1 inhibitionT cellsInhibits HDAC1, promotes Foxp3 and blocks the IL-6/STAT3/IL-17 downstream pathway contributing to the maintain of Th17/Treg balanceIBD patients (n = 9) and healthy control (n = 6); DSS-induced colitis in miceZhou L, et al. (2018) [132]
Faecalibacterium prausnitziiAnti-inflammatory activityHDAC3 inhibitionT cellsProduces butyrate to decrease Th17 differentiation and attenuate colitis through inhibiting HDAC3 and c-Myc-related metabolism in T cellsIBD patients; TNBS-induced colitis in miceZhang M, et al. (2019) [133]
Trichinella spiralisAnti-inflammatory activitymiRsT cellsExtra-vesicles-derived miR are involved in the regulation of the host immune response, including inflammation, including increase of Th2 and Treg cellsTNBS-induced colitis in miceYang Y, et al. (2020) [134]
Enterotoxigenic Bacteroides fragilis (ETBF)Proinflammatory activitymiR-149-3pT cellsDownregulates miR-149-3p, which play a role in modulation of T-helper type 17 cell differentiation (with increased number of T-helper type 17 cell contributing to intestinal inflammation)ETBF cellsCao Y, et al. (2021) [135]
Bacterial component
Roseburia intestinalis-derived flagellinAnti-inflammatory activitylncRNAIECsFlagellin induces p38-stat1 activation, activated HIF1A-AS2 promotor, induced HIF1A-AS2 (a lncRNA) expression in gut epithelium in a dose- and time-dependent manner. HIF1A-AS2 inactivates NF-κB/Jnk pathway and thus inhibits inflammatory responsesDSS/Flagellin-challenged mice; Caco-2 cellsQuan Y, et al. (2018) [124]
Roseburia intestinalis-derived flagellinAnti-inflammatory activitymiR-223-3pMacrophagesFlagellin inhibited activation of the NLRP3 inflammasome and pyroptosis via miR-223-3p/NLRP3 signaling in macrophagesDSS-induced colitis model in C57Bl/6 mice and the LPS/ATP-induced THP-1 macrophagesWu X, et al. (2020) [123]
LPSProinflammatory activityH3K4me1, H3K4me3, and H3K27ac histoneMacrophagesIncreases H3K4me1, H3K4me3, and H3K27ac histone marks, particularly in genes associated with an inflammatory response such as IL-12a and IL-18IL-10-deficient (Il10(−/−)) miceSimon JM, et al. (2016) [136]
LPS and flagellinAnti-inflammatory activitymiR-146IECsStimulate miR-146a overexpression in IECs, induces immune tolerance, inhibiting cytokine production (MCP-1 and GROα/IL-8)TNBS and DSS-induced colitis in miceAnzola A, et al. (2018) [137]
LPSProinflammatory activitylncRNA H19IECsIncreases levels of H19 lncRNA in epithelial cells in the intestine. H19 lncRNA bound to p53 and miR (miR-34a and let-7) that inhibit cell proliferation (alters regeneration of the epithelium)Intestinal tissues of UC patients and miceGeng H, et al. (2018) [138]
LPSProinflammatory activitymiR-19bIECsLPS significantly induces cell inflammatory injury, downregulated miR-19b expression and activates NF-κB and PI3K/AKT pathwayCaco2 cellsQiao CX, et al. (2018) [120]
LPSProinflammatory activitylncRNAMonocytes/macrophagesLPS promotes a downregulation of the lncRNA growth arrest-specific transcript 5 (GAS5), could mediate tissue damage by modulating the expression of matrix metalloproteinasesIBD patients (n = 25)Lucafò M, et al. (2019) [139]
LPSProinflammatory activitymiR-215FibroblastsLPS upregulates the expression of miR-215, increases oxidative stress in LPS-treated intestinal fibroblast by downregulating GDF11 (Growth differentiation factor 11) expression and activating the TLR4/NF-κB and JNK/p38 signaling pathwaysCCD-18Co cellsSun B, et al. (2020) [122]
LPSProinflammatory activitymiR-506 and DNMT1 modificationIECsLPS inhibits miR-506, leading to reduced expression of anion exchange protein 2 and inositol-1,4,5-trisphosphate-receptor but was accompanied by a substantial increase in DNMT1 and SPHK1 (sphingosine kinase 1) expression. The enhanced levels of kinase SPHK1 resulte in upregulation of bioactive sphingosine-1-phosphate (S1P) which led to further activation of S1P-dependent signaling pathways. The net effect of these responses is severe inflammationPatients with PSC, PSC with concurrent UC (PSC  +  UC), UC alone, and healthy controls (n  =  10 each); Caco2 cellsKempinska-Podhorodecka A, et al. (2021) [140]
LPSProinflammatory activitymiR-497MacrophagesReduces miR-497, promotes the activation of NF-κB pathway and the release of cytokinesIBD patients, mice with colitis and LPS-treated RAW264.7 cellsZhang M, et al. (2021) [121]
Although the taxonomic composition of the microbiota is stable at year 3, its composition can be influenced by a range of other environmental factors (including dietary habits, smoking and drugs, as discussed below), which may be responsible for the third step towards the occurrence of IBD [64].

4.3. Gut Microbiota-Derived Metabolites

Influenced by both the microbiota and diet, SCFAs are gut microbiota-derived metabolites that result from the anaerobic fermentation of nondigestible dietary fibers (found in fruits and vegetables). Acetate, butyrate and propionate, the three principal SCFAs, exert an anti-inflammatory role and promote the integrity of the epithelial barrier functions partly via the epigenetic pathways (Table 2) [141,142]. Among the SCFAs, butyrate is the most studied one. By inhibiting, in a reversible way, HDACs [143,144], cells exposed to butyrate present higher acetylation at specific lysine residues in histones, resulting in increased transcription of genes in both intestinal epithelial and immune cells [145]. The inhibition of HDAC in cells contributes to the reduction of inflammation by (1) the induction of IκBα expression, with a subsequent inhibition of the NF-κB pathway, (2) the inhibition of the IFN-γ/STAT1 (signal transducer and activator of transcription) signaling pathway and (3) the activation of the anti-inflammatory function of PPARγ (peroxisome proliferator-activated receptor γ) [145]. Butyrate also has more specific epigenetic actions on certain cell types. At the epithelial level, butyrate plays a role in the integrity of the epithelial barrier (by restoring tight junction proteins [146]) and the defense against the invading microorganisms (via a nucleotide-binding oligomerization domain 2 (NOD2)-dependent pathway or via autophagy [147]). Butyrate also has an effect on various immune cells, such as (1) monocytes/macrophages (in which it induces monocyte-to-macrophage differentiation, promotes their antimicrobial activity through inhibition of HDAC3 [148], reduces the production of their inflammatory mediators [149] and induces the polarization of M2 macrophages [150]); (2) T cells (promotes Treg [151] and inhibits Th17 cell development [151]); (3) neutrophils (in which HDAC inhibition leads to proinflammatory cytokine reduction [152]) and (4) dendritic cells (inhibit IL-12 [153]). The epigenetic role of propionate and acetate has been less studied. Propionate promotes epithelial cell migration and contributes to intestinal epithelial restitution, a complex process important for tissue regeneration in IBD [142].

4.4. Diet

Next, compared to a low-fiber diet [163], impacting the level of these SCFAs [163], other diets have been shown to induce epigenetic changes related to IBD (Table 3). Regarding the literature, elements of the Western diet, characterized by a low-fiber, low-fruit, low-vegetable and deficiency in micronutrients, as well a high-fat diet, may be associated with epigenetic changes in IBD. The Western diet has been shown to lead to a decrease in miR-143/145a, miR-148a and miR-152 in colonocytes with a consequent increase in ADAM17 (a disintegrin and metalloprotease 17) expression protein and colitis aggravation [164]. A low or deficient methyl diet can also contribute to intestinal inflammation by reducing SIRT1 (sirtuin 1) expression (a histone deacetylase), contributing to endoplasmic reticulum stress [165] and demethylating HIF-1-responsive elements (HRE), which leads to the abnormal gut expression of CEACAM6 (CEA Cell Adhesion Molecule 6), favoring AIEC colonization and subsequent inflammation [166]. Finally, it was shown that a high-fat diet can change the miR profile of the visceral adipose exosomes (switching the exosomes from an anti-inflammatory to a proinflammatory phenotype with an increase of miR-155, for example), predisposing the intestine to inflammation via promoting macrophage M1 polarization [167].
Polyphenols, found mainly in fruits and vegetables, are complex molecules produced by plants with antioxidant properties able to scavenge free radicals. Divided into flavonoids (such as alpinetin, fortunellin, baicalin, quercetin, berberine, cardamonin and lonicerin) [168,169,170,171,172,173,174] and non-flavonoids (such as resveratrol [175,176] and chlorogenic acid [177]), they reduce the risk of intestinal inflammation, mainly by modifying the miRs profile and inhibiting HDACs. Other foods have also been shown to influence host epigenetics and could potentially play a role in gut inflammation. Milk [178,179,180], common sweeteners [181], galacto-oligosaccharides [182], corn cobs [183], cinnamaldehyde (a major active compound from cinnamon) [184,185], limonin (a triterpenoid extracted from citrus) [186], ginger [187], ginseng [188] and black raspberries [189,190] have anti-inflammatory properties. In contrast, chronic alcohol exposure increases miR-122a and miR-155 expression in the intestine, which decreases occludins expression, leading to increased intestinal permeability and modulates cytokines and the T-cell immune response in the gut, leading to intestinal TNFα (tumor necrosis factor α) and NF-κB activation, respectively [191,192].
Table 3. Impact of the diet on the epigenome in intestinal inflammation. ADAM17, a disintegrin and metalloprotease-17; AIEC, Adherent-invasive Escherichia coli; CD, Crohn’s disease; CEACAM6, carcinoembryonic antigen-related cell adhesion molecule 6; CREB, C-AMP response element-binding protein; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; HDAC, histone deacetylase; HMGB1, high mobility group box 1; HRE, HIF-1-responsive elements; IBD, inflammatory bowel disease; ICAM, intercellular adhesion molecule; IEC, intestinal epithelial cell; IFN, interferon; IL, interleukin; lncRNA, long non-coding RNA; LPS, lipopolysaccharide; miR, microRNA; MMP9, Matrix Metallopeptidase 9; PBMC, peripheral blood mononuclear cell; PTEN, phosphatase and tensin homolog; NF-κB, nuclear factor-kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; PECAM, Platelet endothelial cell adhesion molecule; RECK, reversion-inducing cysteine-rich protein with Kazal motifs; SCFAs, short-chain fatty acids; STAT, signal transducer and activator of transcription; TGF, transforming growth factor; TNBS, 2,4,6-trinitrobenzenesulfonic acid; TNF, tumor necrosis factor; UC, ulcerative colitis; VCAM, vascular cell adhesion molecule 1; WT, wild-type; ZO, zonula occludens.
Table 3. Impact of the diet on the epigenome in intestinal inflammation. ADAM17, a disintegrin and metalloprotease-17; AIEC, Adherent-invasive Escherichia coli; CD, Crohn’s disease; CEACAM6, carcinoembryonic antigen-related cell adhesion molecule 6; CREB, C-AMP response element-binding protein; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; HDAC, histone deacetylase; HMGB1, high mobility group box 1; HRE, HIF-1-responsive elements; IBD, inflammatory bowel disease; ICAM, intercellular adhesion molecule; IEC, intestinal epithelial cell; IFN, interferon; IL, interleukin; lncRNA, long non-coding RNA; LPS, lipopolysaccharide; miR, microRNA; MMP9, Matrix Metallopeptidase 9; PBMC, peripheral blood mononuclear cell; PTEN, phosphatase and tensin homolog; NF-κB, nuclear factor-kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; PECAM, Platelet endothelial cell adhesion molecule; RECK, reversion-inducing cysteine-rich protein with Kazal motifs; SCFAs, short-chain fatty acids; STAT, signal transducer and activator of transcription; TGF, transforming growth factor; TNBS, 2,4,6-trinitrobenzenesulfonic acid; TNF, tumor necrosis factor; UC, ulcerative colitis; VCAM, vascular cell adhesion molecule 1; WT, wild-type; ZO, zonula occludens.
FoodActivityEpigenetic MechanismTissue/CellsMechanismModelAuthor
Diet
Western dietProinflammatory activitymiR-143, miR-145A, miR-148a, miR-152IECsLeads to a decrease in miR-143/145a, miR-148a and miR-152 in colonocytes with a consequent increase in ADAM17 expression protein (these miRs regulating ADAM17) and aggravates colitis.DSS-induced colitis in miceDougherty U, et al. (2021) [164]
High fat dietProinflammatory activitymiR-155Visceral adipocytesHigh fat diet changes the miR profile (among which miR-155) of the visceral adipose exosomes, switching the exosomes from anti-inflammatory to a proinflammatory phenotype.MacrophagesWei M, et al. (2020) [193]
High fat diet rich in n-6 linoleic acidProinflammatory activityDNA methylationColonic mucosaeEpigenetically modifies farnesoid-X-receptor (FXR), leading to the activation of downstream factors that participate in bile acid homeostasis and epigenetically activates prostaglandin-endoperoxide synthase-2 (Ptsg-2) coupled accumulation of c-JUN and proliferative cyclin D1(Ccnd1) and increase the risk of inflammationC57BL/6J mice; Human colonic foetal cellsRomagnolo DF, et al. (2019) [194]
Methyl-deficient dietProinflammatory activitySirtuin 1IECsReduces sirtuin 1 (SIRT1) expression level and promotes greater acetylation of (heat shock factor protein 1) HSF1, in relation with a dramatic decrease of chaperones (binding immunoglobulin protein (BIP), heat shock protein (HSP)27 and HSP90)DSS-induced colitis in mice; Caco-2 cellsMelhem H, et al. (2016) [165]
Low-methyl dietProinflammatory activityDNA methylationIECsLow-methyl diet-dependent HRE demethylation led to abnormal gut expression of CEACAM6 (carcinoembryonic antigen-related cell adhesion molecule 6), favouring AIEC colonisation and subsequent inflammationTransgenic mice; Caco-2, T-84 and sh-HIF1-α-T-84 cellsDenizot J, et al. (2015) [166]
Methyl-donnor supplemented diet (folate, B12 vitamin)Anti-inflammatory activityDNA methylationIECsMethyl-donor supplemented diet contributes to hypermethylation of CEACAM6 promoter in IECs, associated with a significant decrease in CEACAM6 expression contributing to less adherence of AIEC bacteria to the enterocytesCEABAC10 miceGimier E, et al. (2020) [195]
Isolated food
Cow’s milk (commercial)Anti-inflammatory activitymiR-21, miR-29b and miR-125bColonic mucosaeExtracellular vesicles (EVs) concentrated from commercial cow’s milk downregulates miR-21, miR-29b and miR-125b. MiR-125b was associated with a higher expression of the NF-κB inhibitor TNFAIP3 (A20)DSS-induced colitis in miceBenmoussa A, et al. (2019) [178]
Human milk derived exosomesAnti-inflammatory activitymiR-320, miR-375, and Let-7 and DNMT1 and DNMT3Colonic mucosaeMiR highly express in milk, such as miR-320, 375, and Let-7, were found to be more abundant in the colon of milk derived exosomes-treated mice compared with untreated mice. These miR downregulate their target genes, mainly DNA methyltransferase 1 (DNMT1) and DNMT3DSS-induced colitis in mice; PBMCReif S, et al. (2020) [179]
Dietary depletion of milk exosomes and their microRNA cargosProinflammatory activitymiR-200a-3pCecum mucosaeElicits a depletion of miR-200a-3p and elevated intestinal inflammation and chemokine (C-X-C Motif) ligand 9 expressionMdr1a−/− miceWu D, et al. (2019) [180]
Saccharin sodium, Stevioside, and Sucralose (three common sweeteners)Anti-inflammatory activitymiR-15bIECsUpregulate the expression of E-cadherin through the miR-15b/RECK/MMP-9 axis to improve intestinal barrier integrity. Saccharin exerts the most pronounced effect, followed by Stevioside and SucraloseDSS-induced colitis in miceZhang X, et al. (2022) [181]
Galacto-oligosaccharides (GOS)Anti-inflammatory activitymiR-19IECsGOS increases of cell viability, the decrease of apoptosis, as well as the suppressed release of TNF-α, IFN-γ and IL-1β by upregulating miR-19bHuman colon epithelial FHC cells; Helicobacter hepaticus induced colitis in ratsSun J, et al. (2019) [182]
Cinnamaldehyde (a major active compound from cinnamon)Anti-inflammatory activitymiR-21 and miR-155MacrophagesCinnamaldehyde inhibits NLRP3 inflammasome activation as well as miR-21 and miR-155 level in colon tissues and macrophage. The decrease in miR-21 and miR-155 suppresses levels of IL-1β and IL-6;DSS-induced colitis in mice; macrophage cell line RAW264.7 and human monocytes U937Qu S, et al. (2018) [185]
CinnamaldehydeAnti-inflammatory activitylncRNAs H19T cellsCinnamaldehyde inhibits Th17 cell differentiation by regulating the expression of lncRNA H19DSS-induced colitis in mice and naïve CD4+ T cellsQu SL, et al. (2021) [101]
Limonin (a triterpenoid extracted from citrus)Anti-inflammatory activitymiR-124IECsDownregulates p-STAT3/miR-214 signaling pathway and represses the productions of proinflammatory cytokines (such as TNF-α and IL-6)DSS-induced colitis in mice; cultured normal colonic epithelial cellsLiu S, et al. (2019) [186]
Edible gingerAnti-inflammatory activityContained around 125 miRNAsIECsIncreases the survival and proliferation of IECs, reduces the proinflammatory cytokines (such as TNF-α, IL-6 and IL-1β), and increases the anti-inflammatory cytokines (including IL-10 and IL-22) in colitisDSS-induced colitis in miceZhang M, et al. (2016) [187]
Ginsenoside Rh2 (active ingredient of ginseng)Anti-inflammatory activitymiR-124IECsInhibits IL-6-induced STAT3 phosphorylation and miR-214 expression (which is an inflammatory effector molecule acting through NF-κB-IL6 pathway)DSS-induced colitis in mice; cultured normal colonic epithelial cellsChen X, et al. (2021) [188]
Black raspberries (BRBs)Anti-inflammatory activityDemethylation the promoter of dkk3; correction of promoter hypermethylation of suppressor genesColonic mucosaeBRBs exert their anti-inflammatory effects is through decreasing NF-κB p65 expression leading to decrease of DNMT3B expression (but also histone deacetylases 1 and 2 (HDAC1 and HDAC2) and methyl-binding domain 2 or MBD2), which in turn reverse aberrant DNA methylation of tumor suppressor genes, e.g., dkk2, dkk3, in the Wnt pathway, resulting in their enhanced mRNA expression locally in colon and systematically in spleen and bone marrow and thus in decreased translocation of β-catenin to the nucleus prohibiting the activation of the pathwayDSS-induced colitis in mice; splenocytes and bone marrow cellsWang LS, et al. (2013) [189]
Black raspberriesAnti-inflammatory activityDemethylationColonic mucosaeBRBs decreas the methylation of wif1, sox17, and qki gene promoters and thus increase their mRNA expression (contributing to Wnt signaling)Interleukin-10 knockout miceWang LS, et al. (2013) [190]
MastihaAnti-inflammatory activitymiR-155T cellsPlays a role in circulating levels of miR-155, a critical player in T helper-17 (Th17) differentiation and functionUC patients (n = 35)Amerikanou C, et al. (2021) [196]
IsoliquiritigeninAnti-inflammatory activityHDACs inhibitionIECsSuppresses acetylated HMGB1 release via the induction of HDAC activity, which is one of the critical mediators of inflammation, which is actively secreted from inflammatory cytokine-stimulated immune or non-immune cellsHT-29 cellsChi JH, et al. (2017) [197]
Chronic ethanol exposureProinflammatory activitymiR-122aIECsIncreases the intestinal miR-122a expression, which decreased occludin (OCLN) expression leading to increased intestinal permeabilityHT-29 cellsChen Y, et al. (2013) [191]
Chronic alcohol feeding (but not acute alcohol binge)Proinflammatory activitymiR-155Intestinal tissueIncreases miR-155 in the small bowel, which is a modulator of cytokine and T-cell immune response in the gut, leading to intestinal TNFα, and NF-κB activationWT-miceLippai D, et al. (2014) [192]
Polyphenol
Polyphenolic red wine extractAnti-inflammatory activitymiR-126FibroblastsPolyphenolic red wine extract downregulates miR-126, leading to downregulation of NF-kB, ICAM-1, VCAM-1, and PECAM-1CCD-18Co myofibroblasts cellsAngel-Morales G, et al. (2012) [198]
Polyphenolic extracts from cowpea (Vigna unguiculata)Anti-inflammatory activitymiR-126FibroblastsCowpea may exert their anti-inflammatory activities at least in part through induction of miR-126 that then downregulate VCAM-1 mRNA and protein expressionsCCD-18Co myofibroblasts cellsOjwang LO, et al. (2015) [199]
Mango (Mangifera indica L.) polyphenolicsAnti-inflammatory activitymiR-126FibroblastsMango polyphenols attenuates inflammatory response by modulating the PI3K/AKT/mTOR pathway at least in part through upregulation of miR-126 expressionCCD-18Co cells; DSS-induced colitis in ratsKim H, et al. (2017) [200]
Baicalin (flavone)Anti-inflammatory activitymiR-191aIECsExerts a protective effect on IECs against TNF-α-induced injury, which is at least partly via inhibiting the expression of miR-191a, thus increasing ZO-1 expressionIEC-6 cellsWang L, et al. (2017) [170]
Pomegranate (Punica granatum L.) polyphenolicsAnti-inflammatory activitymiR-145MyofibroblastsPomegranate polyphenols attenuate colitis by modulating the miR-145/p70S6K/HIF1α axisDSS-induced colitis in rats; CCD-18Co colon-myofibroblastic cellsKim H, et al. (2017) [201]
Alpinetin, a flavonoid compound extracted from the seeds of Alpinia katsumadai HayataAnti-inflammatory activitymiR-302T cellsActivates Aryl hydrocarbon receptor (AhR), promoting expression of miR-302, downregulating expression of DNA methyltransferase 1 (DNMT-1), reducing methylation level of Foxp3 promoter region, facilitating combination of CREB and promoter region of Foxp3, and upregulating the expression of Foxp3. Alpinetin ameliorates colitis in mice by recovering Th17/Treg balance.DSS-induced colitis in miceLv Q, et al. (2018) [168]
Fortunellin, a citrus flavonoidAnti-inflammatory activitymiR-374aIECsFortunellin targets miR-374a, which is a negative regulator of PTEN, known to induce cell apoptosisTNBS-induced colitis in ratsXiong Y, et al. (2018) [169]
Quercetin (flavonoid)Anti-inflammatory activitymiR-369-3pDCsQuercetin-induced miR-369-3p which reduce C/EBP-β, TNF-α, and IL-6 productionLPS-stimulated DCsGalleggiante V, et al. (2019) [171]
Resveratrol (a natural plant product)Anti-inflammatory activitymiR-31, Let7a, miR-132T cellsResveratrol decreases the expression of several miRs (miR-31, Let7a, miR-132) that targets cytokines and transcription factors involved in anti-inflammatory T cell responses (Foxp3 and TGF-β). MiR-31 regulates the expression of Foxp3 with increase of CD4+ Foxp3+ regulatory T cells (Tregs)TNBS-induced colitis in miceAlrafas HR, et al. (2020) [176]
Resveratrol (an anti-oxidant)Anti-inflammatory activityHDACs inhibitionT cellsInhibits HDACs, increases anti-inflammatory CD4+ FOXP3+ (Tregs) and CD4+ IL10+ cells, and decreases proinflammatory Th1 and Th17 cellsAOM and DSS-induced colitis in miceAlrafas HR, et al. (2020) [175]
Chlorogenic acid (found in the coffee)Anti-inflammatory activitymiR-155MacrophagesDownregulates miR-155 expression, inactivates the NF-κB/NLRP3 inflammasome pathway in macrophages and prevent colitisDSS-induced colitis in mice; LPS/ATP-induced RAW264.7 cellsZeng J, et al. (2020) [177]
Lonicerin (constituant of herb Lonicera japonica Thunb.)Anti-inflammatory activityH3K27me3 modificationMacrophagesBinds to enhancer of zeste homolog 2 (EZH2) histone methyltransferase, which mediate modification of H3K27me3 and promotes the expression of autophagy-related protein 5, which in turn leads to enhanced autophagy and accelerates autolysosome-mediated NLRP3 degradationDSS-induced colitis in mice and isolated colonic macrophages and IECs; bone marrow-derived macrophagesLv Q, et al. (2021) [174]
Pristimerin (Pris), which is a natural triterpenoid compound extracted from the Celastraceae plantAnti-inflammatory activitymiR-155Colonic mucosaePris may reduce DSS-induced colitis in mice by inhibiting the expression of miR-155Blood and colon tissue of IBD patients; DSS-induced colitis in miceTian M, et al. (2021) [202]
Cardamonin is a naturally occurring chalcone (majorly from the Zingiberaceae family incluging a wide range of spices from India)Anti-inflammatory activityModulation of miR expressionMacrophagesCardamonin modulates miR expression, protects the mice from DSS-induced colitis, decreases the expression of iNOS, TNF-α, and IL-6, and inhibited NF-kB signaling which emphasizes the role of cardamonin as an anti-inflammatory moleculeRAW 264.7 Cells (monocyte/macrophage-like cells); DSS-induced colitis in miceJames S, et al. (2021) [172]
BerberineAnti-inflammatory activitymiR-103a-3pIECsRepresses Wnt/β-catenin pathway activation via modulating the miR-103a-3p/Bromodomain-containing protein 4 axis, thereby refraining pyroptosis and reducing the intestinal mucosal barrier defect induced via colitisDSS-induced colitis in mice; Caco-2 cells and human NCM460 cellsZhao X, et al. (2022) [173]

