Abstract
Environmental triggers in the context of genetic susceptibility drive phenotypes of complex immune disorders such as Inflammatory bowel disease (IBD). One such trigger of IBD is perturbations in enteric commensal bacteria, fungi or viruses that shape both immune and neuronal state. The epigenome acts as an interface between microbiota and context-specific gene expression and is thus emerging as a third key contributor to IBD. Here we review evidence that the host epigenome plays a significant role in orchestrating the bidirectional crosstalk between mammals and their commensal microorganisms. We discuss disruption of chromatin regulatory regions and epigenetic enzyme mutants as a causative factor in IBD patients and mouse models of intestinal inflammation and consider the possible translation of this knowledge. Furthermore, we present emerging insights into the intricate connection between the microbiome and epigenetic enzyme activity via host or bacterial metabolites and how these interactions fine-tune the microorganism-host relationship.
Epigenetics as a contributor to immunological disease, including IBD
DNA is organized in each cell nucleus by interacting with histones to form chromatin. Chromatin is regulated by modifications on DNA or histones and protein complexes that remodel its architecture [1]. At the interface of the environment and gene expression, chromatin alterations enable cell type-specific and kinetic regulation of gene expression from identical DNA sequences and are essential for proper cell development and function. More than 100 unique chromatin modifications have been identified, including common ones like methylation, acetylation, phosphorylation, and less studied modifications like proprionylation, butyrylation, crotonylation, and succinylation [1,2]. These modifications are regulated by over 400 chromatin modifying enzymes. Epigenetic ‘writers’ catalyze post-translational modifications on histones or DNA (e.g. DNA and histone methyltransferases, histone acetyltransferases), while ‘erasers’ remove these dynamic modifications (e.g. demethylases, histone deacetylases (HDACs), TET proteins). Finally, epigenetic modifications are interpreted by ‘readers’, that dock to defined modified histones via distinct protein domains (e.g. bromodomain, PHD, YEATS) and recruit appropriate transcriptional machinery to facilitate or prevent transcription [1].
Genome wide association studies (GWAS) have identified over 200 genetic susceptibility loci for IBD [3] comprising Crohn’s disease (CD) and Ulcerative Colitis (UC). Many of these mutations result in loss-of-function variants of immune components that are normally responsible for protective responses to enteric microorganisms, emphasizing the importance of the host-microorganism relationship for intestinal homeostasis [3]. However, the majority of susceptibility gene mutations contribute individually to IBD with low odds ratio [3], have low concordance rates in monozygotic twins [3] and are in non-coding regions [4]. Moreover, the rapid rise in the incidence of complex immune disorders such as IBD indicates that additional factors to genetics must contribute to disease. Recent studies in monozygotic twins showed that by 65 years of age almost 70% of the immune epigenome is environmentally derived [5], thus demonstrating the profound impact of non-heritable environmental influences on immune output and disease susceptibility. Furthermore, dysregulation of epigenetic enzymes and chromatin regulatory processes is an established event in human disease, but current knowledge is limited mostly to cancer. This has made proteins that regulate the epigenome some of the most promising and intently pursued targets in drug discovery today [6,7]; yet, a critical gap exists in our understanding of altered epigenetic enzymes or chromatin that result in immune disorders, including IBD. Chromatin alterations have been consistently observed in IBD patients and recent GWAS studies have reveals mutants of certain epigenetic enzymes significantly associated with IBD susceptibility. Furthermore, intestinal microorganisms and their metabolites are increasingly being recognized for their ability to alter activity of host chromatin modifying enzymes in order to immunomodulate and affect disease susceptibility. A greater mechanistic understanding of epigenetics in IBD will illuminate important, potentially reversible, mechanisms that link genetic predisposition and intestinal microorganisms in the pathogenesis of disease.
Epigenetic alterations as a trigger and target in intestinal inflammation?
Numerous genome wide methylation profiling studies in humans have shown aberrant DNA methylation in sera and intestinal tissues of young and adult IBD patients [8•,9,10]. In addition, histone methylation profiling of intestinal epithelial cells (IECs) from a CD cohort revealed differences in H3K4me3 signatures [11], with enriched H3K4me3 patterns in genes associated with immune processes to bacterial factors, cytokine signaling and reduced H3K4me3 signatures associated with intestinal epithelial ion absorption and lipid and protein metabolism [11]. Moreover, some studies revealed the highly differentially methylated or histone modified-genes were in fact for IBD risk loci, connecting possible epigenetic signatures with predisposed risk loci [9]. However, it is often challenging in human studies to cleanly separate correlation from causation.
In mice, demonstration that perturbations of individual epigenetic enzymes profoundly influence both immune and intestinal homeostasis in vivo has provided some evidence of causation. Loss of DNA ‘writers’ (DNMT1), histone ‘writers’ (SETDB1, EZH2, ASH1L), DNA ‘erasers’ (MBD2, TET), histone ‘erasers’ (HDACs 2, 3, 7), histone ‘readers’ (UHRF1, TRIM28, SP140) or chromatin remodelers (SMARCA4, PBRM1) disrupted cell identity and prevented appropriate integration of microbiota cues and maintenance of intestinal homeostasis (Table 1). Importantly, deletion of some epigenetic enzymes protected in models of IBD and thus may offer some therapeutic avenues for amelioration of intestinal inflammation. For instance, deletion of histone ‘writers’ (G9a, KAT8), histone ‘erasers’ (JMJD3, KDM2B, HDAC 6, 9, 10) or chromatin remodeler SMARCD1 protected in various models of IBD by altering the T regulatory (Treg):T helper (Th)17 ratio, suppressing macrophages or promoting protective IEC functions (Table 1).