4.5. Smoking

Smoking habits are the single best-established environmental factor that influences the CD phenotype, behavior and response to therapy [203]. While nicotine is the most prominent component released during smoking (and therefore the best-studied), other chemical components could also induce epigenetic changes, including polycyclic aromatic hydrocarbons; heavy metals (nickel, cadmium, chromium and arsenic); carbon monoxide and reactive oxygen species [203]. Well-studied in lung diseases (but never in IBD, to our knowledge), smoking-induced epigenetic modifications seem to be strongly associated with smoking habits, the dose and the duration of smoke exposure [204,205,206,207,208,209]. The methylation of certain genetic loci, post-translational modifications of histones and the level of expressed miR may be reversible after smoking cessation (after 5 years, according to some studies) [93,204,205,206,207,208,209,210]. In contrast to these reversible epigenetic changes, others remain unchanged even after 30 years of smoking cessation, explaining that epigenetic modifications induced by smoking exposition confer long-term risks of adverse health outcomes but could also be transmitted to the next generation [93,204,207,208,209,210,211]. The mechanisms by which tobacco may contribute to inflammation are multiple and involve changes in the enzymes involved in DNA methylation, post-transcriptional histone modifications and ncRNAs [65,93,203,212,213,214].
Regarding smoking-induced DNA methylation, a meta-analysis performed by Joehanes and colleagues highlighted various genome-wide association studies showing that smoking-induced genes differentially methylated are enriched for variants associated with smoking-related diseases, including IBD, CD and UC [210,215,216]. The findings suggest that changes in methylation of the BCL3, FKBP5, AHRR and GPR15 genes are involved in the mechanism by which smoking increases the risk of CD [217,218].
Concerning smoking-induced histone modifications, smoking also contributes to histone hyperacetylation (H4 histone in active smoking and H3 histones in ex-smokers) by upregulating HATs and downregulating SIRT 1–7 (which belong to the family of class III HDACs) [65,219,220,221,222,223,224,225,226,227]. This imbalance, in favor of histones acetylation, contributes to the increased transcription of proinflammatory genes, mainly controlled by NF-κβ [65,219,221,224,228,229,230,231,232], and the increase of expression of proinflammatory mediators (including IL-1β, TNF-α and IL-6), contributing to chronic inflammation [228,229,233,234]. In a colitis model, Lo and colleagues showed that the reduction of SIRT2 could also be associated with a reduction of the M2-associated anti-inflammatory pathway [229]. The SIRT3 reduction is associated with less activation of the NALP3 inflammasome [235]. Cigarette smoke exposure upregulates the enzyme that catabolizes HMTs, leading to an increase of the H3 and H4 histone residue methylation [226], which may contribute to the proinflammatory cascade [236].
Smoking exposure also alters ncRNAs in a dose-and-time-dependent manner, high doses of and long-lasting exposure being necessary to induce irreversible ncRNA alterations, which may be involved in smoking-related diseases [237,238]. While there are no data on lncRNAs, the impact of smoking on miRs in IBD has been better studied. Interestingly, these IBD-induced epigenetic changes could partly explain why smoking is rather protective in UC, whereas it is an important risk factor in CD. Indeed, nicotine enhances the miR-124 expression, which targets and downregulates IL6R, resulting in a shifting Th1/Th2 balance toward Th1 (in peripheral blood lymphocytes and colon tissues), thereby protecting against Th2-type UC and worsening Th1-type CD [239]. This increase in miR-124 in epithelial cells, lymphocytes and macrophages in response to nicotine also results in the phosphorylation of STAT3, in a decreased production of IL-6 at the transcriptional level, and prevents the conversion of pro-TNF-α to TNF-α, which also explains the protective role of tobacco in the UC [240,241]. Tobacco also induces changes in several miRs that are functionally related to inflammation [65]. Among those highlighted in the IBDs are miR-21, miR-132, miR-195 and miR-223 [65]. MiR-21 (increased in the colon of IBD patients [242]) is known to increase the intestinal epithelial permeability (through an action on the tight junctions) [242,243,244] and plays a crucial role in T-cell differentiation, apoptosis and activation [242,245,246,247] and promotes the production of inflammatory cytokines (including TNF-α, IFN-γ and IL-1β) by immune cells, contributing to tissue inflammation and IBD pathogenesis [248,249,250]. The overexpression of a miR-195 precursor lowered the cellular levels of the Smad7 protein, leading to a decrease in c-Jun and p65 expression, and might contribute to the protective effect of tobacco in UC [251]. Lastly, smoking also downregulates miR-200 [252], known to repress epithelial-to-mesenchymal transition (or EMT), a process involved in intestinal fibrosis [242,252]. Consequently, the decrease of miR-200 in response to smoking could partly explain why smoking IBD patients are more likely to develop intestinal fibrosis (and fibrostenosis) [253,254,255].

4.6. Drugs

While the impact of NSAIDs (non-steroidal anti-inflammatory drugs) and oral contraceptives [256,257], whose long-term consumption is known to be associated with IBD, on the epigenome of IBD patients has apparently not yet been investigated, other molecules have a known impact (Table 4). Several treatments used in IBD exert their anti-inflammatory action via epigenetic modifications, such as 5ASA [258,259], anti-TNF [125,127,260,261,262,263,264], exclusive enteral nutrition [265] and mesenchymal stem cells [266,267]. Antibiotics [268,269,270] and probiotics [271,272,273,274,275,276] can also reduce gut inflammation through various epigenetic mechanisms. Finally, a range of Chinese herbs have been shown to have an epigenetically mediated anti-inflammatory action in the gut [277,278,279,280,281,282,283,284,285,286,287].

4.7. Vitamin D

Vitamin D is an environmental factor involved in IBD pathogenesis. Its deficiency, which can be both a cause and a consequence of IBD, is associated to an increased risk of disease activity, mucosal inflammation, clinical relapse and a lower quality of life [301]. A vitamin D-deficient diet contributes to IBD through the following epigenetic mechanisms: (1) increase in miR-142-3p expression in intestinal tissues leading to autophagy dysregulation [302]; (2) reduction of the interaction between VDR and HDAC11, an important complex for the maintenance of the epithelial barrier [303], and (3) the upregulation of miR-125b expression and reduction of M1 macrophage polarization to the M2 subtype [304].

4.8. Physical Activity

Despite all the known benefits of physical activity (PA) in IBD [16], the way in which it modifies epigenetics has never been studied to date to our knowledge. In other inflammatory diseases, the general view is that regular moderate-intensity physical activity could have an anti-inflammatory effect, while prolonged or high-intensity PA can trigger inflammation, both by leading to epigenetic changes that, in turn, regulate inflammatory responses in peripheral tissues [305,306,307]. These peripheral epigenetic changes appear to be largely induced by the muscle secretome, also known as “myokinome”, which corresponds to all the cytokines or proteins produced by the myocyte in response to muscular contractions [308,309]. These sport-induced epigenetic modulations (including both DNA methylations, histone modifications and miR modulations [309]) seem to vary according to the type of performed exercise (and the frequency, the intensity and the duration) [309]; the individual (in terms of age, gender and body composition) [305,309] and can vary from one tissue to another [310].
Studies suggest that PA is associated with DNA hypermethylation (although these results are not unanimous), contributing to a decreased expression of inflammation-related genes (such as the hypermethylation of IL-17A and IFN-γ promoter regions or TNF gene and the hypomethylation of IL-10) [311]. Exercise can also induce histones acetylation/deacetylation in a body mass index-dependent manner [312]. Finally, physical activity also leads to the release of a range of miRs from the muscle, known to play roles in macrophage polarization, dendritic cell activation, dendritic cell-mediated T-cell activation and the Th1 and Th17 differentiation of T cells, which are all pathophysiological processes involved in IBD [309].

5. Limitations to the Analysis of the Exposome Impact on the Epigenome in IBD

The study of the impact of the exposome on the epigenome is difficult because of the limitations of both the exposome study and the epigenome study. Human epidemiological studies are necessary to assess exposome-related epimutation. The first way to study the impact of the exposome is using retrospective case–control epidemiological studies (which compare the life of IBD patients with control cases to identify environmental influences based on surveys), but these studies are subject to a recall bias of past exposure factors. The second way of studying the exposome is a multi-omics approach (via the quantification and detection of external influences in a group of patients compared to a control group by different technologies), but unfortunately, it does not allow the detection of the factors responsible for the crucial pathophysiological changes explaining the occurrence of the pathology, it is very difficult to obtain a cause-and-effect relationship when using this method. A third approach, which counters this, consists of directly studying the supposed factors based on the basis of the pathophysiological hypotheses, but this exposes a selection bias and is not always easy to carry out in humans. Easier to perform on in vivo animal models or in vitro models, it does not reflect what is happening in humans [100,313].
All patients exposed to an environmental factor do not develop epimutation [314,315]. The onset of this and related diseases may depend on the duration, frequency and intensity of exposure to the environmental factor and the period of life during which it occurs [81]. Epigenetic changes can be influenced by age, sex and race but, also, possibly by the underlying host genotype [316,317,318]. Furthermore, the epigenetic changes induced by the exposome may vary according to the cell type and analyzed tissue (peripheral blood mononuclear cells, epithelial cells and biopsies) [316]. The tissue isolation and manipulation may also induce nonspecific epigenetic changes and mask the exposome-related one [100]. Finally, whatever the methodology used, it is always subject to several environmental factors at the same time that may induce concomitant and potentially interactive epigenetic changes, and the impact of an individual factor is not always easy to identify [319].

6. Conclusions and Challenge for the Future

This review is the first, to our knowledge, to study the impact of the exposome on the epigenome in IBD. Different elements of the exposome such as the maternal lifestyle, microbiota, diet, smoking, infection and vitamin D, as well as different drugs, may induce epigenetic changes related to IBD (Figure 1). Next to these factors, the impact of other environmental factors known to be involved in the pathophysiology of IBD on host epigenetics has not yet been studied. The influence of physical activity [16], appendicectomy in UC [16], processed and fast food or dietary [320,321] or psychologic stress/anxiety/depression [19], NSAIDs [256], oral contraception [257] or infections [20], as well as other factors potentially involved, deserve to be investigated. Regarding the identified environmental factors, how environmental exposure (in terms of the duration, frequency and timing at which it occurs) induces an epimutation, and whether this involves an exposome–microbiome–epigenome axis and becomes a critical factor for IBD development is largely unknown and remains to be further investigated. Finally, the exposome could be a tool to predict relapses [322]. The development of electronic technologies to continuously record a patient’s exposome could allow disease-modifying exposures to be detected and acted on early to prevent relapse or disease progression [322].

Author Contributions

L.P.-B. conceived the study. S.V. wrote the article and created tables. B.C., V.H., G.B., J.-Y.J., P.N., A.-C.H., N.C.N., D.M., S.D. and L.P.-B. critically reviewed the contents of the paper. The manuscript was approved by all authors. All authors have read and agreed to the published version of the manuscript.

Funding

S.V. was financially supported by the National Fund for Scientific Research (F.R.S.-FNRS) (grant numbers 32729160 and 40001034).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

S. Vieujean received speaker’s fees from Abbvie and Janssen Pfizer Shire. B. Caron reports lecture and/or consulting fees from Abbvie, Amgen, Celltrion, Ferring, Janssen and Takeda. S. Danese has served as a speaker, consultant and advisory board member for Schering-Plough, AbbVie, Actelion, Alphawasserman, AstraZeneca, Cellerix, Cosmo Pharmaceuticals, Ferring, Genentech, Grunenthal, Johnson and Johnson, Millenium Takeda, MSD, Nikkiso Europe GmbH, Novo Nordisk, Nycomed, Pfizer, Pharmacosmos, UCB Pharma and Vifor. L. Peyrin-Biroulet: personal fees from Galapagos, AbbVie, Janssen, Genentech, Ferring, Tillots, Pharmacosmos, Celltrion, Takeda, Boerhinger Ingelheim, Pfizer, Index Pharmaceuticals, Sandoz, Celgene, Biogen, Samsung Bioepis, Alma, Sterna, Nestle, Inotrem, Enterome, Allergan, MSD, Roche, Arena, Gilead, Hikma, Amgen, BMS, Vifor and Norgine. Mylan, Lilly: Fresenius Kabi, Oppilan Pharma, Sublimity Therapeutics, Applied Molecular Transport, OSE Immunotherapeutics, Enther and, Theravance, Pandion Therapeutics and grants from Abbvie, MSD, Takeda and Fresenius Kabi and stock options: CTMA.

Abbreviations

5hmC, 5-hydroxymethyl cytosine; ADAM17, a disintegrin and metalloprotease17; AIEC, Adherent-invasive Escherichia coli; CD, Crohn’s disease; CEACAM6, CEA Cell Adhesion Molecule 6; CpG, cytosine-phosphate-guanine; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; EMT, epithelial-to-mesenchymal transition; HAT, histone acetyltransferase; HDAC, histone deacetylases; HDM, histone demethylases; HMT, histone methyltransferase; HRE, HIF-1-responsive elements; IBD, inflammatory bowel disease; IEC, intestinal epithelial cell; IL, interleukin; Jnk, c-Jun N-terminal kinase; LPS, lipopolysaccharide; lncRNA, long non-coding RNA; MAP, Mycobacterium avium subspecies paratuberculosis; MeSH, Medical Subject Heading; miR, micro-RNA; mRNA, messenger RNAs; ncRNA, non-coding RNA; NF-κB, nuclear factor-kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; NOD2, nucleotide-binding oligomerization domain 2; NSAID, non-steroidal anti-inflammatory drug; PPAR-γ, peroxisome proliferator-activated receptor γ; RNA, ribonucleic acid; SAM, S-adenosyl-L-methionine; SCFA, short-chain fatty acid; SIRT, sirtuin; STAT, signal transducer and activator of transcription; TET, ten-eleven translocation enzymes; TNF, tumor necrosis factor; UC, ulcerative colitis.