Table 1.
Epigenetic regulators in mouse IBD models
Gene/Protein | Epigenetic enzyme function | Mouse disease phenotype | Cell(s) Implicated |
---|---|---|---|
Exacerbated intestinal inflammation | |||
DNMT1 | Writer of DNA methylation | Spontaneous early epithelial and crypt damage with IEC specific deletion; however, recover with Dnmt3b compensation. Dnmt1-Dnmt3b double IEC KO don’t recover [12] | IEC |
SETDB1 | Writer of H3K9me3 | Spontaneous and lethal terminal ileitis and colitis with IEC specific deletion [13••] | IEC |
EZH2 | Writer of H3K27me3 | ↑ DSS and TNBS colitis with IEC specific deletion [14] | IEC |
ASH1L or KMT2H | Writer and Reader of H3K4me, H3K36me and H3K9me1 | ↑ TNBS colitis and adoptive transfer colitis model with deletion [15] | Treg |
MBD2 | Eraser and Reader of CpG island Hemi-methylated DNA |
↑ DSS colitis with deletion [16] | IEC DC T cells |
TET2 | Eraser of DNA to produce 5-hydroxymethylcytosine | ↑ DSS colitis with deletion [17] | Macrophages DCs |
HDAC2 | Eraser | ↑ DSS colitis with IEC specific deletion [18] ↑ Adoptive T cell transfer colitis and TNBS colitis model with deletion in CD4 T cells [19] |
IEC Th17 |
HDAC3 | Eraser | ↑ Adoptive T cell transfer colitis with Treg specific deletion [20] ↑ DSS colitis with IEC specific deletion [21] ↑ Citrobacter rodentium infection with IEC specific deletion [22•] |
Treg IEC |
HDAC7 | Eraser | Mice with phosphorylation deficient human HDAC7 (HDAC7-ΔP) developed spontaneous autoimmunity [23] | iNKT |
SIRT6 | Eraser of H3K9Ac and H3K56Ac | ↑ DSS colitis with IEC-specific deletion [24] | IEC |
UHRF1 | Reader of H3K9me2/ 3 Hemi-methylated DNA |
Spontaneous development of colitis with CD4 T cell specific deletion [25] ↑ DSS colitis with myeloid specific deletion or mutation in the DNA methylation site (Uhrf1YP187/188AA) [26] |
Treg Macrophages |
TRIM28 | Reader H3K9me3 |
Autoimmunity with global T cell deletion [27,28] ↓ Adoptive transfer T cell colitis with CD4+ T cell deletion [29] |
Treg |
SP140 | Reader | ↑ DSS colitis with hematopoietic knockdown [30••] | Macrophages |
SMARCA4 or BRG1 | Chromatin remodeler, Reader | Spontaneous colitis with IEC specific deletion and ↑ DSS colitis [31] Spontaneous colitis with ILC3 specific deletion of Rag1−/− background [32] |
IEC ILC3 |
PBRM1/Baf180 | Chromatin remodeler, Reader | ↑ Citrobacter rodentium infection with IEC deletion [33] | IEC |
Protected from intestinal inflammation | |||
G9a | Writer of H3K9me1/2 | ↓ Adoptive T cell transfer colitis with G9a deletion in CD4+ T cells [34] | Th17 Treg |
KAT8/Mof | Writer of H416ac | ↓ DSS colitis with conditional deletion [35] | Th17 |
JMJD3 | Eraser of H3K27me3 | ↓ Adoptive T cell transfer colitis with CD4+ T cell specific deletion [36] | Th17 |
KDM2B/JHDM1B | Eraser of H3K4me3 and H3K36me3 | ↓ DSS colitis with deletion [37] | Macrophages DCs |
HDAC6 | Eraser | ↓ Adoptive T cell transfer colitis with Treg specific deletion [38] | Treg |
HDAC9 | Eraser | ↓ DSS colitis with complete deletion [39] ↓ Adoptive T cell transfer colitis with Treg specific deletion [40] |
Treg |
HDAC10 | Eraser | ↓ Adoptive T cell transfer colitis with Treg specific deletion [41] | Treg |
SMARCD1 or BAF60A | Chromatin remodeler | ↓ DSS colitis with IEC specific deletion [42•] | IEC |
Genetic variation within epigenetic regulators and regulatory regions associate with IBD
In humans, variants of epigenetic regulators or regulatory regions are emerging as direct contributors to IBD susceptibility. Of the 90% of IBD-associated genetic variants that occur in non-coding regions, many are in chromatin regulatory regions, such as immune cell enhancers marked by H3K27ac, and often near sites for master regulators for immune cell identity, differentiation, and stimulus-specific gene expression [4]. Indeed, an IBD risk loci in a distal enhancer of CD4+ Tregs was necessary for limiting colitis [43••]. Furthermore, genetic variants within loci that code for three epigenetic regulators: SP140, DNMT3A, and DNMT3B have been found to associate with increased susceptibility to IBD using GWAS (Table 2). Fine mapping GWAS and studies in non-European targeted cohorts have identified newer risk loci associated with epigenetic regulators, such as Baz1a and HDAC11. In addition, profiling of colonic biopsy samples have revealed epigenetic regulators, EZH2, SIRT6, ASH1L, SETDB1 and BRG1, which were identified to have colitis phenotypes in mice (Table 1) had reduced expression levels in human IBD [13••,14,24,31].