References

  1. Danese, S.; Fiocchi, C. Etiopathogenesis of inflammatory bowel diseases. World J. Gastroenterol. 2006, 12, 4807–4812. [Google Scholar] [CrossRef] [PubMed]
  2. Park, J.H.; Wacholder, S.; Gail, M.H.; Peters, U.; Jacobs, K.B.; Chanock, S.J.; Chatterjee, N. Estimation of effect size distribution from genome-wide association studies and implications for future discoveries. Nat. Genet. 2010, 42, 570–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shanahan, F. The gut microbiota-A clinical perspective on lessons learned. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 609–614. [Google Scholar] [CrossRef]
  4. Bernstein, C.N.; Shanahan, F. Disorders of a modern lifestyle: Reconciling the epidemiology of inflammatory bowel diseases. Gut 2008, 57, 1185–1191. [Google Scholar] [CrossRef] [PubMed]
  5. Rubio, K.; Dobersch, S.; Barreto, G. Functional interactions between scaffold proteins, noncoding RNAs, and genome loci induce liquid-liquid phase separation as organizing principle for 3-dimensional nuclear architecture: Implications in cancer. FASEB J. 2019, 33, 5814–5822. [Google Scholar] [CrossRef]
  6. Dobersch, S.; Rubio, K.; Singh, I.; Günther, S.; Graumann, J.; Cordero, J.; Castillo-Negrete, R.; Huynh, M.B.; Mehta, A.; Braubach, P.; et al. Positioning of nucleosomes containing γ-H2AX precedes active DNA demethylation and transcription initiation. Nat. Commun. 2021, 12, 1072. [Google Scholar] [CrossRef] [PubMed]
  7. Rogler, G.; Vavricka, S. Exposome in IBD: Recent insights in environmental factors that influence the onset and course of IBD. Inflamm. Bowel Dis. 2015, 21, 400–408. [Google Scholar] [CrossRef]
  8. Fraga, M.F.; Ballestar, E.; Paz, M.F.; Ropero, S.; Setien, F.; Ballestar, M.L.; Heine-Suñer, D.; Cigudosa, J.C.; Urioste, M.; Benitez, J.; et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 2005, 102, 10604–10609. [Google Scholar] [CrossRef] [Green Version]
  9. Christensen, B.C.; Houseman, E.A.; Marsit, C.; Zheng, S.; Wrensch, M.R.; Wiemels, J.L.; Nelson, H.; Karagas, M.R.; Padbury, J.F.; Bueno, R.; et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CPG island context. PLoS Genet. 2009, 5, e1000602. [Google Scholar] [CrossRef] [Green Version]
  10. Szarc Vel Szic, K.; Declerck, K.; Vidaković, M.; Vanden Berghe, W. From inflammaging to healthy aging by dietary lifestyle choices: Is epigenetics the key to personalized nutrition? Clin. Epigenetics 2015, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  11. Wild, C.P. Complementing the genome with an “exposome”: The outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1847–1850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Rappaport, S.M. Implications of the exposome for exposure science. J. Expo. Sci. Environ. Epidemiol. 2011, 21, 5–9. [Google Scholar] [CrossRef] [PubMed]
  13. Waterland, R.A.; Michels, K.B. Epigenetic epidemiology of the developmental origins hypothesis. Annu. Rev. Nutr. 2007, 27, 363–388. [Google Scholar] [CrossRef]
  14. Barclay, A.R.; Russell, R.K.; Wilson, M.L.; Gilmour, W.H.; Satsangi, J.; Wilson, D.C. Systematic Review: The Role of Breastfeeding in the Development of Pediatric Inflammatory Bowel Disease. J. Pediatr. 2009, 155, 421–426. [Google Scholar] [CrossRef] [PubMed]
  15. Tysk, C.; Lindberg, E.; Jarnerot, G.; Floderus-Myrhed, B. Ulcerative colitis and Crohn’s disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut 1988, 29, 990–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Nikolopoulos, G.K.; Lytras, T.; Bonovas, S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Meta-Analyses. Gastroenterology 2019, 157, 647–659.e4. [Google Scholar] [CrossRef] [Green Version]
  17. Boyko, E.J.; Theis, M.K.; Vaughan, T.L.; Nicol-blades, B. Increased risk of inflammatory bowel disease associated with oral contraceptive use. Am. J. Epidemiol. 1994, 140, 268–278. [Google Scholar] [CrossRef]
  18. Shaw, S.Y.; Blanchard, J.F.; Bernstein, C.N. Association between the use of antibiotics and new diagnoses of Crohn’s disease and ulcerative colitis. Am. J. Gastroenterol. 2011, 106, 2133–2142. [Google Scholar] [CrossRef]
  19. Bernstein, C.N. Psychological Stress and Depression: Risk Factors for IBD? Dig. Dis. 2016, 34, 58–63. [Google Scholar] [CrossRef]
  20. Singh, S.; Graff, L.A.; Bernstein, C.N. Do NSAIDs, antibiotics, infections, or stress trigger flares in IBD? Am. J. Gastroenterol. 2009, 104, 1298–1313. [Google Scholar] [CrossRef]
  21. DeFilippis, E.M.; Longman, R.; Harbus, M.; Dannenberg, K.; Scherl, E.J. Crohn’s Disease: Evolution, Epigenetics, and the Emerging Role of Microbiome-Targeted Therapies. Curr. Gastroenterol. Rep. 2016, 18, 13. [Google Scholar] [CrossRef] [PubMed]
  22. Fiocchi, C. Inflammatory bowel disease pathogenesis: Where are we? J. Gastroenterol. Hepatol. 2015, 30 (Suppl. 1), 12–18. [Google Scholar] [CrossRef] [PubMed]
  23. Ray, G.; Longworth, M.S. Epigenetics, DNA Organization, and Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2019, 25, 235–247. [Google Scholar] [CrossRef] [PubMed]
  24. Bernstein, B.E.; Meissner, A.; Lander, E.S. The Mammalian Epigenome. Cell 2007, 128, 669–681. [Google Scholar] [CrossRef] [Green Version]
  25. Zhang, Y. Recent progress in the epigenetics and chromatin field. Cell Res. 2011, 21, 373–374. [Google Scholar] [CrossRef] [Green Version]
  26. Ozturk, N.; Singh, I.; Mehta, A.; Braun, T.; Barreto, G. HMGA proteins as modulators of chromatin structure during transcriptional activation. Front. Cell Dev. Biol. 2014, 2, 5. [Google Scholar] [CrossRef] [Green Version]
  27. Richmond, T.J.; Finch, J.T.; Rushton, B.; Rhodes, D.; Klug, A. Structure of the nucleosome core particle at 7 resolution. Nature 1984, 311, 532–537. [Google Scholar] [CrossRef]
  28. Bentley, G.A.; Lewit-Bentley, A.; Finch, J.T.; Podjarny, A.D.; Roth, M. Crystal structure of the nucleosome core particle at 16 Å resolution. J. Mol. Biol. 1984, 176, 55–75. [Google Scholar] [CrossRef]
  29. Singh, I.; Contreras, A.; Cordero, J.; Rubio, K.; Dobersch, S.; Günther, S.; Jeratsch, S.; Mehta, A.; Krüger, M.; Graumann, J.; et al. MiCEE is a ncRNA-protein complex that mediates epigenetic silencing and nucleolar organization. Nat. Genet. 2018, 50, 990–1001. [Google Scholar] [CrossRef]
  30. Cedar, H.; Bergman, Y. Linking DNA methylation and histone modification: Patterns and paradigms. Nat. Rev. Genet. 2009, 10, 295–304. [Google Scholar] [CrossRef]
  31. Deaton, A.M.; Bird, A. CpG islands and the regulation of transcription. Genes Dev. 2011, 25, 1010–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Skinner, M.K. Environmental epigenetics and a unified theory of the molecular aspects of evolution: A neo-Lamarckian concept that facilitates neo-Darwinian evolution. Genome Biol. Evol. 2015, 7, 1296–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dobersch, S.; Rubio, K.; Barreto, G. Pioneer Factors and Architectural Proteins Mediating Embryonic Expression Signatures in Cancer. Trends Mol. Med. 2019, 25, 287–302. [Google Scholar] [CrossRef]
  34. Esteller, M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet. 2007, 8, 286–298. [Google Scholar] [CrossRef] [PubMed]
  35. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002, 16, 6–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Schübeler, D. Function and information content of DNA methylation. Nature 2015, 517, 321–326. [Google Scholar] [CrossRef]
  37. Bird, A.P.; Wolffe, A.P. Methylation-induced repression-belts, braces, and chromatin. Cell 1999, 99, 451–454. [Google Scholar] [CrossRef] [Green Version]
  38. Gardiner-Garden, M.; Frommer, M. CpG Islands in vertebrate genomes. J. Mol. Biol. 1987, 196, 261–282. [Google Scholar] [CrossRef]
  39. Lim, D.H.K.; Maher, E.R. DNA methylation: A form of epigenetic control of gene expression. Obstet. Gynaecol. 2010, 12, 37–42. [Google Scholar] [CrossRef]
  40. Yang, I.V.; Schwartz, D.A. Epigenetic control of gene expression in the lung. Am. J. Respir. Crit. Care Med. 2011, 183, 1295–1301. [Google Scholar] [CrossRef] [Green Version]
  41. Reik, W.; Dean, W.; Walter, J. Epigenetic Reprogramming in Mammalian Development. Science 2001, 293, 1089–1093. [Google Scholar] [CrossRef] [Green Version]
  42. Jones, P.A.; Liang, G. Rethinking how DNA methylation patterns are maintained. Nat. Rev. Genet. 2009, 10, 805–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Law, J.A.; Jacobsen, S.E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 2010, 11, 204–220. [Google Scholar] [CrossRef]
  44. Jin, B.; Robertson, K.D. DNA methyltransferases, DNA damage repair, and cancer. Adv. Exp. Med. Biol. 2013, 754, 3–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [Green Version]
  46. Nimmo, E.R.; Prendergast, J.G.; Aldhous, M.C.; Kennedy, N.A.; Henderson, P.; Drummond, H.E.; Ramsahoye, B.H.; Wilson, D.C.; Semple, C.A.; Satsangi, J. Genome-wide methylation profiling in Crohn’s disease identifies altered epigenetic regulation of key host defense mechanisms including the Th17 pathway. Inflamm. Bowel Dis. 2012, 18, 889–899. [Google Scholar] [CrossRef]
  47. Häsler, R.; Feng, Z.; Bäckdahl, L.; Spehlmann, M.E.; Franke, A.; Teschendorff, A.; Rakyan, V.K.; Down, T.A.; Wilson, G.A.; Feber, A.; et al. A functional methylome map of ulcerative colitis. Genome Res. 2012, 22, 2130–2137. [Google Scholar] [CrossRef] [Green Version]
  48. Cooke, J.; Zhang, H.; Greger, L.; Silva, A.L.; Massey, D.; Dawson, C. Mucosal genome-wide methylation changes in inflammatory bowel disease. Inflamm. Bowel Dis. 2012, 18, 2128–2137. [Google Scholar] [CrossRef] [PubMed]
  49. Saito, S.; Kato, J.; Hiraoka, S.; Horii, J.; Suzuki, H.; Higashi, R.; Kaji, E.; Kondo, Y.; Yamamoto, K. DNA methylation of colon mucosa in ulcerative colitis patients: Correlation with inflammatory status. Inflamm. Bowel Dis. 2011, 17, 1955–1965. [Google Scholar] [CrossRef] [Green Version]
  50. Harris, R.; Nagy-Szakal, R.; Mir, S.A.V.; Frank, E.; Szigeti, R.; Kaplan, J.L.; Bronsky, J.; Opekun, A.; Ferry, G.D.; Winter, H.; et al. DNA methylation-associated colonic mucosal immune and defense responses in treatmentnaïve pediatric ulcerative colitis. Epigenetics 2014, 9, 1131–1137. [Google Scholar] [CrossRef] [Green Version]
  51. Koizumi, K.; Alonso, S.; Miyaki, Y.; Okada, S.; Ogura, H.; Shiiya, N.; Konishi, F.; Taya, T.; Perucho, M.; Suzuki, K. Array-based identification of common DNA methylation alterations in ulcerative colitis. Int. J. Oncol. 2012, 40, 983–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Lin, Z.; Hegarty, J.P.; Yu, W.; Cappel, J.A.; Chen, X.; Faber, P.W.; Wang, Y.; Poritz, L.S.; Fan, J.-B.; Koltun, W.A. Identification of disease-associated DNA methylation in B cells from Crohn’s disease and ulcerative colitis patients. Dig. Dis. Sci. 2012, 57, 3145–3153. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Y.; Fischle, W.; Cheung, W.; Jacobs, S.; Khorasanizadeh, S.; Allis, C.D. Beyond the double helix: Writing and reading the histone code. Novartis Found. Symp. 2004, 259, 3–17; discussion 17–21, 163–169. [Google Scholar] [PubMed]
  54. Singh, I.; Ozturk, N.; Cordero, J.; Mehta, A.; Hasan, D.; Cosentino, C.; Sebastian, C.; Krüger, M.; Looso, M.; Carraro, G.; et al. High mobility group protein-mediated transcription requires DNA damage marker γ-H2AX. Cell Res. 2015, 25, 837–850. [Google Scholar] [CrossRef] [Green Version]
  55. Zhang, M.; Zhao, J.; Lv, Y.; Wang, W.; Feng, C.; Zou, W.; Su, L.; Jiao, J. Histone Variants and Histone Modifications in Neurogenesis. Trends Cell Biol. 2020, 30, 869–880. [Google Scholar] [CrossRef]
  56. Nakayama, T.; Takami, Y. Participation of histones and histone-modifying enzymes in cell functions through alterations in chromatin structure. J. Biochem. 2001, 129, 491–499. [Google Scholar] [CrossRef]
  57. Morgan, M.A.J.; Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 2020, 52, 1271–1281. [Google Scholar] [CrossRef]
  58. Stillman, B. Histone Modifications: Insights into Their Influence on Gene Expression. Cell 2018, 175, 6–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Nie, L.; Wu, H.J.; Hsu, J.M.; Chang, S.S.; LaBaff, A.M.; Li, C.W.; Wang, Y.; Hsu, J.L.; Hung, M.C. Long non-coding RNAs: Versatile master regulators of gene expression and crucial players in cancer. Am. J. Transl. Res. 2012, 4, 127–150. [Google Scholar]
  60. Aalto, A.P.; Pasquinelli, A.E. Small non-coding RNAs mount a silent revolution in gene expression. Curr. Opin. Cell Biol. 2012, 24, 333–340. [Google Scholar] [CrossRef] [Green Version]
  61. Pekow, J.R.; Kwon, J.H. MicroRNAs in inflammatory bowel disease. Inflamm. Bowel Dis. 2012, 18, 187–193. [Google Scholar] [CrossRef] [PubMed]
  62. Coskun, M.; Bjerrum, J.T.; Seidelin, J.B.; Nielsen, O.H. MicroRNAs in inflammatory bowel disease-pathogenesis, diagnostics and therapeutics. World J. Gastroenterol. 2012, 18, 4629–4634. [Google Scholar] [CrossRef] [PubMed]
  63. Annese, V. Genetics and epigenetics of IBD. Pharmacol. Res. 2020, 159, 104892. [Google Scholar] [CrossRef] [PubMed]
  64. Kellermayer, R.; Zilbauer, M. The Gut Microbiome and the Triple Environmental Hit Concept of Inflammatory Bowel Disease Pathogenesis. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 589–595. [Google Scholar] [CrossRef] [PubMed]
  65. Zong, D.; Liu, X.; Li, J.; Ouyang, R.; Chen, P. The role of cigarette smoke-induced epigenetic alterations in inflammation. Epigenetics Chromatin 2019, 12, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Marciniak, A.; Patro-Małysza, J.; Kimber-Trojnar, Ż.; Marciniak, B.; Oleszczuk, J.; Leszczyńska-Gorzelak, B. Fetal programming of the metabolic syndrome. Taiwan. J. Obstet. Gynecol. 2017, 56, 133–138. [Google Scholar] [CrossRef] [PubMed]
  67. Dunford, A.R.; Sangster, J.M. Maternal and paternal periconceptional nutrition as an indicator of offspring metabolic syndrome risk in later life through epigenetic imprinting: A systematic review. Diabetes Metab. Syndr. Clin. Res. Rev. 2017, 11, S655–S662. [Google Scholar] [CrossRef]
  68. Rasmussen, L.; Knorr, S.; Antoniussen, C.S.; Bruun, J.M.; Ovesen, P.G.; Fuglsang, J.; Kampmann, U. The impact of lifestyle, diet and physical activity on epigenetic changes in the offspring—A systematic review. Nutrients 2021, 13, 2821. [Google Scholar] [CrossRef]
  69. Aguilera, O.; Fernández, A.F.; Muñoz, A.; Fraga, M.F. Epigenetics and environment: A complex relationship. J. Appl. Physiol. 2010, 109, 243–251. [Google Scholar] [CrossRef] [Green Version]
  70. Tschurtschenthaler, M.; Kachroo, P.; Heinsen, F.A.; Adolph, T.E.; Rühlemann, M.C.; Klughammer, J.; Offner, F.A.; Ammerpohl, O.; Krueger, F.; Smallwood, S.; et al. Paternal chronic colitis causes epigenetic inheritance of susceptibility to colitis. Sci. Rep. 2016, 6, 31640. [Google Scholar] [CrossRef]
  71. Morgan, H.D.; Sutherland, H.G.E.; Martin, D.I.K.; Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet. 1999, 23, 314–318. [Google Scholar] [CrossRef]
  72. Youngson, N.A.; Whitelaw, E. Transgenerational epigenetic effects. Annu. Rev. Genom. Hum. Genet. 2008, 9, 233–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Schaible, T.D.; Harris, R.A.; Dowd, S.E.; Smith, C.W.; Kellermayer, R. Maternal methyl-donor supplementation induces prolonged murine offspring colitis susceptibility in association with mucosal epigenetic and microbiomic changes. Hum. Mol. Genet. 2011, 20, 1687–1696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Waterland, R.A.; Jirtle, R.L. Transposable Elements: Targets for Early Nutritional Effects on Epigenetic Gene Regulation. Mol. Cell. Biol. 2003, 23, 5293–5300. [Google Scholar] [CrossRef] [Green Version]
  75. Tobi, E.; Lumey, L.H.; Talens, R.P.; Kremer, D.; Putter, H.; Stein, A.; Slagboom, P.; Heijmans, B.T. DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum. Mol. Genet. 2009, 18, 4046–4053. [Google Scholar] [CrossRef] [PubMed]
  76. Heijmans, B.T.; Tobi, E.W.; Lumey, L.H.; Slagboom, P.E. The epigenome: Archive of the prenatal environment. Epigenetics 2009, 4, 526–531. [Google Scholar] [CrossRef] [Green Version]
  77. Ventham, N.T.; Kennedy, N.A.; Nimmo, E.R.; Satsangi, J. Beyond gene discovery in inflammatory bowel disease: The emerging role of epigenetics. Gastroenterology 2013, 145, 293–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Blewitt, M.E.; Vickaryous, N.K.; Paldi, A.; Koseki, H.; Whitelaw, E. Dynamic reprogramming of DNA methylation at an epigenetically sensitive allele in mice. PLoS Genet. 2006, 2, 399–405. [Google Scholar] [CrossRef] [PubMed]
  79. Grossniklaus, U.; Kelly, B.; Ferguson-Smith, A.C.; Pembrey, M.; Lindquist, S. Transgenerational epigenetic inheritance: How important is it? Nat. Rev. Genet. 2013, 14, 228–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Lane, N.; Dean, W.; Erhardt, S.; Hajkova, P.; Surani, A.; Reik, W. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 2003, 35, 88–93. [Google Scholar] [CrossRef]
  81. Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 2010, 465, 721–727. [Google Scholar] [CrossRef] [PubMed]
  82. Einstein, F.H. Multigenerational effects of maternal undernutrition. Cell Metab. 2014, 19, 893–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Li, X.; Sundquist, J.; Hemminki, K.; Sundquist, K. Risk of inflammatory bowel disease in first- and second-generation immigrants in Sweden: A nationwide follow-up study. Inflamm. Bowel Dis. 2011, 17, 1784–1791. [Google Scholar] [CrossRef]
  84. Pinsk, V.; Lemberg, D.A.; Grewal, K.; Barker, C.C.; Schreiber, R.A.; Jacobson, K. Inflammatory bowel disease in the South Asian pediatric population of British Columbia. Am. J. Gastroenterol. 2007, 102, 1077–1083. [Google Scholar] [CrossRef]
  85. Benchimol, E.I.; Mack, D.R.; Guttmann, A.; Nguyen, G.C.; To, T.; Mojaverian, N.; Quach, P.; Manuel, D.G. Inflammatory bowel disease in immigrants to Canada and their children: A population-based cohort study. Am. J. Gastroenterol. 2015, 110, 553–563. [Google Scholar] [CrossRef] [PubMed]
  86. Lim, A.I.; McFadden, T.; Link, V.M.; Han, S.J.; Karlsson, R.M.; Stacy, A.; Farley, T.K.; Lima-Junior, D.S.; Harrison, O.J.; Desai, J.V.; et al. Prenatal maternal infection promotes tissue-specific immunity and inflammation in offspring. Science 2021, 373, eabf3002. [Google Scholar] [CrossRef]
  87. Wiklund, P.; Karhunen, V.; Richmond, R.C.; Parmar, P.; Rodriguez, A.; De Silva, M.; Wielscher, M.; Rezwan, F.I.; Richardson, T.G.; Veijola, J.; et al. DNA methylation links prenatal smoking exposure to later life health outcomes in offspring. Clin. Epigenetics 2019, 11, 97. [Google Scholar] [CrossRef] [Green Version]
  88. Martin, E.M.; Fry, R.C. Environmental Influences on the Epigenome: Exposure-Associated DNA Methylation in Human Populations. Annu. Rev. Public Health 2018, 39, 309–333. [Google Scholar] [CrossRef] [Green Version]
  89. Flom, J.D.; Ferris, J.S.; Liao, Y.; Tehranifar, P.; Richards, C.B.; Cho, Y.H.; Gonzalez, K.; Santella, R.M.; Terry, M.B. Prenatal smoke exposure and genomic DNA methylation in a multiethnic birth cohort. Cancer Epidemiol. Biomark. Prev. 2011, 20, 2518–2523. [Google Scholar] [CrossRef] [Green Version]
  90. Breton, C.V.; Byun, H.M.; Wenten, M.; Pan, F.; Yang, A.; Gilliland, F.D. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am. J. Respir. Crit. Care Med. 2009, 180, 462–467. [Google Scholar] [CrossRef] [Green Version]
  91. Guerrero-Preston, R.; Goldman, L.R.; Brebi-Mieville, P.; Ili, C.; Lebron, C.; Witter, F.R.; Apelberg, B.J.; Hernández-Roystacher, M.; Jaffe, A.; Halden, R.; et al. Global DNA hypomethylation is associated with in utero exposure to cotinine and perfluorinated alkyl compounds. Epigenetics 2010, 5, 539–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Janssen, B.G.; Gyselaers, W.; Byun, H.-M.; Roels, H.A.; Cuypers, A.; Baccarelli, A.A.; Nawrot, T.S. Placental mitochondrial DNA and CYP1A1 gene methylation as molecular signatures for tobacco smoke exposure in pregnant women and the relevance for birth weight. J. Transl. Med. 2017, 15, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kaur, G.; Begum, R.; Thota, S.; Batra, S. A systematic review of smoking-related epigenetic alterations. Arch. Toxicol. 2019, 93, 2715–2740. [Google Scholar] [CrossRef] [PubMed]
  94. Richmond, R.C.; Suderman, M.; Langdon, R.; Relton, C.L.; Smith, G.D. DNA methylation as a marker for prenatal smoke exposure in adults. Int. J. Epidemiol. 2018, 47, 1120–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Herberth, G.; Bauer, M.; Gasch, M.; Hinz, D.; Röder, S.; Olek, S.; Kohajda, T.; Rolle-Kampczyk, U.; von Bergen, M.; Sack, U.; et al. Maternal and cord blood miR-223 expression associates with prenatal tobacco smoke exposure and low regulatory T-cell numbers. J. Allergy Clin. Immunol. 2014, 133, 543–550.e4. [Google Scholar] [CrossRef]
  96. Jenkins, T.G.; James, E.R.; Alonso, D.F.; Hoidal, J.R.; Murphy, P.J.; Hotaling, J.M.; Cairns, B.R.; Carrell, D.T.; Aston, K.I. Cigarette smoking significantly alters sperm DNA methylation patterns. Andrology 2017, 5, 1089–1099. [Google Scholar] [CrossRef]
  97. Knopik, V.S.; Maccani, M.A.; Francazio, S.; McGeary, J.E. The epigenetics of maternal cigarette smoking during pregnancy and effects on child development. Dev. Psychopathol. 2012, 24, 1377–1390. [Google Scholar] [CrossRef] [Green Version]
  98. Suter, M.; Ma, J.; Harris, A.S.; Patterson, L.; Brown, K.A.; Shope, C.; Showalter, L.; Abramovici, A.; Aagaard-Tillery, K.M. Maternal tobacco use modestly alters correlated epigenome-wide placental DNA methylation and gene expression. Epigenetics 2011, 6, 1284–1294. [Google Scholar] [CrossRef] [Green Version]
  99. Ventham, N.T.; IBD BIOM consortium; Kennedy, N.; Adams, A.T.; Kalla, R.; Heath, S.C.; O’Leary, K.R.; Drummond, H.E.; Wilson, D.C.; Gut, I.G.; et al. Integrative epigenome-wide analysis demonstrates that DNA methylation may mediate genetic risk in inflammatory bowel disease. Nat. Commun. 2016, 7, 13507. [Google Scholar] [CrossRef] [Green Version]
  100. Kellermayer, R. Epigenetics and the developmental origins of inflammatory bowel diseases. Can. J. Gastroenterol. 2012, 26, 909–915. [Google Scholar] [CrossRef]
  101. Guo, F.; Cai, D.; Li, Y.; Gu, H.; Qu, H.; Zong, Q.; Bao, W.; Chen, A.; Liu, H.-Y. How Early-Life Gut Microbiota Alteration Sets Trajectories for Health and Inflammatory Bowel Disease? Front. Nutr. 2021, 8, 690073. [Google Scholar] [CrossRef]
  102. Dominguez-Bello, M.G.; Blaser, M.J.; Ley, R.E.; Knight, R. Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology 2011, 140, 1713–1719. [Google Scholar] [CrossRef] [PubMed]
  103. Yu, D.-H.; Gadkari, M.; Zhou, Q.; Yu, S.; Gao, N.; Guan, Y.; Schady, D.; Roshan, T.N.; Chen, M.-H.; Laritsky, E.; et al. Postnatal epigenetic regulation of intestinal stem cells requires DNA methylation and is guided by the microbiome. Genome Biol. 2015, 16, 211. [Google Scholar] [CrossRef] [Green Version]
  104. Poupeau, A.; Garde, C.; Sulek, K.; Citirikkaya, K.; Treebak, J.T.; Arumugam, M.; Simar, D.; Olofsson, L.E.; Bäckhed, F.; Barrès, R. Genes controlling the activation of natural killer lymphocytes are epigenetically remodeled in intestinal cells from germ-free mice. FASEB J. 2019, 33, 2719–2731. [Google Scholar] [CrossRef] [Green Version]
  105. Amatullah, H.; Jeffrey, K.L. Epigenome-metabolome-microbiome axis in health and IBD. Curr. Opin. Microbiol. 2020, 56, 97–108. [Google Scholar] [CrossRef] [PubMed]
  106. Wu, Y.; Wang, C.-Z.; Wan, J.-Y.; Yao, H.; Yuan, C.-S. Dissecting the interplay mechanism between epigenetics and gut microbiota: Health maintenance and disease prevention. Int. J. Mol. Sci. 2021, 22, 6933. [Google Scholar] [CrossRef]
  107. Liu, S.; da Cunha, A.P.; Rezende, R.M.; Cialic, R.; Wei, Z.; Bry, L.; Comstock, L.E.; Gandhi, R.; Weiner, H.L. The Host Shapes the Gut Microbiota via Fecal MicroRNA. Cell Host Microbe 2016, 19, 32–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ansari, I.; Raddatz, G.; Gutekunst, J.; Ridnik, M.; Cohen, D.; Abu-Remaileh, M.; Tuganbaev, T.; Shapiro, H.; Pikarsky, E.; Elinav, E.; et al. The microbiota programs DNA methylation to control intestinal homeostasis and inflammation. Nat. Microbiol. 2020, 5, 610–619. [Google Scholar] [CrossRef]
  109. Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species roseburia hominis and faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef]
  111. Frank, D.N.; St. Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Rossi, M.; Amaretti, A.; Raimondi, S. Folate production by probiotic bacteria. Nutrients 2011, 3, 118–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Sartor, R.B. Microbial Influences in Inflammatory Bowel Diseases. Gastroenterology 2008, 134, 577–594. [Google Scholar] [CrossRef] [PubMed]
  114. Meconi, S.; Vercellone, A.; Levillain, F.; Payré, B.; Al Saati, T.; Capilla, F.; Desreumaux, P.; Darfeuille-Michaud, A.; Altare, F. Adherent-invasive Escherichia coli isolated from Crohn’s disease patients induce granulomas in vitro. Cell. Microbiol. 2007, 9, 1252–1261. [Google Scholar] [CrossRef] [PubMed]
  115. Nguyen, H.T.T.; Dalmasso, G.; Müller, S.; Carrière, J.; Seibold, F.; Darfeuille-Michaud, A. Crohn’s disease-associated adherent invasive escherichia coli modulate levels of microRNAs in intestinal epithelial cells to reduce autophagy. Gastroenterology 2014, 146, 508–519. [Google Scholar] [CrossRef] [PubMed]
  116. Larabi, A.; Dalmasso, G.; Delmas, J.; Barnich, N.; Nguyen, H.T.T. Exosomes transfer miRNAs from cell-to-cell to inhibit autophagy during infection with Crohn’s disease-associated adherent-invasive E. Coli. Gut Microbes 2020, 11, 1677–1694. [Google Scholar] [CrossRef]
  117. Guo, Z.; Cai, X.; Guo, X.; Xu, Y.; Gong, J.; Li, Y.; Zhu, W. Let-7b ameliorates Crohn’s disease-associated adherent-invasive E coli induced intestinal inflammation via modulating Toll-Like Receptor 4 expression in intestinal epithelial cells. Biochem. Pharmacol. 2018, 156, 196–203. [Google Scholar] [CrossRef]
  118. Timms, V.J.; Daskalopoulos, G.; Mitchell, H.M.; Neilan, B.A. The association of mycobacterium avium subsp. paratuberculosis with inflammatory bowel disease. PLoS ONE 2016, 11, e0148731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Mostoufi-Afshar, S.; Tabatabaei, M.; Ghahramani Seno, M.M. Mycobacterium avium subsp. Paratuberculosis induces differential cytosine methylation at miR-21 transcription start site region. Iran. J. Vet. Res. 2018, 19, 262–269. [Google Scholar]
  120. Qiao, C.X.; Xu, S.; Wang, D.D.; Gao, S.Y.; Zhao, S.F.; Zhang, M.L.; Yu, B.; Yin, Q.; Zhao, G. MicroRNA-19b alleviates lipopolysaccharide-induced inflammatory injury in human intestinal cells by up-regulation of Runx3. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5284–5294. [Google Scholar] [CrossRef]
  121. Zhang, M.; Yang, D.; Yu, H.; Li, Q. MicroRNA-497 inhibits inflammation in DSS-induced IBD model mice and lipopolysaccharide-induced RAW264.7 cells via Wnt/β-catenin pathway. Int. Immunopharmacol. 2021, 101, 108318. [Google Scholar] [CrossRef] [PubMed]
  122. Sun, B.; Xing, K.; Qi, C.; Yan, K.; Xu, Y. Down-regulation of mir-215 attenuates lipopolysaccharide-induced inflammatory injury in ccd-18co cells by targeting gdf11 through the tlr4/nf-kb and jnk/p38 signaling pathways. Histol. Histopathol. 2020, 35, 1473–1481. [Google Scholar] [CrossRef] [PubMed]
  123. Wu, X.; Pan, S.; Luo, W.; Shen, Z.; Meng, X.; Xiao, M.; Tan, B.; Nie, K.; Tong, T.; Wang, X. Roseburia intestinalis-derived flagellin ameliorates colitis by targeting miR-223-3p-mediated activation of NLRP3 inflammasome and pyroptosis. Mol. Med. Rep. 2020, 22, 2695–2704. [Google Scholar] [CrossRef] [PubMed]
  124. Quan, Y.; Song, K.; Zhang, Y.; Zhu, C.; Shen, Z.; Wu, S.; Luo, W.; Tan, B.; Yang, Z.; Wang, X. Roseburia intestinalis-derived flagellin is a negative regulator of intestinal inflammation. Biochem. Biophys. Res. Commun. 2018, 501, 791–799. [Google Scholar] [CrossRef]
  125. Xue, X.; Feng, T.; Yao, S.; Wolf, K.J.; Liu, C.-G.; Liu, X. Microbiota Downregulates Dendritic Cell Expression of miR-10a, Which Targets IL-12/IL-23p40. J. Immunol. 2011, 187, 5879–5886. [Google Scholar] [CrossRef] [Green Version]
  126. Xue, X.; Cao, A.T.; Cao, X.; Yao, S.; Carlsen, E.D.; Soong, L. Downregulation of microRNA-107 in intestinal CD11c+ myeloid cells in response to microbiota and proinflammatory cytokines increases IL-23p19 expression. Eur. J. Immunol. 2014, 44, 673–682. [Google Scholar] [CrossRef] [Green Version]
  127. Wu, W.; He, C.; Liu, C.; Cao, A.T.; Xue, X.; Evans-Marin, H.L. miR-10a inhibits dendritic cell activation and Th1/Th17 cell immune responses in IBD. Gut 2015, 64, 1755–1764. [Google Scholar] [CrossRef]
  128. Nakata, K.; Sugi, Y.; Narabayashi, H.; Kobayakawa, T.; Nakanishi, Y.; Tsuda, M. Commensal Microbiota-induced microRNA modulates intestinal epithelial permeability through the small GTPase ARF4. J. Biol. Chem. 2017, 292, 15426–15433. [Google Scholar] [CrossRef] [Green Version]
  129. Johnston, D.G.W.; Williams, M.A.; Thaiss, C.A.; Cabrera-Rubio, R.; Raverdeau, M.; McEntee, C. Loss of microRNA-21 influences the gut microbiota, causing reduced susceptibility in a murine model of colitis. J. Crohn’s Colitis 2018, 12, 835–848. [Google Scholar] [CrossRef]
  130. Ryan, F.J.; Ahern, A.M.; Fitzgerald, R.S.; Laserna-Mendieta, E.J.; Power, E.M.; Clooney, A.G. Colonic microbiota is associated with inflammation and host epigenomic alterations in inflammatory bowel disease. Nat. Commun. 2020, 11, 1512. [Google Scholar] [CrossRef] [Green Version]
  131. Kim, W.K.; Han, D.H.; Jang, Y.J.; Park, S.; Jang, S.J.; Lee, G. Alleviation of DSS-induced colitis: Via Lactobacillus acidophilus treatment in mice. Food Funct. 2021, 12, 340–350. [Google Scholar] [CrossRef]
  132. Zhou, L.; Zhang, M.; Wang, Y.; Dorfman, R.G.; Liu, H.; Yu, T. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm. Bowel Dis. 2018, 24, 1926–1940. [Google Scholar] [CrossRef] [Green Version]
  133. Zhang, M.; Zhou, L.; Wang, Y.; Dorfman, R.G.; Tang, D.; Xu, L. Faecalibacterium prausnitzii produces butyrate to decrease c-Myc-related metabolism and Th17 differentiation by inhibiting histone deacetylase 3. Int. Immunol. 2019, 31, 499–514. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, Y.; Liu, L.; Liu, X.; Zhang, Y.Y.; Shi, H.; Jia, W. Extracellular Vesicles Derived from Trichinella spiralis Muscle Larvae Ameliorate TNBS-Induced Colitis in Mice. Front. Immunol. 2020, 11, 1174. [Google Scholar] [CrossRef] [PubMed]
  135. Cao, Y.; Wang, Z.; Yan, Y.; Ji, L.; He, J.; Xuan, B. Enterotoxigenic Bacteroides fragilis Promotes Intestinal Inflammation and Malignancy by Inhibiting Exosome-Packaged miR-149-3p. Gastroenterology 2021, 161, 1552–1566.e12. [Google Scholar] [CrossRef] [PubMed]
  136. Simon, J.M.; Davis, J.P.; Lee, S.E.; Schaner, M.R.; Gipson, G.R.; Weiser, M. Alterations to chromatin in intestinal macrophages link IL-10 deficiency to inappropriate inflammatory responses. Eur. J. Immunol. 2016, 46, 1912–1925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Anzola, A.; González, R.; Gámez-Belmonte, R.; Ocón, B.; Aranda, C.J.; Martínez-Moya, P. miR-146a regulates the crosstalk between intestinal epithelial cells, microbial components and inflammatory stimuli. Sci. Rep. 2018, 8, 17350. [Google Scholar] [CrossRef]
  138. Geng, H.; Bu, H.F.; Liu, F.; Wu, L.; Pfeifer, K.; Chou, P.M. In Inflamed Intestinal Tissues and Epithelial Cells, Interleukin 22 Signaling Increases Expression of H19 Long Noncoding RNA, Which Promotes Mucosal Regeneration. Gastroenterology 2018, 155, 144–155. [Google Scholar] [CrossRef] [PubMed]
  139. Lucafò, M.; Pugnetti, L.; Bramuzzo, M.; Curci, D.; Di Silvestre, A.; Marcuzzi, A. Long non-coding RNA gas5 and intestinal mmp2 and mmp9 expression: A translational study in pediatric patients with IBD. Int. J. Mol. Sci. 2019, 20, 5280. [Google Scholar] [CrossRef] [Green Version]
  140. Kempinska-Podhorodecka, A.; Adamowicz, M.; Ostrycharz, E.; Chmielarz, M.; Wójcicki, M.; Milkiewicz, P. Role of miR-506 in ulcerative colitis associated with primary sclerosing cholangitis. Sci. Rep. 2021, 11, 10134. [Google Scholar] [CrossRef]
  141. Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Bilotta, A.J.; Ma, C.; Yang, W.; Yu, Y.; Yu, Y.; Zhao, X.; Zhou, Z.; Yao, S.; Dann, S.M.; Cong, Y. Propionate Enhances Cell Speed and Persistence to Promote Intestinal Epithelial Turnover and Repair. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 1023–1044. [Google Scholar] [CrossRef] [PubMed]
  143. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  144. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
  145. Couto, M.R.; Gonçalves, P.; Magro, F.; Martel, F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol. Res. 2020, 159, 104947. [Google Scholar] [CrossRef] [PubMed]
  146. Plöger, S.; Stumpff, F.; Penner, G.B.; Schulzke, J.-D.; Gäbel, G.; Martens, H.; Shen, Z.; Günzel, D.; Aschenbach, J.R. Microbial butyrate and its role for barrier function in the gastrointestinal tract. Ann. N. Y. Acad. Sci. 2012, 1258, 52–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Leung, C.H.; Lam, W.; Ma, D.L.; Gullen, E.A.; Cheng, Y.C. Butyrate mediates nucleotide-binding and oligomerisation domain (NOD) 2-dependent mucosal immune responses against peptidoglycan. Eur. J. Immunol. 2009, 39, 3529–3537. [Google Scholar] [CrossRef]
  148. Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.W.; Pires, E.; et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity 2019, 50, 432–445.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Ji, J.; Shu, D.; Zheng, M.; Wang, J.; Luo, C.; Wang, Y.; Guo, F.; Zou, X.; Lv, X.; Li, Y.; et al. Microbial metabolite butyrate facilitates M2 macrophage polarization and function. Sci. Rep. 2016, 6, 24838. [Google Scholar] [CrossRef]
  151. Chen, L.; Sun, M.; Wu, W.; Yang, W.; Huang, X.; Xiao, Y.; Ma, C.; Xu, L.; Yao, S.; Liu, Z.; et al. Microbiota Metabolite Butyrate Differentially Regulates Th1 and Th17 Cells’ Differentiation and Function in Induction of Colitis. Inflamm. Bowel Dis. 2019, 25, 1450–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Li, G.; Lin, J.; Zhang, C.; Gao, H.; Lu, H.; Gao, X.; Zhu, R.; Li, Z.; Li, M.; Liu, Z. Microbiota metabolite butyrate constrains neutrophil functions and ameliorates mucosal inflammation in inflammatory bowel disease. Gut Microbes 2021, 13, 1968257. [Google Scholar] [CrossRef] [PubMed]
  153. Berndt, B.E.; Zhang, M.; Owyang, S.Y.; Cole, T.S.; Wang, T.W.; Luther, J.; Veniaminova, N.A.; Merchant, J.L.; Chen, C.-C.; Huffnagle, G.B.; et al. Butyrate increases IL-23 production by stimulated dendritic cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, 1384–1392. [Google Scholar] [CrossRef] [Green Version]
  154. Park, J.; Kim, M.; Kang, S.G.; Jannasch, A.H.; Cooper, B.; Patterson, J. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol. 2015, 8, 80–93. [Google Scholar] [CrossRef] [Green Version]
  155. Zou, F.; Qiu, Y.; Huang, Y.; Zou, H.; Cheng, X.; Niu, Q. Effects of short-chain fatty acids in inhibiting HDAC and activating p38 MAPK are critical for promoting B10 cell generation and function. Cell Death Dis. 2021, 12, 582. [Google Scholar] [CrossRef]
  156. Liu, Q.; Peng, Z.; Zhou, L.; Peng, R.; Li, X.; Zuo, W. Short-Chain Fatty Acid Decreases the Expression of CEBPB to Inhibit miR-145-Mediated DUSP6 and Thus Further Suppresses Intestinal Inflammation. Inflammation 2022, 45, 372–386. [Google Scholar] [CrossRef]
  157. Zimmerman, M.A.; Singh, N.; Martin, P.M.; Thangaraju, M.; Ganapathy, V.; Waller, J.L. Butyrate suppresses colonic inflammation through HDAC1-dependent fas upregulation and fas-mediated apoptosis of T cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, 1405–1415. [Google Scholar] [CrossRef]
  158. Lee, C.; Kim, B.G.; Kim, J.H.; Chun, J.; Im, J.P.; Kim, J.S. Sodium butyrate inhibits the NF-kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL-10 independent manner. Int. Immunopharmacol. 2017, 51, 47–56. [Google Scholar] [CrossRef]