Table 2.
Variants of epigenetic regulators that associate with susceptibility of IBD
Gene | IBD-associated SNP | Epigenetic enzyme subclass | Protein domain/Function | Validated or predicted outcome of SNP | Validated in mice |
---|---|---|---|---|---|
SP140 | rs6716753 [45–47] rs28445040 [45,46] rs7423615 [46,48] |
Reader | SAND, PHD Bromodomain |
Validated Loss-of-function | YES |
DNMT3A | rs13428812 [48] rs201014116 [49] |
Writer | DNA methyltransferase | Unknown | NO |
DNMT3B | rs4911259 [45,47] rs6058869 [50] rs6087990 [51,52] rs1474738 [52] |
Writer | DNA methyltransferase | Unknown | NO |
BAZ1A | rs712303 [49] | Reader, Chromatin Remodeler | Bromodomain | Unknown | NO |
BRD2 | rs1049526 [53] | Reader | Bromodomain | Predicted loss-of-function | NO |
HDAC7 | rs11168249[45] | Eraser | Histone deacetylase | Unknown | NO |
HDAC11 | rs2655211[54] | Eraser | Histone deacetylase | Unknown | NO |
Of the GWAS identified IBD-associated epigenetic enzyme variants, SP140 is the only one that has been functionally validated thus far. SP140 is an immune-restricted PHD and bromodomain containing epigenetic ‘reader’ [44] and CD-associated SP140 SNPs resulted in altered splicing of SP140 mRNA and ultimately diminished SP140 protein. SP140 was identified to be essential for macrophage transcriptional programs that dictate cell identity by preferentially occupying silenced, lineage-inappropriate genes bearing H3K27me3 and ensuring their repression [30••]. Consequently, loss of SP140 in mouse or human macrophages and cells carrying CD-associated SP140 SNPs displayed hypo-responsiveness to Toll-like Receptor (TLR) stimulation suggesting a critical role for this epigenetic reader in immune response to microbiota. Importantly, a loss of this epigenetic reader is causative of intestinal inflammation since hematopoietic-specific knockdown of Sp140 in mice resulted in exacerbated DSS–induced colitis [30••]. Thus, an inability to interpret the epigenetic landscape in immune cells due to loss of SP140 contributes to intestinal inflammation.
Global effects of microbiota on host chromatin
Globally, enteric microbiota and their metabolites profoundly influence host chromatin, both at the level of DNA methylation as well as histone post translational modifications (PTMs). Employment of germ-free (GF) raised mice or antibiotic (ABX) treated mice demonstrated that absence of microbiota greatly disrupts host chromatin in the intestine [55,56••,57]. Microbiota maintains DNA methylation of genes regulating bacterial sensing, immunity and homeostasis. In a recent study, DNA methylation at regulatory elements was reduced in intestinal epithelial cells (IECs) of conventional (CV) mice compared with GF mice and associated with inflammatory genes and bacterial sensing pathways, such as MyD88 and this demethylation was mediated by TETs [56••]. Differential DNA methylation occurred early in postnatal development in mice pointing to a critical developmental window for imprinting by the microbiota, in a Dnmt1-dependent manner, which correlated with transcription of genes responsible for mucus barrier formation and intestinal maturation [58], as well as innate immune and phagocytosis genes [59].
Similarly, global alterations in histone modifications (H3K4me3, H3K27me3) were observed in GF and ABX treated mice. In particular, microbiota-dependent histone modifications were enriched for innate and adaptive immune related and metabolic pathways [11,55,60]. In addition, histone crotonylation, a modification that associates with active transcription, was abundantly observed in the colon and was reduced with ABX treatment [61•]. Furthermore, chromatin accessibility, particularly in the enhancer landscape of various intestinal cells, is altered by enteric microbiota that is in concordance with the direction of gene expression changes [56••,60,62]. Conventionalization of GF mice and re-establishing commensal bacteria rescued DNA methylation, histone modification and chromatin accessibility changes [55,56••,63]. Hence, the microbiota is a major influencer of host epigenetic state.
Epigenetics as a sensor of metabolic state
As has been thoroughly reviewed previously [64], epigenetics and metabolism are intimately linked. Almost all chromatin modifying enzymes rely on available metabolites for their catalytic activity and thus will underpin the intricate co-evolutionary relationship between microorganisms and epigenetics for adaptive cellular transcription. DNA methyltransferases (DNMTs) require the metabolite S-adenosyl methionine (SAM) and histone acetyltransferases require acetyl coenzyme coA (acetyl-CoA) [64]. Other metabolites such as α-ketoglutarate, S-adenosyl homocysteine (SAH), β-oxybutyrate similarly serve as substrates, donors, cofactors, or competitive inhibitors for various epigenetic enzymes [64] (Figure 1). Recent discoveries of novel epigenetic marks also exemplify this tight coupling, including histone succinylation, crotonylation and lactylation which rely on substrates succinyl-coA, crotonyl-coA and lactate levels, respectively [65–67]. These metabolites are generated in diverse cellular metabolic pathways, including tricarboxylic acid (TCA) and methionine cycle. However, the microbiome is also a major source of metabolites in mammals, including lipids, vitamins, organic acids, indoles, oxazoles and secondary bile acids (SBAs) [68], and is also emerging as prominent regulator of host epigenetic enzyme activity. In particular, microbiota exclusively metabolize complex carbohydrates derived from dietary fibers via fermentative reactions to produce small organic acids in the colonic lumen, the bulk of which are short chain fatty acids (SCFAs): acetate, propionate and butyrate – all of which are HDAC inhibitors [21,69,70••]. Notably, supplementation of SCFAs in GF mice could recapitulate a large portion of global chromatin states and gene expression patterns observed with complete gut colonization [55]. Insights gleaned from colorectal cancer mouse models also show how other microbial derived metabolites, such as gallic acid, influence chromatin by promoting TCF4-chromatin association and subsequent H3K4me3 at WNT promoters [71•]. Thus, both cellular and microbial metabolites are major drivers of epigenetic activity for malleable transcriptional and cellular output.
Figure 1.
Intimate relationship between metabolism and epigenetics. Host cellular and bacteria-derived metabolites as essential co-factors for chromatin modifying enzyme function. Green arrows indicate promotion of enzyme activity, red arrows indicate inhibition of enzyme activity. This figure was created using Biorender.
Any alteration in microbiota and their metabolic products, a hallmark of IBD, therefore will profoundly alter host epigenetic enzyme activity. Certainly, a reduction of species diversity in microbial communities is consistently observed in IBD patients [8•,72••,73••]. Importantly, this decrease in microbial diversity parallels a decrease in the metabolite pool with reduced bacteria strains being ones that produce beneficial metabolites such as butyrate and phytate [72••,74••]. In addition to SCFAs, pantothenate and nicotinate (vitamins B5 and B3) are also depleted, while polyunsaturated fatty acids such as adrenate and arachidonate are increased in IBD patients [72••]. Other metabolites like bile acids and acylcarnitines are also differential in IBD patients [72••,73••]. Alterations in nicotinate affect the NAD+ pool and thus would be predicted to directly impact the activity of the Sirtuin subfamily of HDACs, while changes in acylcarnitines alters acetyl-coA pools and thus would be predicted to influence HAT activity [64]. However, the precise effects, both acutely and permanently, of all these altered metabolites in IBD on epigenetic enzymes as the critical intermediaries between host and microbiota in human IBD will be a fertile area of future investigation.
Crosstalk between the microbiome and epigenome in intestinal epithelial cells
Intestinal epithelial cells (IECs) act both as a physical barrier to maintain intestinal homeostasis and express innate immune receptors that recognize and respond to commensal microorganisms. It is well established that microbiota regulate intestinal epithelial cell (IEC) barrier integrity, especially at the suckling-to-weaning transition, where onset of solid food ingestion and metabolites is critical for establishing barrier integrity [75]. Lactate produced from commensal bacteria Bifidobacterium and Lactobacillus spp. led to expansion of intestinal stem cells and differentiation into Paneth and goblet cells [76]. Interesting, histone lactylation in macrophages was recently proposed to drive expression of genes involved in tissue homeostasis [67], suggesting its role in IECs may be similar. One of the seminal papers highlighting the critical platform IECs hold for epigenetic programming by microbiota came from Alenghat et al. that depleted histone ‘eraser’ HDAC3 specifically in IECs (HDAC3ߡIEC) [21]. HDAC3ΔIEC mice exhibited greater weight loss, disease severity, loss of crypt architecture, edema and inflammation following DSS-induced colitis. Moreover, a large portion of gene expression regulated by HDAC3 was microbiota-dependent [21]. Epithelial HDAC3 was also essential in integrating metabolic cues and regulated diet-induced obesity in mice [77]. Recent insights expand on how microbiota programmed circadian rhythmicity of H3K4me3 and HDAC3-dependent H3K9ac and H3K27ac, in small intestines and colons [78,79]. HDAC3 increased in IECs after conventionalization of GF mice and further could be sufficiently induced by monocolonization of GF mice with Bacteriodetes thetaiotamicron [78]. Moreover, binding of HDAC3 to its target genes was reduced in the absence of circadian clock repressor REV-ERBα [78], implicating the requirement for sensing circadian rhythmicity. While HDAC3 activity was inhibited with colonization of butyrate producing Faecalibacterium prausnitzii, HDAC3 activity in IECs actually increased in microbiome-replete mice compared to GF mice. It was subsequently revealed that microbial-metabolite inositol triphosphate (IP3) and phytate could activate HDAC activity [74••] demonstrating divergent HDAC modulation by microbial derived metabolites. IECs from mice colonized with phytate producing Escherichia coli had decreased H3K9ac at loci regulated by HDAC3 [74••] and administration of phytate attenuated DSS colitis in mice [74••]. In terms of innate immune functions of IECs, microbiota-dependent transcriptional suppression of the C-type lectin receptor Clec2e in IECs correlated with decreased histone acetylation and recruitment of HDAC3 to reduce adherence of Citrobacter rodentium [80]. In another study, enteric infection with C. rodentium led to overall increased HDAC activity in IECs which was required for proper IFNγ production by interspersed CD8 lymphocytes to clear infection [22•]. Thus, a key mechanism of how microbiota dictate intestinal epithelial cell function is via histone deacetylases, particularly HDAC3, that senses metabolism, circadian rhythmicity and microbial cues in order to coordinate responses and maintain gut homeostasis (Figure 2).