  159. Kespohl, M.; Vachharajani, N.; Luu, M.; Harb, H.; Pautz, S.; Wolff, S. The microbial metabolite butyrate induces expression of Th1-associated factors in CD4+ T cells. Front. Immunol. 2017, 8, 1036. [Google Scholar] [CrossRef]
  160. Zhang, F.; Wang, W.; Niu, J.; Yang, G.; Luo, J.; Lan, D. Heat-shock transcription factor 2 promotes sodium butyrate-induced autophagy by inhibiting mTOR in ulcerative colitis. Exp. Cell Res. 2020, 388, 111820. [Google Scholar] [CrossRef] [PubMed]
  161. Wang, R.X.; Lee, J.S.; Campbell, E.L.; Colgan, S.P. Microbiota-derived butyrate dynamically regulates intestinal homeostasis through regulation of actin-associated protein synaptopodin. Proc. Natl. Acad. Sci. USA 2020, 117, 11648–11657. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, J.; Huang, N.; Xiong, J.; Wei, H.; Jiang, S.; Peng, J. Caprylic acid and nonanoic acid upregulate endogenous host defense peptides to enhance intestinal epithelial immunological barrier function via histone deacetylase inhibition. Int. Immunopharmacol. 2018, 65, 303–311. [Google Scholar] [CrossRef] [PubMed]
  163. Krautkramer, K.A.; Kreznar, J.H.; Romano, K.A.; Vivas, E.I.; Barrett-Wilt, G.A.; Rabaglia, M.E.; Keller, M.P.; Attie, A.D.; Rey, F.E.; Denu, J.M. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol. Cell 2016, 64, 982–992. [Google Scholar] [CrossRef] [Green Version]
  164. Dougherty, U.; Mustafi, R.; Zhu, H.; Zhu, X.; Deb, D.; Meredith, S.C.; Ayaloglu-Butun, F.; Fletcher, M.; Sanchez, A.; Pekow, J.; et al. Upregulation of polycistronic microRNA-143 and microRNA-145 in colonocytes suppresses colitis and inflammation-associated colon cancer. Epigenetics 2021, 16, 1317–1334. [Google Scholar] [CrossRef] [PubMed]
  165. Melhem, H.; Hansmannel, F.; Bressenot, A.; Battaglia-Hsu, S.-F.; Billioud, V.; Alberto, J.M.; Gueant, J.L.; Peyrin-Biroulet, L. Methyl-deficient diet promotes colitis and SIRT1-mediated endoplasmic reticulum stress. Gut 2016, 65, 595–606. [Google Scholar] [CrossRef] [PubMed]
  166. Denizot, J.; Desrichard, A.; Agus, A.; Uhrhammer, N.; Dreux, N.; Vouret-Craviari, V.; Hofman, P.; Darfeuille-Michaud, A.; Barnich, N. Diet-induced hypoxia responsive element demethylation increases CEACAM6 expression, favouring Crohn’s disease-associatedEscherichia colicolonisation. Gut 2014, 64, 428–437. [Google Scholar] [CrossRef]
  167. Wei, M.; Gao, X.; Liu, L.; Li, Z.; Wan, Z.; Dong, Y.; Chen, X.; Niu, Y.; Zhang, J.; Yang, G. Visceral Adipose Tissue Derived Exosomes Exacerbate Colitis Severity via Pro-inflammatory MiRNAs in High Fat Diet Fed Mice. ACS Nano 2020, 14, 5099–5110. [Google Scholar] [CrossRef]
  168. Lv, Q.; Shi, C.; Qiao, S.; Cao, N.; Guan, C.; Dai, Y.; Wei, Z. Alpinetin exerts anti-colitis efficacy by activating AhR, regulating miR-302/DNMT-1/CREB signals, and therefore promoting Treg differentiation. Cell Death Dis. 2018, 9, 1–25. [Google Scholar] [CrossRef]
  169. Xiong, Y.; Qiu, J.; Li, C.; Qiu, Y.; Guo, L.; Liu, Y.; Wan, J.; Li, Y.; Wu, G.; Wang, L.; et al. Fortunellin-induced modulation of phosphatase and tensin homolog by MicroRNA-374a decreases inflammation and maintains intestinal barrier function in colitis. Front. Immunol. 2018, 9, 83. [Google Scholar] [CrossRef] [Green Version]
  170. Wang, L.; Zhang, R.; Chen, J.; Wu, Q.; Kuang, Z. Baicalin protects against TNF-α-induced injury by down-regulating mir-191a that targets the tight junction protein ZO-1 in IEC-6 cells. Biol. Pharm. Bull. 2017, 40, 435–443. [Google Scholar] [CrossRef] [Green Version]
  171. Galleggiante, V.; De Santis, S.; Liso, M.; Verna, G.; Sommella, E.; Mastronardi, M.; Campiglia, P.; Chieppa, M.; Serino, G. Quercetin-Induced miR-369-3p Suppresses Chronic Inflammatory Response Targeting C/EBP-β. Mol. Nutr. Food Res. 2019, 63, 1801390. [Google Scholar] [CrossRef] [PubMed]
  172. James, S.; Aparna, J.; Babu, A.; Paul, A.; Lankadasari, M.; Athira, S.; Kumar, S.; Vijayan, Y.; Namitha, N.; Mohammed, S.; et al. Cardamonin attenuates experimental colitis and associated colorectal cancer. Biomolecules 2021, 11, 661. [Google Scholar] [CrossRef] [PubMed]
  173. Zhao, X.; Cui, D.J.; Yuan, W.Q.; Chen, C.; Liu, Q. Berberine represses Wnt/β-catenin pathway activation via modulating the microRNA-103a-3p/Bromodomain-containing protein 4 axis, thereby refraining pyroptosis and reducing the intestinal mucosal barrier defect induced via colitis. Bioengineered 2022, 13, 7392–7409. [Google Scholar] [CrossRef] [PubMed]
  174. Lv, Q.; Xing, Y.; Liu, J.; Dong, D.; Liu, Y.; Qiao, H.; Zhang, Y.; Hu, L. Lonicerin targets EZH2 to alleviate ulcerative colitis by autophagy-mediated NLRP3 inflammasome inactivation. Acta. Pharm. Sin. B 2021, 11, 2880–2899. [Google Scholar] [CrossRef] [PubMed]
  175. Alrafas, H.R.; Busbee, P.B.; Chitrala, K.N.; Nagarkatti, M.; Nagarkatti, P. Alterations in the gut microbiome and suppression of histone deacetylases by resveratrol are associated with attenuation of colonic inflammation and protection against colorectal cancer. J. Clin. Med. 2020, 9, 1796. [Google Scholar] [CrossRef]
  176. Alrafas, H.R.; Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol Downregulates miR-31 to Promote T Regulatory Cells during Prevention of TNBS-Induced Colitis. Mol. Nutr. Food Res. 2020, 64, 1900633. [Google Scholar] [CrossRef]
  177. Zeng, J.; Zhang, D.; Wan, X.; Bai, Y.; Yuan, C.; Wang, T.; Yuan, D.; Zhang, C.; Liu, C. Chlorogenic Acid Suppresses miR-155 and Ameliorates Ulcerative Colitis through the NF-κB/NLRP3 Inflammasome Pathway. Mol. Nutr. Food Res. 2020, 64, e2000452. [Google Scholar] [CrossRef]
  178. Benmoussa, A.; Diallo, I.; Salem, M.; Michel, S.; Gilbert, C.; Sévigny, J.; Provost, P. Concentrates of two subsets of extracellular vesicles from cow’s milk modulate symptoms and inflammation in experimental colitis. Sci. Rep. 2019, 9, 14661. [Google Scholar] [CrossRef] [Green Version]
  179. Reif, S.; Elbaum-Shiff, Y.; Koroukhov, N.; Shilo, I.; Musseri, M.; Golan-Gerstl, R. Cow and human milk-derived exosomes ameliorate colitis in DSS murine model. Nutrients 2020, 12, 2589. [Google Scholar] [CrossRef]
  180. Wu, D.; Kittana, H.; Shu, J.; Kachman, S.D.; Cui, J.; Ramer-Tait, A.E.; Zempleni, J. Dietary Depletion of Milk Exosomes and their MicroRNA Cargos Elicits a Depletion of miR-200a-3p and Elevated Intestinal Inflammation and CXCL9 Expression in Mdr1a−/− Mice. Curr. Dev. Nutr. 2019, 3, nzz122. [Google Scholar] [CrossRef]
  181. Zhang, X.; Gu, J.; Zhao, C.; Hu, Y.; Zhang, B.; Wang, J.; Lv, H.; Ji, X.; Wang, S. Sweeteners Maintain Epithelial Barrier Function Through the miR-15b/RECK/MMP-9 Axis, Remodel Microbial Homeostasis, and Attenuate Dextran Sodium Sulfate-Induced Colitis in Mice. J. Agric. Food Chem. 2022, 70, 171–183. [Google Scholar] [CrossRef] [PubMed]
  182. Sun, J.; Liang, W.; Yang, X.; Li, Q.; Zhang, G. Cytoprotective effects of galacto-oligosaccharides on colon epithelial cells via up-regulating miR-19b. Life Sci. 2019, 231, 116589. [Google Scholar] [CrossRef]
  183. Zha, Z.; Lv, Y.; Tang, H.; Li, T.; Miao, Y.; Cheng, J. An orally administered butyrate-releasing xylan derivative reduces inflammation in dextran sulphate sodium-induced murine colitis. Int. J. Biol. Macromol. 2020, 156, 1217–1233. [Google Scholar] [CrossRef] [PubMed]
  184. Qu, S.-L.; Chen, L.; Wen, X.-S.; Zuo, J.-P.; Wang, X.-Y.; Lu, Z.-J.; Yang, Y.-F. Suppression of Th17 cell differentiation via sphingosine-1-phosphate receptor 2 by cinnamaldehyde can ameliorate ulcerative colitis. Biomed. Pharmacother. 2021, 134, 111116. [Google Scholar] [CrossRef] [PubMed]
  185. Qu, S.; Shen, Y.; Wang, M.; Wang, X.; Yang, Y. Suppression of miR-21 and miR-155 of macrophage by cinnamaldehyde ameliorates ulcerative colitis. Int. Immunopharmacol. 2019, 67, 22–34. [Google Scholar] [CrossRef] [PubMed]
  186. Liu, S.; Zhang, S.; Lv, X.; Lu, J.; Ren, C.; Zeng, Z. Limonin ameliorates ulcerative colitis by regulating STAT3/miR-214 signaling pathway. Int. Immunopharmacol. 2019, 75, 105768. [Google Scholar] [CrossRef]
  187. Zhang, M.; Viennois, E.; Prasad, M.; Zhang, Y.; Wang, L.; Zhang, Z. Edible ginger-derived nanoparticles: A novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer. Biomaterials 2016, 101, 321–340. [Google Scholar] [CrossRef] [Green Version]
  188. Chen, X.; Xu, T.; Lv, X.; Zhang, J.; Liu, S. Ginsenoside Rh2 alleviates ulcerative colitis by regulating the STAT3/miR-214 signaling pathway. J. Ethnopharmacol. 2021, 274, 113997. [Google Scholar] [CrossRef]
  189. Wang, L.S.; Kuo, C.T.; Stoner, K.; Yearsley, M.; Oshima, K.; Yu, J. Dietary black raspberries modulate DNA methylation in dextran sodium sulfate (DSS)-induced ulcerative colitis. Carcinogenesis 2013, 34, 2842–2850. [Google Scholar] [CrossRef] [Green Version]
  190. Wang, L.S.; Kuo, C.T.; Huang, T.H.M.; Yearsley, M.; Oshima, K.; Stoner, G.D. Black raspberries protectively regulate methylation of Wnt pathway genes in precancerous colon tissue. Cancer Prev. Res. 2013, 6, 1317–1327. [Google Scholar] [CrossRef] [Green Version]
  191. Chen, Y.; Wang, C.; Liu, Y.; Tang, L.; Zheng, M.; Xu, C. MiR-122 targets NOD2 to decrease intestinal epithelial cell injury in Crohn’s disease. Biochem. Biophys. Res. Commun. 2013, 438, 133–139. [Google Scholar] [CrossRef] [PubMed]
  192. Lippai, D.; Bala, S.; Catalano, D.; Kodys, K.; Szabo, G. Micro-RNA-155 Deficiency Prevents Alcohol-Induced Serum Endotoxin Increase and Small Bowel Inflammation in Mice. Alcohol. Clin. Exp. Res. 2014, 38, 2217–2224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Jia, Y.; Wei, Y. Modulators of microrna function in the immune system. Int. J. Mol. Sci. 2020, 21, 2357. [Google Scholar] [CrossRef] [PubMed]
  194. Romagnolo, D.F.; Donovan, M.G.; Doetschman, T.C.; Selmin, O.I. N-6 linoleic acid induces epigenetics alterations associated with colonic inflammation and cancer. Nutrients 2019, 11, 171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Gimier, E.; Chervy, M.; Agus, A.; Sivignon, A.; Billard, E.; Privat, M. Methyl-donor supplementation prevents intestinal colonization by Adherent-Invasive, E. coli in a mouse model of Crohn’s disease. Sci. Rep. 2020, 10, 12922. [Google Scholar] [CrossRef]
  196. Amerikanou, C.; Papada, E.; Gioxari, A.; Smyrnioudis, I.; Kleftaki, S.A.; Valsamidou, E. Mastiha has efficacy in immune-mediated inflammatory diseases through a microRNA-155 Th17 dependent action. Pharmacol. Res. 2021, 171, 105753. [Google Scholar] [CrossRef]
  197. Chi, J.H.; Seo, G.S.; Cheon, J.H.; Lee, S.H. Isoliquiritigenin inhibits TNF-α-induced release of high-mobility group box 1 through activation of HDAC in human intestinal epithelial HT-29 cells. Eur. J. Pharmacol. 2017, 796, 101–109. [Google Scholar] [CrossRef]
  198. Angel-Morales, G.; Noratto, G.; Mertens-Talcott, S. Red wine polyphenolics reduce the expression of inflammation markers in human colon-derived CCD-18Co myofibroblast cells: Potential role of microRNA-126. Food Funct. 2012, 3, 745–752. [Google Scholar] [CrossRef]
  199. Ojwang, L.O.; Banerjee, N.; Noratto, G.D.; Angel-Morales, G.; Hachibamba, T.; Awika, J.M. Polyphenolic extracts from cowpea (Vigna unguiculata) protect colonic myofibroblasts (CCD18Co cells) from lipopolysaccharide (LPS)-induced inflammation–modulation of microRNA 126. Food Funct. 2015, 6, 145–153. [Google Scholar] [CrossRef]
  200. Kim, H.; Banerjee, N.; Barnes, R.C.; Pfent, C.M.; Talcott, S.T.; Dashwood, R.H. Mango polyphenolics reduce inflammation in intestinal colitis—involvement of the miR-126/PI3K/AKT/mTOR axis in vitro and in vivo. Mol. Carcinog. 2017, 56, 197–207. [Google Scholar] [CrossRef] [Green Version]
  201. Kim, H.; Banerjee, N.; Sirven, M.A.; Minamoto, Y.; Markel, M.E.; Suchodolski, J.S. Pomegranate polyphenolics reduce inflammation and ulceration in intestinal colitis—involvement of the miR-145/p70S6K1/HIF1α axis in vivo and in vitro. J. Nutr. Biochem. 2017, 43, 107–115. [Google Scholar] [CrossRef] [PubMed]
  202. Tian, M.; Peng, S.; Wang, S.; Li, X.; Li, H.; Shen, L. Pristimerin reduces dextran sulfate sodium-induced colitis in mice by inhibiting microRNA-155. Int. Immunopharmacol. 2021, 94, 107491. [Google Scholar] [CrossRef] [PubMed]
  203. Papoutsopoulou, S.; Satsangi, J.; Campbell, B.J.; Probert, C.S. Review article: Impact of cigarette smoking on intestinal inflammation—Direct and indirect mechanisms. Aliment. Pharmacol. Ther. 2020, 51, 1268–1285. [Google Scholar] [CrossRef] [PubMed]
  204. Ambatipudi, S.; Cuenin, C.; Hernandez-Vargas, H.; Ghantous, A.; Le Calvez-Kelm, F.; Kaaks, R. Tobacco smoking-associated genome-wide DNA methylation changes in the EPIC study. Epigenomics 2016, 8, 599–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Breitling, L.P.; Yang, R.; Korn, B.; Burwinkel, B.; Brenner, H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am. J. Hum. Genet. 2011, 88, 450–457. [Google Scholar] [CrossRef] [Green Version]
  206. Li, S.; Wong, E.M.; Bui, M.; Nguyen, T.L.; Joo, J.H.E.; Stone, J. Causal effect of smoking on DNA methylation in peripheral blood: A twin and family study. Clin. Epigenetics 2018, 10, 18. [Google Scholar] [CrossRef] [Green Version]
  207. Wan, E.S.; Qiu, W.; Baccarelli, A.; Carey, V.J.; Bacherman, H.; Rennard, S.I. Cigarette smoking behaviors and time since quitting are associated with differential DNA methylation across the human genome. Hum. Mol. Genet. 2012, 21, 3073–3082. [Google Scholar] [CrossRef]
  208. McCartney, D.L.; Stevenson, A.J.; Hillary, R.F.; Walker, R.M.; Bermingham, M.L.; Morris, S.W. Epigenetic signatures of starting and stopping smoking. EBioMedicine 2018, 37, 214–220. [Google Scholar] [CrossRef] [Green Version]
  209. Wilson, R.; Wahl, S.; Pfeiffer, L.; Ward-Caviness, C.K.; Kunze, S.; Kretschmer, A. The dynamics of smoking-related disturbed methylation: A two time-point study of methylation change in smokers, non-smokers and former smokers. BMC Genom. 2017, 18, 805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Joehanes, R.; Just, A.C.; Marioni, R.E.; Pilling, L.C.; Reynolds, L.M.; Mandaviya, P.R. Epigenetic Signatures of Cigarette Smoking. Circ. Cardiovasc. Genet. 2016, 9, 436–447. [Google Scholar] [CrossRef] [Green Version]
  211. Wang, G.; Wang, R.; Strulovici-Barel, Y.; Salit, J.; Staudt, M.R.; Ahmed, J. Persistence of smoking-induced dysregulation of MiRNA expression in the small airway epithelium despite smoking cessation. PLoS ONE 2015, 10, e0120824. [Google Scholar] [CrossRef] [PubMed]
  212. Parker, M.M.; Chase, R.P.; Lamb, A.; Reyes, A.; Saferali, A.; Yun, J.H. RNA sequencing identifies novel non-coding RNA and exon-specific effects associated with cigarette smoking. BMC Med. Genom. 2017, 10, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Vrijens, K.; Bollati, V.; Nawrot, T.S. MicroRNAs as potential signatures of environmental exposure or effect: A systematic review. Environ. Health Perspect. 2015, 123, 399–411. [Google Scholar] [CrossRef] [Green Version]
  214. Banerjee, A.; Luettich, K. MicroRNAs as potential biomarkers of smoking-related diseases. Biomark. Med. 2012, 6, 671–684. [Google Scholar] [CrossRef] [Green Version]
  215. National Center for Chronic Disease Prevention and Health Promotion (US) Office on Smoking and Health. The Health Consequences of Smoking—50 Years of Progress; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2014; pp. 1–36.
  216. Hindorff, L.A.; Sethupathy, P.; Junkins, H.A.; Ramos, E.M.; Mehta, J.P.; Collins, F.S. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl. Acad. Sci. USA 2009, 106, 9362–9367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Wang, T.; Xia, P.; Su, P. High-Dimensional DNA Methylation Mediates the Effect of Smoking on Crohn’s Disease. Front. Genet. 2022, 13, 302. [Google Scholar] [CrossRef]
  218. Andersen, A.M.; Lei, M.K.; Beach, S.R.H.; Philibert, R.A.; Sinha, S.; Colgan, J.D. Cigarette and cannabis smoking effects on GPR15+ helper T cell levels in peripheral blood: Relationships with epigenetic biomarkers. Genes 2020, 11, 149. [Google Scholar] [CrossRef] [Green Version]
  219. Szulakowski, P.; Crowther, A.J.L.; Jiménez, L.A.; Donaldson, K.; Mayer, R.; Leonard, T.B. The effect of smoking on the transcriptional regulation of lung inflammation in patients with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2006, 174, 41–50. [Google Scholar] [CrossRef] [Green Version]
  220. Ito, K.; Ito, M.; Elliott, W.M.; Cosio, B.; Caramori, G.; Kon, O.M. Decreased Histone Deacetylase Activity in Chronic Obstructive Pulmonary Disease. N. Engl. J. Med. 2005, 352, 1967–1976. [Google Scholar] [CrossRef] [Green Version]
  221. Chen, X.; Guan, X.-J.; Peng, X.-H.; Cui, Z.-L.; Luan, C.-Y.; Guo, X.-J. Acetylation of lysine 9 on histone H3 is associated with increased pro-inflammatory cytokine release in a cigarette smoke-induced rat model through HDAC1 depression. Inflamm. Res. 2015, 64, 513–526. [Google Scholar] [CrossRef]
  222. Sundar, I.K.; Rahman, I. Gene expression profiling of epigenetic chromatin modification enzymes and histone marks by cigarette smoke: Implications for COPD and lung cancer. Am. J. Physiol. Lung Cell. Mol. Physiol. 2016, 311, L1245–L1258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Ferraro, M.; Di Vincenzo, S.; Dino, P.; Bucchieri, S.; Cipollina, C.; Gjomarkaj, M. Budesonide, Aclidinium and Formoterol in combination limit inflammaging processes in bronchial epithelial cells exposed to cigarette smoke. Exp. Gerontol. 2019, 118, 78–87. [Google Scholar] [CrossRef]
  224. To, M.; Yamamura, S.; Akashi, K.; Charron, C.E.; Haruki, K.; Barnes, P.J. Defect of adaptation to hypoxia in patients with COPD due to reduction of histone deacetylase 7. Chest 2012, 141, 1233–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Jeong, I.; Lim, J.H.; Oh, D.K.; Kim, W.J.; Oh, Y.M. Gene expression profile of human lung in a relatively early stage of COPD with emphysema. Int. J. Chronic Obstr. Pulm. Dis. 2018, 13, 2643–2655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Sundar, I.K.; Nevid, M.Z.; Friedman, A.E.; Rahman, I. Cigarette smoke induces distinct histone modifications in lung cells: Implications for the pathogenesis of COPD and lung cancer. J. Proteome Res. 2014, 13, 982–996. [Google Scholar] [CrossRef] [Green Version]