Figure 2.
Epigenetic mechanisms as the intermediary of microorganism-host communication. Schematic of enteric microorganism and metabolic cues that regulate intestinal epithelial cell and immune cell development and function via epigenomic mechanisms. Individual chromatin modifying enzymes demonstrated to be important in this cross-talk are indicated. This figure was created using Biorender.
Crosstalk between the microbiome and epigenome in intestinal-resident immune cells
Intestinal immune cells play an important role in sensing and discriminating between symbionts and pathobionts. Microbiota drive differentiation of unique immune cell populations, including colon macrophage or innate lymphoid cell populations and are indispensable for imprinting tissue specific functions on these cells [60,81] (Figure 2). For instance, macrophages differentiated in the presence of microbial-derived metabolite butyrate displayed enhanced antimicrobial activity against pathogenic bacteria [70••]. This was less so for propionate and not noted with acetate, suggesting unique roles of the various SCFAs [70••]. Other studies similarly observed increased macrophage antibacterial activity [82], reduced NF-κB activation and cytokine production with butyrate or propionate co-stimulation [83,84] (Figure 2). The converse was observed in intestinal macrophages from ABX treated mice [85]. Importantly, butyrate was unable to enhance antimicrobial activity in macrophages with HDAC3 silencing [70••]. Lactobacillus reuteri, a common commensal bacteria, produced a novel form of folate (vitamin B9) that can transfer 2 carbons onto homocysteine resulting in ethionine [86]. Mass spectrometry of human cell line THP-1 histones revealed incorporation of ethionine instead of methionine into proteins, a reduction of histone methylation, and increased ethylation of histone lysine residues [86]. Furthermore, ethionine pre-treatment in THP-1 cells prevented NF-κB activation and TNF transcription following LPS stimulation [86]. Conversely, lactate derived from pathogenic Staphylococcus aureus inhibited HDAC11, unchecked activation of HDAC6, and increased histone 3 acetylation at the IL10 promoter, resulting in enhanced IL10 transcription in mouse host macrophages [87]. These findings present a complex picture of how metabolites from commensal or pathogenic bacteria immunomodulate host chromatin in diverse ways and the ability of epigenetic enzymes to calibrate host responses to diverse microbial signals. Recent findings further support the idea that microbiota epigenetically ‘train’ the state of immune cells. Intestinal bacterium Clostridium scindens provided microbial processed-bile salt, deoxycholate mediated-protection against later Entamoeba histolytica infection by altering H3K4me3 and H3K27me3 in bone marrow granulocyte progenitor cells, leading to expansion and differentiation of intestinal neutrophils [88]. Similarly, microbiota were critical for dendritic cell (DC) responses since DCs from GF and ABX mice could not adequately produce type I interferon (IFN) [89] (Figure 2). This was not due to a defect in intrinsic innate signaling but rather due to chromatin changes with lower levels of H3K4me3 on transcriptional start sites of many inflammatory primary response genes or IFN stimulatory genes and enriched H3K27me3 on metabolic processes that were critical in priming subsequent T cell responses [89,90••]. Thus, commensal microbiota and their metabolites instruct intestinal innate immune cells via chromatin mechanisms to calibrate and fine-tune transcriptional responses.
In parallel, the microbiota is critical for regulation of adaptive immune responses and maintenance of tolerance towards commensal microbes via the epigenome (Figure 2). Butyrate, and to a lesser extent propionate, programs generation of T regulatory cells (Tregs) in the periphery and colon by inhibiting HDAC activity at the Foxp3 locus [91–93]. Conversely, butyrate suppressed Th17 differentiation [94–96]. Butyrate, or supernatant of F. prausnitzii, administration in vivo alleviated colitis [94,97]. Other less abundant SCFAs, such as pentanoate, also affect Th17 proliferation and IL17A production [98•]. Comparative assessment of SCFAs revealed pentanoate, butyrate and propionate had potent HDAC-inhibitory activity in T lymphocytes, while acetate and hexanoate had little activity [93,98•]. Pentanoate impacted B regulatory cells (Bregs) but had no impact on Tregs [98•]. Other metabolites such as secondary bile acids (SBA) also modulate Th17 and Treg differentiation with some evidence that this was via chromatin architecture [99,100•]. 3-oxolithocholic acid and isoallolithocholic acid had reciprocal roles with 3-oxoLCA inhibiting Th17 cells differentiation while isoalloLCA increased Tregs [100•]. Administration of SBAs, lithocholic acid, deoxycholic acid and ursodeoxycholic acid alleviated DSS-induced, TNBS-induced, T cell transfer colitis and Clostridiodes difficile infection models [73••,101]. Furthermore, there is some evidence that they may similarly act as an HDAC inhibitor [102]. These studies collectively highlight the cell-specific, metabolite-specific programming by microbiota. However, most work has focused thus far on SCFAs and HDACs in this process, as HDAC activity is an easier output to assay. Understanding the underlying chromatin mechanisms governing immunomodulation by other microbial metabolites, at defined loci, will be an exciting avenue of future research.
Concluding remarks, perspectives and future directions
IBD is a multifactorial disease, that is more likely multiple subsets of diseases, making both accurate diagnosis and therapeutic management challenging. Substantial progress has been made in the genetic understanding of IBD subtypes and onset, yet genetic factors explain only a small portion of overall disease incidence. Environmental triggers such as perturbation of enteric microorganisms are also a major factor for disease stratification; however, pathways underlying how cues from microbiota are integrated during development of IBD remain poorly understood. Epigenetics links genetic predisposition with disturbances of microorganisms in the pathogenesis of IBD and may offer novel precision medicine diagnosis and therapeutic opportunities.
Indeed, the importance of epigenetics in mediating host-microorganism-metabolism relationships in health and IBD is rapidly emerging. Furthermore, alterations in the epigenome or genetic variants of chromatin enzymes is materializing as a causative factor in IBD. However, our grasp of the mechanisms, the relationship between lesser-studied microorganisms such as enteric fungi and viruses and the epigenome, the role of novel microorganism-derived metabolites in altering the host epigenome and the extent to which altered epigenetics in specific cell types directly contribute to disease is still in its infancy. One of the fundamental questions that still remains unanswered is whether rescue of epigenetic, microbiome or metabolic alterations with the ever-expanding array of epigenetic-targeting drugs is a feasible strategy in complex immune disorders such as IBD. Deciphering the critical epithelial, immune or neuronal pathways specifically regulated by certain chromatin-modifying enzymes in response to changing commensal microorganisms and their metabolites and how this contributes to distinct phenotypes of IBD will be essential. The rapid improvement of chromatin techniques in limited cell numbers that are becoming more accessible to immunologists will help achieve this goal. Finally, a more expansive and unbiased assessment of regulation of lesser-studied aspects of the epigenome by commensal microbiota or novel metabolites in individual cell types is warranted and may also reveal unexpected avenues for therapeutic intervention of the epigenome-metabolome-microbiome axis.
Funding
Canadian Institute of Health Research (CIHR) postdoctoral fellowship (HA). Kenneth Rainin Foundation (Innovator and Synergy Awards to KLJ), NIH R21AI144877 (KLJ), NIH R01DK119996 (KLJ), Harvard Catalyst | The Harvard Clinical and Translational Science Center (National Center for Advancing Translational Sciences, National Institutes of Health Award UL 1TR002541) and financial contributions from Harvard University and its affiliated academic healthcare centers (KLJ), MGH Research Scholar, class of 2020 (KLJ).
Footnotes
Conflict of interest statement
Nothing declared.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1.Allis CD, Jenuwein T: The molecular hallmarks of epigenetic control. Nat Rev Genet 2016, 17:487–500. [DOI] [PubMed] [Google Scholar]
- 2.Zhao Y, Garcia BA: Comprehensive catalog of currently documented histone modifications. Cold Spring Harb Perspect Biol 2015, 7:a025064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Graham DB, Xavier RJ: Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020, 578:527–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Farh KK, Marson A, Zhu J, Kleinewietfeld M, Housley WJ, Beik S, Shoresh N, Whitton H, Ryan RJ, Shishkin AA et al. : Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 2015, 518:337–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cheung P, Vallania F, Warsinske HC, Donato M, Schaffert S, Chang SE, Dvorak M, Dekker CL, Davis MM, Utz PJ et al. : Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 2018, 173:1385–1397 e1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M: Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov 2012, 11:384–400. [DOI] [PubMed] [Google Scholar]
- 7.Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H et al. : Suppression of inflammation by a synthetic histone mimic. Nature 2010, 468:1119–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ryan FJ, Ahern AM, Fitzgerald RS, Laserna-Mendieta EJ, Powe EM, Cloone AG, O’Donoghue KW, McMurdie PJ, Iwai S, Crits-Christoph A et al.: Colonic microbiota is associated with inflammation and host epigenomic alterations in inflammatory bowel disease. Nat Commun 2020, 11:1512.• Report details microbiota association with host transcriptomics, epigenomics and genetics from matched inflamed and non-inflamed colonic mucosa of IBD patients and controls.