  227. Marwick, J.A.; Kirkham, P.A.; Stevenson, C.S.; Danahay, H.; Giddings, J.; Butler, K. Cigarette smoke alters chromatin remodeling and induces proinflammatory genes in rat lungs. Am. J. Respir. Cell Mol. Biol. 2004, 31, 633–642. [Google Scholar] [CrossRef] [Green Version]
  228. Schug, T.T.; Xu, Q.; Gao, H.; Peres-da-Silva, A.; Draper, D.W.; Fessler, M.B. Myeloid Deletion of SIRT1 Induces Inflammatory Signaling in Response to Environmental Stress. Mol. Cell. Biol. 2010, 30, 4712–4721. [Google Scholar] [CrossRef] [Green Version]
  229. Lo Sasso, G.; Menzies, K.J.; Mottis, A.; Piersigilli, A.; Perino, A.; Yamamoto, H. SIRT2 deficiency modulates macrophage polarization and susceptibility to experimental colitis. PLoS ONE 2014, 9, e103573. [Google Scholar] [CrossRef]
  230. Winkler, A.R.; Nocka, K.N.; Williams, C.M.M. Smoke exposure of human macrophages reduces HDAC3 activity, resulting in enhanced inflammatory cytokine production. Pulm. Pharmacol. Ther. 2012, 25, 286–292. [Google Scholar] [CrossRef]
  231. Poralla, L.; Stroh, T.; Erben, U.; Sittig, M.; Liebig, S.; Siegmund, B. Histone deacetylase 5 regulates the inflammatory response of macrophages. J. Cell. Mol. Med. 2015, 19, 2162–2171. [Google Scholar] [CrossRef]
  232. Zhao, Y.; Ma, G.; Yang, X. HDAC5 promotes Mycoplasma pneumoniae-induced inflammation in macrophages through NF-κB activation. Life Sci. 2019, 221, 13–19. [Google Scholar] [CrossRef] [PubMed]
  233. Lappas, M. Anti-inflammatory properties of sirtuin 6 in human umbilical vein endothelial cells. Mediat. Inflamm. 2012, 2012, 597514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Chen, Y.; Wang, H.; Luo, G.; Dai, X. SIRT4 inhibits cigarette smoke extracts-induced mononuclear cell adhesion to human pulmonary microvascular endothelial cells via regulating NF-κB activity. Toxicol. Lett. 2014, 226, 320–327. [Google Scholar] [CrossRef] [PubMed]
  235. Liu, P.; Huang, G.; Wei, T.; Gao, J.; Huang, C.; Sun, M. Sirtuin 3-induced macrophage autophagy in regulating NLRP3 inflammasome activation. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 764–777. [Google Scholar] [CrossRef] [PubMed]
  236. Liang, H.; Huang, Q.; Liao, M.-J.; Xu, F.; Zhang, T.; He, J.; Zhang, L.; Liu, H.-Z. EZH2 plays a crucial role in ischemia/reperfusion-induced acute kidney injury by regulating p38 signaling. Inflamm. Res. 2019, 68, 325–336. [Google Scholar] [CrossRef] [PubMed]
  237. Izzotti, A.; Larghero, P.; Longobardi, M.; Cartiglia, C.; Camoirano, A.; Steele, V.E. Dose-responsiveness and persistence of microRNA expression alterations induced by cigarette smoke in mouse lung. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2011, 717, 9–16. [Google Scholar] [CrossRef] [PubMed]
  238. Izzotti, A.; Larghero, P.; Balansky, R.; Pfeffer, U.; Steele, V.E.; de Flora, S. Interplay between histopathological alterations, cigarette smoke and chemopreventive agents in defining microRNA profiles in mouse lung. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2011, 717, 17–24. [Google Scholar] [CrossRef] [PubMed]
  239. Qin, Z.; Wang, P.Y.; Wan, J.J.; Zhang, Y.; Wei, J.; Sun, Y. MicroRNA124-IL6R Mediates the Effect of Nicotine in Inflammatory Bowel Disease by Shifting Th1/Th2 Balance toward Th1. Front. Immunol. 2020, 11, 235. [Google Scholar] [CrossRef] [Green Version]
  240. Sun, Y.; Li, Q.; Gui, H.; Xu, D.P.; Yang, Y.L.; Su, D.F. MicroRNA-124 mediates the cholinergic anti-inflammatory action through inhibiting the production of pro-inflammatory cytokines. Cell Res. 2013, 23, 1270–1283. [Google Scholar] [CrossRef] [Green Version]
  241. Qin, Z.; Wan, J.J.; Sun, Y.; Wu, T.; Wang, P.Y.; Du, P. Nicotine protects against DSS colitis through regulating microRNA-124 and STAT3. J. Mol. Med. 2017, 95, 221–233. [Google Scholar] [CrossRef]
  242. Yan, H.; Zhang, X.; Xu, Y. Aberrant expression of miR-21 in patients with inflammatory bowel disease A protocol for systematic review and meta analysis. Medicine 2020, 99, e19693. [Google Scholar] [CrossRef] [PubMed]
  243. Shi, C.; Liang, Y.; Yang, J.; Xia, Y.; Chen, H.; Han, H. MicroRNA-21 Knockout Improve the Survival Rate in DSS Induced Fatal Colitis through Protecting against Inflammation and Tissue Injury. PLoS ONE 2013, 8, e66814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Zhang, L.; Shen, J.; Cheng, J.; Fan, X. MicroRNA-21 regulates intestinal epithelial tight junction permeability. Cell Biochem. Funct. 2015, 33, 235–240. [Google Scholar] [CrossRef]
  245. Smigielska-Czepiel, K.; van den Berg, A.; Jellema, P.; Slezak-Prochazka, I.; Maat, H.; van den Bos, H. Dual Role of miR-21 in CD4+ T-Cells: Activation-Induced miR-21 Supports Survival of Memory T-Cells and Regulates CCR7 Expression in Naive T-Cells. PLoS ONE 2013, 8, e76217. [Google Scholar] [CrossRef] [Green Version]
  246. Ruan, Q.; Wang, P.; Wang, T.; Qi, J.; Wei, M.; Wang, S. MicroRNA-21 regulates T-cell apoptosis by directly targeting the tumor suppressor gene Tipe2. Cell Death Dis. 2014, 5, e1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Ando, Y.; Mazzurana, L.; Forkel, M.; Okazaki, K.; Aoi, M.; Schmidt, P.T. Downregulation of MicroRNA-21 in colonic CD3+ T cells in UC remission. Inflamm. Bowel Dis. 2016, 22, 2788–2793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Lu, T.X.; Munitz, A.; Rothenberg, M.E. MicroRNA-21 Is Up-Regulated in Allergic Airway Inflammation and Regulates IL-12p35 Expression. J. Immunol. 2009, 182, 4994–5002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Hashimi, S.T.; Fulcher, J.A.; Chang, M.H.; Gov, L.; Wang, S.; Lee, B. MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 and WNT1 in the coordinate regulation of dendritic cell differentiation. Blood 2009, 114, 404–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Sheedy, F.J. Turning 21, Induction of miR-21 as a key switch in the inflammatory response. Front. Immunol. 2015, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  251. Chen, G.; Cao, S.; Liu, F.; Liu, Y. miR-195 plays a role in steroid resistance of ulcerative colitis by targeting Smad7. Biochem. J. 2015, 471, 357–367. [Google Scholar] [CrossRef]
  252. Zhao, Y.; Xu, Y.; Li, Y.; Xu, W.; Luo, F.; Wang, B. NF-κB-Mediated Inflammation Leading to EMT via miR-200c is involved in cell transformation induced by cigarette smoke extract. Toxicol. Sci. 2013, 135, 265–276. [Google Scholar] [CrossRef]
  253. Louis, E.; Michel, V.; Hugot, J.P.; Reenaers, C.; Fontaine, F.; Delforge, M. Early development of stricturing or penetrating pattern in Crohn’s disease is influenced by disease location, number of flares, and smoking but not by NOD2/CARD15 genotype. Gut 2003, 52, 552–557. [Google Scholar] [CrossRef] [PubMed]
  254. Flier, S.N.; Tanjore, H.; Kokkotou, E.G.; Sugimoto, H.; Zeisberg, M.; Kalluri, R. Identification of epithelial to mesenchymal transition as a novel source of fibroblasts in intestinal fibrosis. J. Biol. Chem. 2010, 285, 20202–20212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Scharl, M.; Huber, N.; Lang, S.; Fürst, A.; Jehle, E.; Rogler, G. Hallmarks of epithelial to mesenchymal transition are detectable in Crohn’s disease associated intestinal fibrosis. Clin. Transl. Med. 2015, 4, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Ananthakrishnan, A.N.; Higuchi, L.M.; Huang, E.S.; Khalili, H.; Richter, J.M.; Fuchs, C.S. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for crohn disease and ulcerative colitis. Ann. Intern. Med. 2012, 156, 350–359. [Google Scholar] [CrossRef] [PubMed]
  257. Cornish, J.A.; Tan, E.; Simillis, C.; Clark, S.K.; Teare, J.; Tekkis, P.P. The risk of oral contraceptives in the etiology of inflammatory bowel disease: A meta-analysis. Am. J. Gastroenterol. 2008, 103, 2394–2400. [Google Scholar] [CrossRef] [PubMed]
  258. Minacapelli, C.D.; Bajpai, M.; Geng, X.; Van Gurp, J.; Poplin, E.; Amenta, P.S. MiR-206 as a biomarker for response to mesalamine treatment in ulcerative colitis. Inflamm. Bowel Dis. 2019, 25, 78–84. [Google Scholar] [CrossRef] [PubMed]
  259. Adamowicz, M.; Milkiewicz, P.; Kempinska-Podhorodecka, A. 5-aminosalicylic acid inhibits the expression of oncomirs and pro-inflammatory microRNAS: An in vitro study. J. Physiol. Pharmacol. 2021, 72, 529–535. [Google Scholar] [CrossRef]
  260. He, C.; Shi, Y.; Wu, R.; Sun, M.; Fang, L.; Wu, W. MIR-301a promotes intestinal mucosal inflammation through induction of IL-17A and TNF-α in IBD. Gut 2016, 65, 1938–1950. [Google Scholar] [CrossRef] [PubMed]
  261. Nie, J.; Zhao, Q. Lnc-ITSN1-2, Derived from RNA Sequencing, Correlates with Increased Disease Risk, Activity and Promotes CD4+ T Cell Activation, Proliferation and Th1/Th17 Cell Differentiation by Serving as a ceRNA for IL-23R via Sponging miR-125a in Inflammatory Bowel Di. Front. Immunol. 2020, 11, 852. [Google Scholar] [CrossRef]
  262. Ye, Y.; Zhang, L.; Hu, T.; Yin, J.; Xu, L.; Pang, Z. CircRNA_103765 acts as a proinflammatory factor via sponging miR-30 family in Crohn’s disease. Sci Rep 2021, 11, 565. [Google Scholar] [CrossRef] [PubMed]
  263. Batra, S.K.; Heier, C.R.; Calderon, L.D.; Tully, C.B.; Fiorillo, A.A.; Van Der Anker, J. Serum mirnas are pharmacodynamic biomarkers associated with therapeutic response in pediatric inflammatory bowel disease. Inflamm. Bowel Dis. 2020, 26, 1597–1606. [Google Scholar] [CrossRef] [PubMed]
  264. Dubois-Camacho, K.; Diaz-Jimenez, D.; De la Fuente, M.; Quera, R.; Simian, D.; Martínez, M. Inhibition of miR-378a-3p by Inflammation Enhances IL-33 Levels: A Novel Mechanism of Alarmin Modulation in Ulcerative Colitis. Front. Immunol. 2019, 10, 2449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Guo, Z.; Gong, J.; Li, Y.; Gu, L.; Cao, L.; Wang, Z. Mucosal microRNAs expression profiles before and after exclusive enteral nutrition therapy in adult patients with Crohn’s disease. Nutrients 2016, 8, 519. [Google Scholar] [CrossRef]
  266. Gu, L.; Ren, F.; Fang, X.; Yuan, L.; Liu, G.; Wang, S. Exosomal MicroRNA-181a Derived from Mesenchymal Stem Cells Improves Gut Microbiota Composition, Barrier Function, and Inflammatory Status in an Experimental Colitis Model. Front. Med. 2021, 8, 898. [Google Scholar] [CrossRef]
  267. Yang, R.; Huang, H.; Cui, S.; Zhou, Y.; Zhang, T.; Zhou, Y. IFN-γ promoted exosomes from mesenchymal stem cells to attenuate colitis via miR-125a and miR-125b. Cell Death Dis. 2020, 11, 603. [Google Scholar] [CrossRef]
  268. Becker, E.; Bengs, S.; Aluri, S.; Opitz, L.; Atrott, K.; Stanzel, C. Doxycycline, metronidazole and isotretinoin: Do they modify microRNA/mRNA expression profiles and function in murine T-cells? Sci. Rep. 2016, 6, 37082. [Google Scholar] [CrossRef]
  269. Garrido-Mesa, J.; Rodríguez-Nogales, A.; Algieri, F.; Vezza, T.; Hidalgo-Garcia, L.; Garrido-Barros, M. Immunomodulatory tetracyclines shape the intestinal inflammatory response inducing mucosal healing and resolution. Br. J. Pharmacol. 2018, 175, 4353–4370. [Google Scholar] [CrossRef]
  270. Hattori, N.; Niwa, T.; Ishida, T.; Kobayashi, K.; Imai, T.; Mori, A. Antibiotics suppress colon tumorigenesis through inhibition of aberrant DNA methylation in an azoxymethane and dextran sulfate sodium colitis model. Cancer Sci. 2019, 110, 147–156. [Google Scholar] [CrossRef] [Green Version]
  271. Veltman, K.; Hummel, S.; Cichon, C.; Sonnenborn, U.; Schmidt, M.A. Identification of specific miRNAs targeting proteins of the apical junctional complex that simulate the probiotic effect of E. coli Nissle 1917 on T84 epithelial cells. Int. J. Biochem. Cell Biol. 2012, 44, 341–349. [Google Scholar] [CrossRef]
  272. Zhang, M.; Zhou, L.; Zhang, S.; Yang, Y.; Xu, L.; Hua, Z. Bifidobacterium longum affects the methylation level of forkhead box P3 promoter in 2, 4, 6-trinitrobenzenesulphonic acid induced colitis in rats. Microb. Pathog. 2017, 110, 426–430. [Google Scholar] [CrossRef] [PubMed]
  273. Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Vezza, T.; Utrilla, M.P.; Chueca, N. Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: Impact on microRNAs expression and microbiota composition. Mol. Nutr. Food Res. 2017, 61, 1700144. [Google Scholar] [CrossRef] [PubMed]
  274. Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Vezza, T.; Utrilla, M.P.; Chueca, N. Intestinal anti-inflammatory effect of the probiotic Saccharomyces boulardii in DSS-induced colitis in mice: Impact on microRNAs expression and gut microbiota composition. J. Nutr. Biochem. 2018, 61, 129–139. [Google Scholar] [CrossRef] [PubMed]
  275. Din, A.U.; Hassan, A.; Zhu, Y.; Zhang, K.; Wang, Y.; Li, T. Inhibitory effect of Bifidobacterium bifidum ATCC 29521 on colitis and its mechanism. J. Nutr. Biochem. 2020, 79, 108353. [Google Scholar] [CrossRef]
  276. Sun, S.; Xu, X.; Liang, L.; Wang, X.; Bai, X.; Zhu, L. Lactic Acid-Producing Probiotic Saccharomyces cerevisiae Attenuates Ulcerative Colitis via Suppressing Macrophage Pyroptosis and Modulating Gut Microbiota. Front. Immunol. 2021, 12, 4847. [Google Scholar] [CrossRef] [PubMed]
  277. Yu, Q.; Zhu, S.; Zhou, R.; Yi, F.; Bing, Y.; Huang, S. Effects of Sinomenine on the Expression of microRNA-155 in 2,4,6-Trinitrobenzenesulfonic Acid-Induced Colitis in Mice. PLoS ONE 2013, 8, e73757. [Google Scholar] [CrossRef] [Green Version]
  278. Wu, R.; Li, Y.; Guo, Z.; Gong, J.; Zhu, W.; Li, N. Triptolide ameliorates ileocolonic anastomosis inflammation in IL-10 deficient mice by mechanism involving suppression of miR-155/SHIP-1 signaling pathway. Mol. Immunol. 2013, 56, 340–346. [Google Scholar] [CrossRef]
  279. Changlin, Z.; Ying, Z.; Shuhua, Z.; Zeng, J.; Chi, H. Research on the Protective Effect of MiR-185-3p Mediated by Huangqin-Tang Decoction (HQT) on the Epithelial Barrier Function of Ulcerative Colitis. Evid.-Based Complement. Altern. Med. 2021, 2021, 4775606. [Google Scholar] [CrossRef]
  280. Wei, K.; Zhang, D.; Hong, J.; Zhang, C.; Feng, X.; Huang, Y. Herb-partitioned moxibustion and the miRNAs related to Crohn’s disease: A study based on rat models. Evid.-Based Complement. Altern. Med. 2015, 2015, 265238. [Google Scholar] [CrossRef]
  281. Xiong, Y.; Wang, J.; Chu, H.; Chen, D.; Guo, H. Salvianolic acid B restored impaired barrier function via downregulation of MLCK by microRNA-1 in rat colitis model. Front. Pharmacol. 2016, 7, 134. [Google Scholar] [CrossRef] [Green Version]
  282. Huang, Y.; Ma, Z.; Cui, Y.H.; Dong, H.S.; Zhao, J.M.; Dou, C.Z. Effects of Herb-Partitioned Moxibustion on the miRNA Expression Profiles in Colon from Rats with DSS-Induced Ulcerative Colitis. Evid.-Based Complement. Altern. Med. 2017, 2017, 1767301. [Google Scholar] [CrossRef] [PubMed]
  283. Hou, H.W.; Wang, J.M.; Wang, D.; Wu, R.; Ji, Z.L. Triptolide exerts protective effects against fibrosis following ileocolonic anastomosis by mechanisms involving the MIR-16-1/HSP70 pathway in IL-10-deficient mice. Int. J. Mol. Med. 2017, 40, 337–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Lv, Q.; Wang, K.; Qiao, S.; Yang, L.; Xin, Y.; Dai, Y. Norisoboldine, a natural AhR agonist, promotes Treg differentiation and attenuates colitis via targeting glycolysis and subsequent NAD+/SIRT1/SUV39H1/H3K9me3 signaling pathway article. Cell Death Dis. 2018, 9, 258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Chen, M.; Wang, J.M.; Wang, D.; Wu, R.; Hou, H.W. Triptolide inhibits migration and proliferation of fibroblasts from ileocolonic anastomosis of patients with Crohn’s disease via regulating the miR-16-1/HSP70 pathway. Mol. Med. Rep. 2019, 19, 4841–4851. [Google Scholar] [CrossRef]
  286. Zong, X.; Xiao, X.; Kai, L.; Cheng, Y.; Fu, J.; Xu, W. Atractylodis macrocephalae polysaccharides protect against DSS-induced intestinal injury through a novel lncRNA ITSN1-OT1. Int. J. Biol. Macromol. 2021, 167, 76–84. [Google Scholar] [CrossRef]
  287. Liu, H.; Liang, J.; Zhong, Y.; Xiao, G.; Efferth, T.; Georgiev, M.I. Dendrobium officinale Polysaccharide Alleviates Intestinal Inflammation by Promoting Small Extracellular Vesicle Packaging of miR-433-3p. J. Agric. Food Chem. 2021, 69, 13510–13523. [Google Scholar] [CrossRef]
  288. Huang, N.; Katz, J.P.; Martin, D.R.; Wu, G.D. Inhibition of IL-8 gene expression in Caco-2 cells by compounds which induce histone hyperacetylation. Cytokine 1997, 9, 27–36. [Google Scholar] [CrossRef]
  289. Simeoli, R.; Mattace Raso, G.; Pirozzi, C.; Lama, A.; Santoro, A.; Russo, R. An orally administered butyrate-releasing derivative reduces neutrophil recruitment and inflammation in dextran sulphate sodium-induced murine colitis. Br. J. Pharmacol. 2017, 174, 1484–1496. [Google Scholar] [CrossRef] [Green Version]
  290. Tazi, J.; Begon-Pescia, C.; Campos, N.; Apolit, C.; Garcel, A.; Scherrer, D. Specific and selective induction of miR-124 in immune cells by the quinoline ABX464, A transformative therapy for inflammatory diseases. Drug Discov. Today 2021, 26, 1030–1039. [Google Scholar] [CrossRef]
  291. Chen, Q.; Duan, X.; Xu, M.; Fan, H.; Dong, Y.; Wu, H. BMSC-EVs regulate Th17 cell differentiation in UC via H3K27me3. Mol. Immunol. 2020, 118, 191–200. [Google Scholar] [CrossRef]
  292. Wang, X.; Wang, H.; Cao, J.; Ye, C. Exosomes from Adipose-Derived Stem Cells Promotes VEGF-C-Dependent Lymphangiogenesis by Regulating miRNA-132/TGF-β Pathway. Cell Physiol. Biochem. 2018, 49, 160–171. [Google Scholar] [CrossRef]
  293. Horniblow, R.D.; Pathak, P.; Balacco, D.L.; Acharjee, A.; Lles, E.; Gkoutos, G. Iron-mediated epigenetic activation of NRF2 targets. J. Nutr. Biochem. 2022, 101, 108929. [Google Scholar] [CrossRef] [PubMed]