- 9.Howell KJ, Kraiczy J, Nayak KM, Gasparetto M, Ross A, Lee C, Mak TN, Koo BK, Kumar N, Lawley T et al. : DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome. Gastroenterology 2018, 154:585–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Somineni HK, Venkateswaran S, Kilaru V, Marigorta UM, Mo A, Okou DT, Kellermayer R, Mondal K, Cobb D, Walters TD et al. : Blood-derived DNA methylation signatures of Crohn’s disease and severity of intestinal inflammation. Gastroenterology 2019, 156:2254–2265 e2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kelly D, Kotliar M, Woo V, Jagannathan S, Whitt J, Moncivaiz J, Aronow BJ, Dubinsky MC, Hyams JS, Markowitz JF et al. : Microbiota-sensitive epigenetic signature predicts inflammation in Crohn’s disease. JCI Insight 2018, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Elliott EN, Sheaffer KL, Kaestner KH: The ‘de novo’ DNA methyltransferase Dnmt3b compensates the Dnmt1-deficient intestinal epithelium. eLife 2016, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang R, Li H, Wu J, Cai ZY, Li B, Ni H, Qiu X, Chen H, Liu W, Yang ZH et al. : Gut stem cell necroptosis by genome instability triggers bowel inflammation. Nature 2020, 580:386–390•• Demonstrated that epigenetic regulator Setdb1 is decreased in IBD patients and mice with Setdb1 deficiency in intestinal epithelial cells had spontaneous colitis due to genomic instability and severe necropsis. Endogenous retroviruses (ERVs) dysregulated in Setdb1-knockout crypts.
- 14.Liu Y, Peng J, Sun T, Li N, Zhang L, Ren J, Yuan H, Kan S, Pan Q, Li X et al. : Epithelial EZH2 serves as an epigenetic determinant in experimental colitis by inhibiting TNFalpha-mediated inflammation and apoptosis. Proc Natl Acad Sci U S A 2017, 114: E3796–E3805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Xia M, Liu J, Liu S, Chen K, Lin H, Jiang M, Xu X, Xue Y, Liu W, Gu Y et al. : Ash1l and lnc-Smad3 coordinate Smad3 locus accessibility to modulate iTreg polarization and T cell autoimmunity. Nat Commun 2017, 8:15818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jones GR, Brown SL, Phythian-Adams AT, Ivens AC, Cook PC, MacDonald AS: The methyl-CpG-binding protein Mbd2 regulates susceptibility to experimental colitis via control of CD11c(+) cells and colonic epithelium. Front Immunol 2020, 11:183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang Q, Zhao K, Shen Q, Han Y, Gu Y, Li X, Zhao D, Liu Y, Wang C, Zhang X et al. : Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature 2015, 525:389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Turgeon N, Gagne JM, Blais M, Gendron FP, Boudreau F, Asselin C: The acetylome regulators Hdac1 and Hdac2 differently modulate intestinal epithelial cell dependent homeostatic responses in experimental colitis. Am J Physiol Gastrointest Liver Physiol 2014, 306:G594–605. [DOI] [PubMed] [Google Scholar]
- 19.Singh AK, Khare P, Obaid A, Conlon KP, Basrur V, DePinho RA, Venuprasad K: SUMOylation of ROR-gammat inhibits IL-17 expression and inflammation via HDAC2. Nat Commun 2018, 9:4515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wang L, Liu Y, Han R, Beier UH, Bhatti TR, Akimova T, Greene MI, Hiebert SW, Hancock WW: FOXP3(+) regulatory T cell development and function require histone/protein deacetylase 3. J Clin Invest 2015, 125:3304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Alenghat T, Osborne LC, Saenz SA, Kobuley D, Ziegler CG, Mullican SE, Choi I, Grunberg S, Sinha R, Wynosky-Dolfi M et al. : Histone deacetylase 3 coordinates commensal-bacteria-dependent intestinal homeostasis. Nature 2013, 504:153–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Navabi N, Whitt J, Wu SE, Woo V, Moncivaiz J, Jordan MB, Vallance BA, Way SS, Alenghat T: Epithelial histone deacetylase 3 instructs intestinal immunity by coordinating local lymphocyte activation. Cell Rep 2017, 19:1165–1175• Support the role for HDAC3 in deciphering external cues and orchestrating other cell type responses to maintain homeostasis. HDAC3 silencing in intestinal epithelial cells significantly impaired the ability of neighbouring intraepithelial CD8 T cells to produce IFNγ against Citrobacter infection.