  294. Liu, M.; Ding, J.; Zhang, H.; Shen, J.; Hao, Y.; Zhang, X. Lactobacillus casei LH23 modulates the immune response and ameliorates DSS-induced colitis via suppressing JNK/p-38 signal pathways and enhancing histone H3K9 acetylation. Food Funct. 2020, 11, 5473–5485. [Google Scholar] [CrossRef] [PubMed]
  295. Li, Y.; Zuo, L.; Zhu, W.; Gong, J.; Zhang, W.; Guo, Z. Telmisartan attenuates the inflamed mesenteric adipose tissue in spontaneous colitis by mechanisms involving regulation of neurotensin/microRNA-155 pathway. Biochem. Pharmacol. 2015, 93, 461–469. [Google Scholar] [CrossRef] [PubMed]
  296. Mannino, G.; Caradonna, F.; Cruciata, I.; Lauria, A.; Perrone, A.; Gentile, C. Melatonin reduces inflammatory response in human intestinal epithelial cells stimulated by interleukin-1β. J. Pineal. Res. 2019, 67, e12598. [Google Scholar] [CrossRef] [PubMed]
  297. Zhang, Y.; Yan, Y.; Meng, J.; Girotra, M.; Ramakrishnan, S.; Roy, S. Immune modulation mediated by extracellular vesicles of intestinal organoids is disrupted by opioids. Mucosal Immunol. 2021, 14, 887–898. [Google Scholar] [CrossRef]
  298. Yang, Z.B.; Qiu, L.Z.; Chen, Q.; Lin, J.D. Artesunate alleviates the inflammatory response of ulcerative colitis by regulating the expression of miR-155. Pharm. Biol. 2021, 59, 97–105. [Google Scholar] [CrossRef]
  299. Felice, C.; Lewis, A.; Iqbal, S.; Gordon, H.; Rigoni, A.; Colombo, M.P. Intestinal Inflammation is Linked to Hypoacetylation of Histone 3 Lysine 27 and can be Reversed by Valproic Acid Treatment in Inflammatory Bowel Disease Patients. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 889–891.e6. [Google Scholar] [CrossRef]
  300. Chu, Y.; Zhu, Y.; Zhang, Y.; Liu, X.; Guo, Y.; Chang, L. Tetrandrine attenuates intestinal epithelial barrier defects caused by colitis through promoting the expression of Occludin via the AhR-miR-429 pathway. FASEB J. 2021, 35, e21502. [Google Scholar] [CrossRef]
  301. Gubatan, J.; Chou, N.D.; Nielsen, O.H.; Moss, A.C. Systematic review with meta-analysis: Association of vitamin D status with clinical outcomes in adult patients with inflammatory bowel disease. Aliment. Pharmacol. Ther. 2019, 50, 1146–1158. [Google Scholar] [CrossRef]
  302. McGillis, L.; Bronte-Tinkew, D.M.; Dang, F.; Capurro, M.; Prashar, A.; Ricciuto, A. Vitamin D deficiency enhances expression of autophagy-regulating miR-142-3p in mouse and “involved” IBD patient intestinal tissues. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 321, G171–G184. [Google Scholar] [CrossRef]
  303. Liu, F.-H.; Li, S.-S.; Li, X.-X.; Wang, S.; Li, M.-G.; Guan, L.; Luan, T.-G.; Liu, Z.-G.; Liu, Z.-J.; Yang, P.-C. Vitamin D3 induces vitamin D receptor and HDAC11 binding to relieve the promoter of the tight junction proteins. Oncotarget 2017, 8, 58781–58789. [Google Scholar] [CrossRef] [PubMed]
  304. Zhu, X.; Zhu, Y.; Li, C.; Yu, J.; Ren, D.D.; Qiu, S. 1,25-Dihydroxyvitamin D regulates macrophage polarization and ameliorates experimental inflammatory bowel disease by suppressing miR-125b. Int. Immunopharmacol. 2019, 67, 106–118. [Google Scholar] [CrossRef] [PubMed]
  305. Barrón-Cabrera, E.; Ramos-Lopez, O.; González-Becerra, K.; Riezu-Boj, J.I.; Milagro, F.I.; Martínez-López, E. Epigenetic Modifications as Outcomes of Exercise Interventions Related to Specific Metabolic Alterations: A Systematic Review. Lifestyle Genom. 2019, 12, 25–44. [Google Scholar] [CrossRef]
  306. White, A.J.; Sandler, D.P.; Bolick, S.C.E.; Xu, Z.; Taylor, J.A.; Deroo, L.A. Recreational and household physical activity at different time points and DNA global methylation. Eur. J. Cancer 2013, 49, 2199–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Sanchis-Gomar, F.; Garcia-Gimenez, J.L.; Perez-Quilis, C.; Gomez-Cabrera, M.C.; Pallardo, F.V.; Lippi, G. Physical exercise as an epigenetic modulator: Eustress, the “positive stress” as an effector of gene expression. J. Strength Cond. Res. 2012, 26, 3469–3472. [Google Scholar] [CrossRef] [PubMed]
  308. Pedersen, B.K.; Åkerström, T.C.A.; Nielsen, A.R.; Fischer, C.P. Role of myokines in exercise and metabolism. J. Appl. Physiol. 2007, 103, 1093–1098. [Google Scholar] [CrossRef] [Green Version]
  309. Tarnowski, M.; Kopytko, P.; Piotrowska, K. Epigenetic regulation of inflammatory responses in the context of physical activity. Genes 2021, 12, 1313. [Google Scholar] [CrossRef]
  310. Wang, J.; Liu, S.; Li, G.; Xiao, J. Exercise Regulates the Immune System. Adv. Exp. Med. Biol. 2020, 1228, 395–408. [Google Scholar] [CrossRef]
  311. Machado, O.A.S.; Diniz, V.L.S.; Passos, M.E.P.; de Oliveira, H.H.; Santos-Oliveira, L.C.; Alecrim, A.L. Physical exercise increases global and gene-specific (interleukin-17 and interferon-γ) DNA methylation in lymphocytes from aged women. Exp. Physiol. 2021, 106, 1878–1885. [Google Scholar] [CrossRef]
  312. Dorneles, G.P.; Boeira, M.C.R.; Schipper, L.L.; Silva, I.R.V.; Elsner, V.R.; Dal Lago, P. Acute Strenuous Exercise Induces an Imbalance on Histone H4 Acetylation/Histone Deacetylase 2 and Increases the Proinflammatory Profile of PBMC of Obese Individuals. Oxid. Med. Cell Longev. 2017, 2017, 1530230. [Google Scholar] [CrossRef] [PubMed]
  313. Seok, J.; Warren, H.S.; Cuenca, A.G.; Mindrinos, M.N.; Baker, H.V.; Xu, W.; Richards, D.R.; McDonald-Smith, G.P.; Gao, H.; Hennessy, L.; et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl. Acad. Sci. USA 2013, 110, 3507–3512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Saha, S.P.; Bhalla, D.K.; Whayne, T.F.; Gairola, C.G. Cigarette smoke and adverse health effects: An overview of research trends and future needs. Int. J. Angiol. 2007, 16, 77–83. [Google Scholar] [CrossRef] [Green Version]
  315. Terzikhan, N.; Verhamme, K.M.C.; Hofman, A.; Stricker, B.H.; Brusselle, G.G.; Lahousse, L. Prevalence and incidence of COPD in smokers and non-smokers: The Rotterdam Study. Eur. J. Epidemiol. 2016, 31, 785–792. [Google Scholar] [CrossRef] [Green Version]
  316. Marcin, W.; Michael, S. Genetics and epigenetics of inflammatory bowel disease. Swiss Med. Wkly. 2018, 148, w14671. [Google Scholar] [CrossRef]
  317. Wallace, K.; Grau, M.V.; Levine, A.J.; Shen, L.; Hamdan, R.; Chen, X. Association between folate levels and CpG island hypermethylation in normal colorectal mucosa. Cancer Prev. Res. 2010, 3, 1552–1564. [Google Scholar] [CrossRef] [Green Version]
  318. Ananthakrishnan, A.N.; Nguyen, D.D.; Sauk, J.; Yajnik, V.; Xavier, R.J. Genetic polymorphisms in metabolizing enzymes modifying the association between smoking and inflammatory bowel diseases. Inflamm. Bowel Dis. 2014, 20, 783–789. [Google Scholar] [CrossRef] [Green Version]
  319. Nagy-Szakal, D.; Ross, M.C.; Dowd, S.E.; Mir, S.A.V.; Schaible, T.D.; Petrosino, J.F. Maternal micronutrients can modify colonic mucosal microbiota maturation in murine offspring. Gut Microbes 2012, 3, 426–433. [Google Scholar] [CrossRef] [Green Version]
  320. Yamamoto, T.; Nakahigashi, M.; Saniabadi, A.R. Review article: Diet and inflammatory bowel disease-Epidemiology and treatment. Aliment. Pharmacol. Ther. 2009, 30, 99–112. [Google Scholar] [CrossRef]
  321. Chassaing, B.; Koren, O.; Goodrich, J.K.; Poole, A.C.; Srinivasan, S.; Ley, R.E. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 2015, 519, 92–96. [Google Scholar] [CrossRef] [Green Version]
  322. Ananthakrishnan, A.N. The exposome in inflammatory bowel disease. Trop. Gastroenterol. 2014, 35, 135–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  323. Sawan, C.; Vaissière, T.; Murr, R.; Herceg, Z. Epigenetic drivers and genetic passengers on the road to cancer. Mutat. Res. 2008, 642, 1–13. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Summary (adapted from Sawan et al. [323]). The environmental factors epigenetically influencing the occurrence of intestinal inflammation are breastfeeding, microbiota, diet, smoking habits, drugs, infections, vitamin D and physical activity. Although present at all times, it is mainly during the prenatal period, at birth and just before the onset of the disease that these factors play a key role in triggering the disease. These environmental factors, by inducing DNA methylation, histone modifications and ncRNAs in different cell types, trigger the pathways involved in IBD pathophysiology and contribute to disease initiation.
Figure 1. Summary (adapted from Sawan et al. [323]). The environmental factors epigenetically influencing the occurrence of intestinal inflammation are breastfeeding, microbiota, diet, smoking habits, drugs, infections, vitamin D and physical activity. Although present at all times, it is mainly during the prenatal period, at birth and just before the onset of the disease that these factors play a key role in triggering the disease. These environmental factors, by inducing DNA methylation, histone modifications and ncRNAs in different cell types, trigger the pathways involved in IBD pathophysiology and contribute to disease initiation.
Ijms 23 07611 g001
Table 2. Impact of the gut microbiota-derived metabolites on the epigenome in intestinal inflammation. CD, Crohn’s disease; CEBPB, CCAAT/enhancer binding protein; DNA, deoxyribonucleic acid; DSS, dextran sulfate sodium; HDAC, histone deacetylases; IBD, inflammatory bowel disease; IECs, intestinal epithelial cells; IFN, interferon; IL, interleukin; lncRNAs, long non-coding RNAs; LPS, lipopolysaccharide; MCP-1, Monocyte chemoattractant protein-1; miR, micro-RNA; NF-κB, nuclear factor-kappa B; NOD2, nucleotide-binding oligomerization domain 2; SCFAs, short-chain fatty acids; STAT, signal transducer and activator of transcription; TNF, tumor necrosis factor; UC, ulcerative colitis.
Table 2. Impact of the gut microbiota-derived metabolites on the epigenome in intestinal inflammation. CD, Crohn’s disease; CEBPB, CCAAT/enhancer binding protein; DNA, deoxyribonucleic acid; DSS, dextran sulfate sodium; HDAC, histone deacetylases; IBD, inflammatory bowel disease; IECs, intestinal epithelial cells; IFN, interferon; IL, interleukin; lncRNAs, long non-coding RNAs; LPS, lipopolysaccharide; MCP-1, Monocyte chemoattractant protein-1; miR, micro-RNA; NF-κB, nuclear factor-kappa B; NOD2, nucleotide-binding oligomerization domain 2; SCFAs, short-chain fatty acids; STAT, signal transducer and activator of transcription; TNF, tumor necrosis factor; UC, ulcerative colitis.
MetaboliteActivityEpigenetic MechanismTissue/CellsMechanismModelAuthor
SCFAs
SCFAsAnti-inflammatory activityHDACs inhibitionT cellsInhibits HDACs in T cells and increases the acetylation of p70 S6 kinase and phosphorylation rS6, regulating the mTOR pathway required for generation of Th17 (T helper type 17), Th1, and IL-10(+) T cellsC57BL/6 mice; CD4+ T cells isolated from the spleen and lymph nodesPark J, et al. (2015) [154]
SCFAsAnti-inflammatory activityHDACs inhibitionB cellsUpregulates regulatory B cells capable of producing IL-10 in a manner dependent on their HDAC inhibitory activityDSS-induced colitis in miceZou F, et al. (2021) [155]
SCFAsAnti-inflammatory activitymiR-145IECsDecreases the CEBPB expression, which could bind to the miR-145 promoter to inhibit its expression, thereby promoting the expression of DUSP6 (dual-specificity phosphatase 6) and thus prevents the development of intestinal inflammationLPS-treated intestinal epithelial cellsLiu Q, et al. (2022) [156]
Butyrate
ButyrateAnti-inflammatory activityHistone acetylationIECsButyrate, by inducing an increase in histone acetylation in the NOD2 promoter region, induces NOD2 upregulation, and impact the defence mechanism against the bacterial membrane component peptidoglycan by inducing IL-8 and GRO-alpha secretionCaco-2 cell lineLeung CH, et al. (2009) [147]
ButyrateAnti-inflammatory activityHDAC inhibitionDendritic cellsButyrate has a role of HDACi on the epigenetic modification of gene expression, inhibits IL-12 and upregulates subunit IL-23p19DSS-induced colitis in miceBerndt BE, et al. (2012) [153]
ButyrateAnti-inflammatory activityHDAC1 inhibitionT cellsButyrate inhibits HDAC1 activity to induce Fas promoter hyperacetylation and Fas upregulation in T cells and promote Fas-mediated apoptosis of T cells to eliminate the source of inflammationBALB/c miceZimmerman MA, et al. (2012) [157]
ButyrateAnti-inflammatory activityHDAC inhibitionIECsButyrate may contribute to the restoration of the tight junction barrier in IBD by affecting the expression of claudin-2, occludin, cingulin, and zonula occludens proteins (ZO-1, ZO-2) via inhibition of histone deacetylaseDSS-induced colitis in micePlöger S, et al. (2012) [146]
ButyrateAnti-inflammatory activityHistone H3 acetylationT cellsButyrate enhances histone H3 acetylation in the promoter and conserves non-coding sequence regions of the Foxp3 locus, regulating the differentiation of Treg cells, ameliorating colitisGerm-free and CRB-associated mice; OT-II (Ly5.2) transgenic CD4+ T cellsFurusawa Y, et al. (2013) [144]
ButyrateAnti-inflammatory activityHDAC inhibitionMacrophagesButyrate reduces de production of proinflammatory mediators by macrophages including nitric oxide, IL-6, and IL-12, but did not affect levels of TNF-α or MCP-1DSS-induced colitis in miceChang PV, et al. (2014) [149]
ButyrateAnti-inflammatory activityH3K9 acetylationMacrophagesButyrate activates STAT6-mediated transcription through H3K9 acetylation driving M2 macrophage polarizationDSS-induced colitis in miceJi J, et al. (2016) [150]
ButyrateAnti-inflammatory activityHistone H3 acetylationMacrophagesOral supplementation with butyrate attenuates experimental murine colitis by blocking NF-κB signaling and reverses histone acetylationDSS-induced colitis in mice, IL-10−/− mice and RAW264.7 cellsLee C, et al. (2017) [158]
ButyrateAnti or proinflammatory activity depending on its concentration and immunological milieuHDACs inhibitionT cellsLower butyrate concentrations facilitates differentiation of Tregs in vitro and in vivo under steady-state conditions. In contrast, higher concentrations of butyrate induces expression of the transcription factor T-bet in all investigated T cell subsets resulting in IFN-γ-producing Tregs or conventional T cells. This effect was mediated by the inhibition of histone deacetylase activityDSS-induced colitis in mice; CD4+ T cellsKespohl M, et al. (2017) [159]
ButyrateAnti-inflammatory activityHDAC3 inhibitionMonocyteMacrophageButyrate induces the monocyte to macrophage differentiation and promotes its antimicrobial activity and restricts bacterial translocation, through HDAC3 inhibitionHuman monocytes isolated from leukocyte cones of healthy blood donorsSchulthess J, et al. (2019) [148]
ButyrateAnti-inflammatory activityHDAC inhibitionT cellsButyrate promotes Th1 cell development by promoting IFN-γ and T-bet expression and inhibits Th17 cell development by suppressing IL-17, Rorα, and Rorγt expression and upregulate IL-10 production in Th1 and Th17CBir1 transgenic T cells; Rag1−/− miceChen L, et al. (2019) [151]
ButyrateAnti-inflammatory activityIncrease of histone acetylationIECsButyrate induces HSF2 (Heat-shock transcription factor 2) expression epigenetically via increasing histone acetylation levels at the promoter region, enhancing autophagy in IECsUC (n = 50) and healthy (n = 30) patients; DSS-induced colitis in mice; HT-29 cellsZhang F, et al. (2020) [160]
ButyrateAnti-inflammatory activityHDAC inhibitionIECsButyrate induces SYNPO (Synaptopodin) in epithelial cell lines through mechanisms possibly involving histone deacetylase inhibition. SYNPO contributes by intestinal homeostasis by controlling intestinal permeabilityEpithelial cell lines; DSS-induced colitis in miceWang RX, et al. (2020) [161]
ButyrateAnti-inflammatory activityHDAC inhibitionNeutrophilsButyrate significantly inhibits IBD neutrophils to produce proinflammatory cytokines, chemokines, and calprotectins through HDAC inhibitionPeripheral neutrophils isolated from IBD patients and healthy donors; DSS-induced colitis in miceLi G, et al. (2021) [152]
PropionateAnti-inflammatory activityHDAC1 inhibitionIECsPropionate promotes intestinal epithelial cell migration by enhancing cell spreading and polarization, a function dependant of the inhibition of class I HDACMouse small intestinal epithelial cells (MSIE) and human Caco-2 cells; DSS-induced colitis in miceBilotta AJ, et al. (2021) [142]
Caprylic acid (C8) and nonanoic acid (C9) (medium chain fatty acids)Anti-inflammatory activityAcetylation of histone 3 lysine 9 (H3K9)IECsReduces bacterial translocation, enhances antibacterial activity, and attenuates the activity of the classical histone deacetylase pathway to facilitate the acetylation of histone 3 lysine 9 (H3K9) at the promoters pBD-1 and pBD-2, remarkably increases the secretion of porcine β-defensins 1 (pBD-1) and pBD-2Porcine jejunal epithelial cell line-J2Wang J, et al. (2018) [162]
Table 4. Impact of drugs on epigenome in intestinal inflammation. 5-ASA, 5-aminosalicylic acid; CD, Crohn’s disease; circRNA, circular RNA; CpG, CpG, cytosine–phosphate–guanine; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; EEN, Exclusive enteral nutrition; EMT, epithelial-to-mesenchymal-transition; EV, extracellular vesicle; HDAC, histone deacetylases; HPM, Herb-partitioned moxibustion; IBD, inflammatory bowel disease; IEC, intestinal epithelial cell; IFN, interferon; IL, interleukin; lncRNA, long non-coding RNA; LPS, lipopolysaccharide; miR, micro-RNA; MSC, mesenchymal stem cell; PPAR-γ, peroxisome proliferator-activated receptor γ; RNA, ribonucleic acid; SCFAs, short-chain fatty acids; SNIP1, Smad Nuclear Interacting Protein 1; STAT, signal transducer and activator of transcription; TLR, toll-like receptor; TNF, tumor necrosis factor; TNBS, 2,4,6-Trinitrobenzene sulfonic acid; UC, ulcerative colitis; VEGF, Vascular Endothelial Growth Factor; ZO, zonula occludens.
Table 4. Impact of drugs on epigenome in intestinal inflammation. 5-ASA, 5-aminosalicylic acid; CD, Crohn’s disease; circRNA, circular RNA; CpG, CpG, cytosine–phosphate–guanine; DNA, deoxyribonucleic acid; DNMT, DNA methyltransferase; DSS, dextran sulfate sodium; EEN, Exclusive enteral nutrition; EMT, epithelial-to-mesenchymal-transition; EV, extracellular vesicle; HDAC, histone deacetylases; HPM, Herb-partitioned moxibustion; IBD, inflammatory bowel disease; IEC, intestinal epithelial cell; IFN, interferon; IL, interleukin; lncRNA, long non-coding RNA; LPS, lipopolysaccharide; miR, micro-RNA; MSC, mesenchymal stem cell; PPAR-γ, peroxisome proliferator-activated receptor γ; RNA, ribonucleic acid; SCFAs, short-chain fatty acids; SNIP1, Smad Nuclear Interacting Protein 1; STAT, signal transducer and activator of transcription; TLR, toll-like receptor; TNF, tumor necrosis factor; TNBS, 2,4,6-Trinitrobenzene sulfonic acid; UC, ulcerative colitis; VEGF, Vascular Endothelial Growth Factor; ZO, zonula occludens.
DrugActivityEpigenetic MechanismTissue/CellsMechanismModelAuthor
IBD medication
MesalamineAnti-inflammatory activitymiR-206IECs and colonic tissuesLong-term treatment donw-regulates miR-206 which confer a protective effect in inducing and maintaining histologic remissionHT29 colon cells; UC patients (n = 10)Minacapelli CD, et al. (2019) [258]
5-ASAAnti-inflammatory activitymiR-125b, miR-150, miR-155, miR-346 and miR-506IECs5-ASA suppressed the levels of miR-125b, miR-150, miR-155, miR-346 and miR-506 in IECs and inhibition of these miR were associated with significant inductions of their target genes such as vitamin D receptor (VDR), suppressor of cytokine signaling (SOCS1), Forkhead box O (FOXO3a) and DNA methyltransferase 1 (DNMT1)Caco-2 cellsAdamowicz M, et al. (2021) [259]
InfliximabAnti-inflammatory activitymiR-10aDCsAnti-TNF mAb treatment significantly promote miR-10a expression, whereas it markedly inhibited NOD2 and IL-12/IL-23p40 in the inflamed mucosaHuman monocyte-derived dendritic cells (DC); IBD patientsWu W, et al. (2015) [127]
InfliximabAnti-inflammatory activitymiR-301aT cellsDecreases miR-301a expression in IBD CD4+ T cells by decreasing Th17 cell differentiation through upregulation of SNIP1Peripheral blood mononuclear cells (PBMC); inflamed mucosa of patients with IBDHe C, et al. (2016) [260]
InfliximabAnti-inflammatory activitylnc-ITSN1-2T cellsLnc-ITSN1-2 promotes IBD CD4+ T cell activation, proliferation, and Th1/Th17 cell differentiation by serving as a competing endogenous RNA for IL-23R via sponging miR-125aIntestinal mucosa from IBD patients (n = 6) and healthy controls (n = 6)Nie J, et al. (2020) [261]
InfliximabAnti-inflammatory activitymiR-30 familyIECsDecreases circRNA_103765 expression, which act as a molecular sponge to adsorb the miR-30 family and impair the negative regulation of Delta-like ligand 4 (DLL4) and protect human IECs from TNF-α-induced apoptosisIBD patients; PBMCsYe Y, et al. (2021) [262]
InfliximabAnti-inflammatory activitymiR-146a and miR-146bSerum and intestinal mucosaeDecreases miR-146a and miR-146b levels in serum. miR-146a probably promotes colitis through TLR4/MyD88/NF-κB signaling pathwaySerum of 19 IBD patientsBatra SK, et al. (2020) [263]
Infliximab (IFX) therapy and longer-term steroids (weeks)Anti-inflammatory activitymiR-320aDecreases miR-320a serum level. miR-320a could play a role in sensitization of the quiescent mucosa to environmental factorsSerum of 19 IBD patients
Anti-TNF and glucocorticoidsAnti-inflammatory activitylet-7clet-7c serum level decreases, thus reduces M2 macrophage polarization (anti-inflammatory) and promote M1 (proinflammatory) polarizationSerum of 19 IBD patients
Anti-TNFAnti-inflammatory activitymiR-10aDCsBlockade TNF with anti-TNF mAb markedly enhances miR10a expression in the intestinal mucosa. miR-10a could block intestinal inflammation and reduce the differentiation Th1 and Th17C57BL/6 (B6) miceXue X, et al. (2011) [125]
Anti-TNFAnti-inflammatory activitymiR-378a-3p, miR-378cColonic mucosaeIncreases levels of miR-378a-3p and miR-378c. Over-expression of miR-378a-3p decreased the levels of an IL-33 target sequence β-gal-reporter geneActive UC patients (n = 24); inactive UC (n = 10); controls (n = 6); HEK293 cellsDubois-Camacho K, et al. (2019) [264]
Enemas containing short chain fatty acids (SCFA) such as butyrate, propionate, and acetateAnti-inflammatory activityHistone acetylationIECsSCFAs increase histone acetylation states and inhibit the production of proinflammatory substances, such as IL-8, by the intestinal epitheliumCaco-2 cellsHuang N, et al. (1997) [288]
N-(1-carbamoyl-2-phenylethyl) butyramide (FBA), a butyrate-releasing derivativeAnti-inflammatory activityHistone deacetylase-9 and H3 histone acetylationColonic mucosaeFBA, similar to its parental compound sodium butyrate, inhibited histone deacetylase-9 and restored H3 histone acetylation, exerting an anti-inflammatory effect through NF-κB inhibition and the upregulation of PPARγDSS-induced colitis in miceSimeoli R, et al. (2017) [289]
Exclusive enteral nutrition (EEN)Anti-inflammatory activityhsa-miR-192-5p, hsa-miR-423-3p, hsa-miR-99a-5p, hsa-miR-124-3p, hsa-miR-301a-5p, hsa-miR-495-5p, and hsa-let-7b-5pIntestinal mucosaeEEN induces mucosal miRNAs expression profile (altered expressions of hsa-miR-192-5p, hsa-miR-423-3p, hsa-miR-99a-5p, hsa-miR-124-3p, hsa-miR-301a-5p, hsa-miR-495-5p, and hsa-let-7b-5p) after EEN therapy was significantly changed compared with inflamed mucosa before treatmentCD patients (n = 30)Guo Z, et al. (2016) [265]
ABX464Anti-inflammatory activitymiR-124Immune cellsUpregulates miR-124 in human immune cells, which is a negative regulator of inflammation and was shown to target RNAs, such as STAT and TLR Tazi J, et al. (2021) [290]
MSCsAnti-inflammatory activitymiR-181aIECsMSC-derived exosomal miR-181a could alleviate colitis by promoting intestinal barrier function decreased (increasing level of Claudin-1, ZO-1, and IκB)DSS-induced colitis in mice and induced human colonic epithelial cell (HCOEPIC)Gu L, et al. (2021) [266]
MSCsAnti-inflammatory activityH3K27me3T cellsExtracellular vesicles from MSCs could inhibit the differentiation of Th17 cells by regulating H3K27me3TNBS-induced colitis in miceChen Q, et al. (2020) [291]
IFN-γ pretreated bone marrow mesenchymal stem cellsAnti-inflammatory activitymiR-125a and miR-125bT cellsIncreases the level of miR-125a and miR-125b of exosomes, which directly targeted on Stat3, to repress Th17 cell differentiationDSS-induced colitis in miceYang R, et al. (2020) [267]
Vascular endothelial growth factor-C-treated adipose-derived stem cells (ADSCs)Anti-inflammatory activitymiR-132Lymphatic endothelial cellsVEGF-C-treated ADSCs have a higher level of miR-132, which promotes lymphangiogenic response by directly targeting Smad-7 and regulating TGF-β/Smad signalingLymphatic endothelial cells (LECs)Wang X, et al. (2018) [292]
Supplementation
IronProinflammatory activityTET1 induction; NRF2, NQ01, GPX2 demethylationIECs and intestinal mucosaeChronic iron exposure leads to induction of TET1 expression leading to demethylation of NRF2 (nuclear factor erythroid 2-related factor 2) pathway targets (including NAD(P)H Quinone Dehydrogenase 1 (NQO1) and Glutathione peroxidase 2 (GPX2). NQO1 and GPX2 hypomethylation led to increased gene and protein expression, and could be a route by which cells overcome persistent and chronic oxidative stressCaco-2 cells and wild-type C57BL/6 miceHorniblow RD, et al. (2022) [293]
Antibiotics
IsotretinoinAnti-inflammatory activitymiRT cells3 miR overexpressed in naive T-cells and potentially downregulate 777 miR targets (cytoskeleton remodelling and the c-Jun N terminal kinase (JNK) signaling pathway)Balb/c miceBecker E, et al. (2016) [268]
MetronidazoleAnti-inflammatory activitymiR5 miR were significantly lower in naive T-cells resulting in the prediction of 340 potentially upregulated miR targets associated with IL-2 activation and signaling, cytoskeleton remodelling and epithelial-to-mesenchymal-transition (EMT).
DoxycyclineAnti-inflammatory activitymiR-144-3pOverexpression of miR-144-3p that resulted in the prediction of 493 potentially downregulated miR targets involved in protein kinase A (PKA), protein kinase B and nuclear factor of activated T-cells (NFAT) signaling pathways
TetracyclinesAnti-inflammatory activitymiR-150, miR-155, miR-375 and miR-146Colonic tissuesReduce miR-150 and miR-155 expression, upregulate miR-375 and miR-142DSS-induced colitis in mice and bone marrow-derived macrophagesGarrido-Mesa J, et al. (2018) [269]
Antibiotics treatmentAnti-inflammatory activityDNA demethylationIECsSuppresses aberrant DNA methylation of three marker CpG islands (Cbln4, Fosb, and Msx1) induced by chronic inflammationAOM/DSS-induced colitis in miceHattori N, et al. (2019) [270]
Probiotics
Probiotic bacterium Escherichia coli Nissle 1917 (EcN)Anti-inflammatory activitymiR-203, miR-483-3p, miR-595IECsIncreases miR-203, miR-483-3p, miR-595 targeting tight junction (TJ) proteins; these miRNAs are involved in the regulation of barrier function by modulating the expression of regulatory and structural components of tight junctional complexes.T84 cellsVeltman K, et al. (2012) [271]
Bifidobacterium longumAnti-inflammatory activityDNA demethylationPeripheral blood mononuclear cellsB. Longum treatment significantly demethylates several CpG sites in Foxp3 promoterTNBS-induced colitis in rat; spleen peripheral blood mononuclear cells (PBMC) cells was extractedZhang M, et al. (2017) [272]
Lactobacillus fermentum and Lactobacillus salivariusAnti-inflammatory activitymiR-155, miR-223, miR-150 and miR-143Colonic tissuesThey increase the expression of miR-155 and miR-223, and miR-150 and miR-143 for L. fermentum, involved in the immune response (restoration of Treg cell population and the Th1/Th2 cytokine balance) and in the intestinal barrier functionC57BL/6J miceRodríguez-Nogales A, et al. (2017) [273]
Saccharomyces boulardiiAnti-inflammatory activitymiR-155 and miR-223; miR-143 and miR-375Colonic tissuessIncreasing the expression of miR-155 and miR-223, whereas decreasing the expression miR-143 and miR-375DSS-induced colitis in miceRodríguez-Nogales A, et al. (2018) [274]
Bifidobacterium bifidum ATCC 29521Anti-inflammatory activitymiR-150, miR-155, miR-223Colonic mucosaeRestorates miR-150, miR-155, miR-223, upregulates anti-inflammatory cytokines (IL-10, PPARγ, IL-6), tight junction proteins (such as ZO-1, MUC-2, Claudin-3, and E Cadherin-1) and downregulates inflammatory genes (TNF-α, IL-1β)DSS-induced colitis in miceDin AU, et al. (2020) [275]
Lactobacillus casei LH23 probioticAnti-inflammatory activityHistone H3K9 acetylationColonic tissuesModulates the immune response and ameliorates colitis via suppressing JNK/p-38 signal pathways and enhancing histone H3K9 acetylationDSS-induced colitis in mice; LPS-induced RAW264.7 cellsLiu M, et al. (2020) [294]
Lactic Acid-Producing Probiotic Saccharomyces cerevisiaeAnti-inflammatory activityHistone H3K9 acetylation and histone H3K18 lactylationMacrophagesPromotes histone H3K9 acetylation and histone H3K18 lactylation and attenuates intestinal inflammation via suppressing macrophage pyroptosisDSS-induced colitis in miceSun S, et al. (2021) [276]
Other medication
Telmisartan (angiotensin II type 1 receptor blocker and a peroxisome proliferator-activated receptor-receptor-γ agonist)Anti-inflammatory activitymiR-155Mesenteric adipocytesRestorates the mesenteric adipose tissue adipocyte morphology and the expression of adipokines by suppressing the neurotensin/miR-155 pathwayIL-10(−)/(−) mice; cultured mesenteric adipose tissue from Crohn’s disease patientsLi Y, et al. (2015) [295]
MelatonineAnti-inflammatory activityPrevent DNA methylationIECsPrevents DNA demethylation, reduces NF-κB activation, decreases the levels of inflammatory mediators (including IL-6, IL-8, COX-2, and NO), and reduces increase in paracellular permeability, attenuating the inflammatory responseCaco-2 cellsMannino G, et al. (2019) [296]
MorphineProinflammatory activityLet7c-5pMacrophages, DCsOpioid treatment can disrupt gut immune homeostasis by inhibiting packaging of miR into EVs secreted by intestinal crypt cells (with a decreased amount of Let7c-5p)C57BL/6J mice; organoid cultureZhang Y, et al. (2021) [297]
ArtesunateAnti-inflammatory activitymiR-155MacrophagesInhibits the expression of miR-155 to inhibit the NF-κB pathwayLPS-induced RAW264.7 cells; BALB/c mice modelYang ZB, et al. (2021) [298]
Valproic acid treatmentAnti-inflammatory activityHDAC inhibitionIntestinal tissueInhibits HDAC activity and increases H3K27ac levels and reduced expression of IL6, IL10, IL1B, and IL23DSS-induced colitis in miceFelice C, et al. (2021) [299]
TetrandrineAnti-inflammatory activitymiR-429IECsTetrandrine can attenuate the intestinal epithelial barrier defects in colitis through promoting occludin expression via the AhR/miR-429 pathwayDSS-induced colitis in miceChu Y, et al. (2021) [300]
Chinese medicine
Sinomenine, a pure alkaloid isolated in Chinese medicineAnti-inflammatory activitymiR-155Colonic tissuesDownregulates the levels of miR-155 and several related inflammatory cytokinesTNBS-induced colitis in miceYu Q, et al. (2013) [277]
Tripterygium wilfordii Hook F (TWHF)Anti-inflammatory activitymiR-155Ileocolonic anastomosisTriptolide could suppress miR-155/SHIP-1 signaling pathway and attenuated expression of inflammatory cytokines after ileocaecal resectionIL-10(−/−) miceWu R, et al. (2013) [278]
Herb-partitioned moxibustion (HPM)Anti-inflammatory activitymiR-147 and miR-205Colonic tissuesUpregulates the expression of miR-147 and miR-205 and then further regulate some of their target genes, thereby indirectly inhibiting the inflammatory signal pathways mediated by TLR, NF-κB, and so forth and decreasing the production of downstream inflammatory cytokines such as TNF-α and IL-1β, so as to alleviate intestinal inflammation in CDExperimental CD rat modelsWei K, et al. (2015) [280]
Salvianolic acid B (Sal B) is isolated from the traditional Chinese medical herb Salvia miltiorrhizaAnti-inflammatory activitymiR-1IECsSal B restores barrier function by miR-1 activation and subsequent myosin light chain kinase (MLCK) inactivationTNBS-induced rat colitis modelXiong Y, et al. (2016) [281]
Herb-partitioned moxibustion (HPM)Anti-inflammatory activitymiR-184 and miR-490-5pColonic tissueHPM regulates miR-184 and miR-490-5p expression, act on the transcription of their target genes to regulate inflammatory signaling pathways, and attenuate inflammation and tissue injury in the colons of rats with DSS-induced UCDSS-induced colitis in miceHuang Y, et al. (2017) [282]
Triptolide (TPL)Anti-inflammatory activitymiR-16-1Ileocolonic anastomosisTPL reduces miR-16-1 levels aggravating anastomotic inflammation and fibrosisIL-10−/− miceHou HW, et al. (2017) [283]
Norisoboldine (NOR), a natural aryl hydrocarbon receptor (AhR)Anti-inflammatory activityH3K9me3 modificationT cellsNOR promoted Treg differentiation and then alleviated the development of colitis by regulating AhR (aryl hydrocarbon receptor)/glycolysis axis and decreases NAD+ and SIRT1 (sirtuin 1), facilitates the ubiquitin-proteasomal degradation of SUV39H1, which is a major member of histone KMTs and catalyses the H3K9me3 modification, which is associated with transcription repression of Foxp3 Lv Q, et al. (2018) [284]
Triptolide (TPL), the most potent bioactive substance in TWHF (Tripterygium wilfordii Hook F) extractAnti-inflammatory activitymiR-16-1FibroblastsInhibits migration and proliferation of fibroblasts from ileocolonic anastomosis of CD patients via regulating the miR-16-1/HSP70 pathwayFibroblasts from strictured anastomosis tissue (SAT) samples and matched anastomosis-adjacent normal tissue (NT) samples of CD patients (n = 10)Chen M, et al. (2019) [285]
Polysaccharide RAMPtp from Atractylodis macrocephalae KoidzAnti-inflammatory activitylncRNA ITSN1-OT1IECsInduces lncRNA ITSN1-OT1, which blocks the nuclear import of phosphorylated STAT2 and prevents the decrease of expression and structural destroy of tight junction proteinsDSS-induced colitis in miceZong X, et al. (2021) [286]
Dendrobium officinale polysaccharide (DOP)Anti-inflammatory activitymiR-433-3pIECs, macrophagesDOP interfered with the secretion of small extracellular vesicles (DIEs) by IEC, with increased miR-433-3p expression. When delivered to macrophages, miR-433-3p targeted the MAPK8 gene, leading to inhibition of the MAPK signaling pathway and reduced production of inflammatory cytokinesIECs, macrophagesLiu H, et al. (2021) [287]
Huangqin-Tang decoction (HQT)Anti-inflammatory activitymiR-185-3pIECsHQT could upregulate miR-185-3p, thereby affecting the myosin light chain kinase (MLCK)/myosin light chain phosphorylation (p-MLC) pathway and leading to increased expression of occludin protein, which ultimately protected the intestinal epithelial barrier functionBalb/c miceChanglin Z, et al. (2021) [279]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vieujean, S.; Caron, B.; Haghnejad, V.; Jouzeau, J.-Y.; Netter, P.; Heba, A.-C.; Ndiaye, N.C.; Moulin, D.; Barreto, G.; Danese, S.; et al. Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models. Int. J. Mol. Sci. 2022, 23, 7611. https://doi.org/10.3390/ijms23147611

AMA Style

Vieujean S, Caron B, Haghnejad V, Jouzeau J-Y, Netter P, Heba A-C, Ndiaye NC, Moulin D, Barreto G, Danese S, et al. Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models. International Journal of Molecular Sciences. 2022; 23(14):7611. https://doi.org/10.3390/ijms23147611

Chicago/Turabian Style

Vieujean, Sophie, Bénédicte Caron, Vincent Haghnejad, Jean-Yves Jouzeau, Patrick Netter, Anne-Charlotte Heba, Ndeye Coumba Ndiaye, David Moulin, Guillermo Barreto, Silvio Danese, and et al. 2022. "Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models" International Journal of Molecular Sciences 23, no. 14: 7611. https://doi.org/10.3390/ijms23147611

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

Article Metrics

Back to TopTop