- 23.Kasler HG, Lim HW, Mottet D, Collins AM, Lee IS, Verdin E: Nuclear export of histone deacetylase 7 during thymic selection is required for immune self-tolerance. EMBO J 2012, 31:4453–4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Liu F, Bu HF, Geng H, De Plaen IG, Gao C, Wang P, Wang X, Kurowski JA, Yang H, Qian J et al. : Sirtuin-6 preserves R-spondin-1 expression and increases resistance of intestinal epithelium to injury in mice. Mol Med 2017, 23:272–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Obata Y, Furusawa Y, Endo TA, Sharif J, Takahashi D, Atarashi K, Nakayama M, Onawa S, Fujimura Y, Takahashi M et al. : The epigenetic regulator Uhrf1 facilitates the proliferation and maturation of colonic regulatory T cells. Nat Immunol 2014, 15:571–579. [DOI] [PubMed] [Google Scholar]
- 26.Qi S, Li Y, Dai Z, Xiang M, Wang G, Wang L, Wang Z: Uhrf1-mediated Tnf-alpha gene methylation controls proinflammatory macrophages in experimental colitis resembling inflammatory bowel disease. J Immunol 2019, 203:3045–3053. [DOI] [PubMed] [Google Scholar]
- 27.Tanaka S, Pfleger C, Lai JF, Roan F, Sun SC, Ziegler SF: KAP1 regulates regulatory T cell function and proliferation in both Foxp3-dependent and -independent manners. Cell Rep 2018, 23:796–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chikuma S, Suita N, Okazaki IM, Shibayama S, Honjo T: TRIM28 prevents autoinflammatory T cell development in vivo. Nat Immunol 2012, 13:596–603. [DOI] [PubMed] [Google Scholar]
- 29.Gehrmann U, Burbage M, Zueva E, Goudot C, Esnault C, Ye M, Carpier JM, Burgdorf N, Hoyler T, Suarez G et al. : Critical role for TRIM28 and HP1beta/gamma in the epigenetic control of T cell metabolic reprograming and effector differentiation. Proc Natl Acad Sci U S A 2019, 116:25839–25849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mehta S, Cronkite DA, Basavappa M, Saunders TL, Adiliaghdam F, Amatullah H, Morrison SA, Pagan JD, Anthony RM, Tonnerre P et al. : Maintenance of macrophage transcriptional programs and intestinal homeostasis by epigenetic reader SP140. Sci Immunol 2017, 2•• First confirmation of a chromatin enzyme variant identified by GWAS to be lost in IBD patients. The immune-restricted epigenetic reader SP140 is critical for macrophage identity and function. A loss of this epigenetic enzyme is causative of intestinal inflammation since hematopoietic knockdown of Sp140 leads to exacerbated DSS-colitis.
- 31.Liu M, Sun T, Li N, Peng J, Fu D, Li W, Li L, Gao WQ: BRG1 attenuates colonic inflammation and tumorigenesis through autophagy-dependent oxidative stress sequestration. Nat Commun 2019, 10:4614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Qi X, Qiu J, Chang J, Ji Y, Yang Q, Cui G, Sun L, Chai Q, Qin J, Qiu J: Brg1 restrains the pro-inflammatory properties of ILC3s and modulates intestinal immunity. Mucosal Immunol 2020. 10.1038/s41385-020-0317-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.He X, Yu J, Wang M, Cheng Y, Han Y, Yang S, Shi G, Sun L, Fang Y, Gong ST et al. : Bap180/Baf180 is required to maintain homeostasis of intestinal innate immune response in Drosophila and mice. Nat Microbiol 2017, 2:17056. [DOI] [PubMed] [Google Scholar]
- 34.Antignano F, Burrows K, Hughes MR, Han JM, Kron KJ, Penrod NM, Oudhoff MJ, Wang SK, Min PH, Gold MJ et al. : Methyltransferase G9A regulates T cell differentiation during murine intestinal inflammation. J Clin Invest 2014, 124:1945–1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yang Y, Guan J, Shaikh AS, Liang Y, Sun L, Wang M, Li D, Qiu C, Li X: Histone acetyltransferase Mof affects the progression of DSS-induced colitis. Cell Physiol Biochem 2018, 47:2159–2169. [DOI] [PubMed] [Google Scholar]
- 36.Liu Z, Cao W, Xu L, Chen X, Zhan Y, Yang Q, Liu S, Chen P, Jiang Y, Sun X et al. : The histone H3 lysine-27 demethylase Jmjd3 plays a critical role in specific regulation of Th17 cell differentiation. J Mol Cell Biol 2015, 7:505–516. [DOI] [PubMed] [Google Scholar]
- 37.Zhou Q, Zhang Y, Wang B, Zhou W, Bi Y, Huai W, Chen X, Chen Y, Liu Z, Liu X et al. : KDM2B promotes IL-6 production and inflammatory responses through Brg1-mediated chromatin remodeling. Cell Mol Immunol 2020, 17:834–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.de Zoeten EF, Wang L, Butler K, Beier UH, Akimova T, Sai H, Bradner JE, Mazitschek R, Kozikowski AP, Matthias P et al. : Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3(+) T-regulatory cells. Mol Cell Biol 2011, 31:2066–2078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, Li B, Turka LA, Olson EN, Greene MI et al. : Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 2007, 13:1299–1307. [DOI] [PubMed] [Google Scholar]
- 40.de Zoeten EF, Wang L, Sai H, Dillmann WH, Hancock WW: Inhibition of HDAC9 increases T regulatory cell function and prevents colitis in mice. Gastroenterology 2010, 138:583–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dahiya S, Beier UH, Wang L, Han R, Jiao J, Akimova T, Angelin A, Wallace DC, Hancock WW: HDAC10 deletion promotes Foxp3 (+) T-regulatory cell function. Sci Rep 2020, 10:424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kazakevych J, Denizot J, Liebert A, Portovedo M, Mosavie M, Jain P, Stellato C, Fraser C, Correa RO, Celestine M et al. : Smarcad1 mediates microbiota-induced inflammation in mouse and coordinates gene expression in the intestinal epithelium. Genome Biol 2020, 21:64.• Highlights how a chromatin remodeling enzyme, Smarcad1, can regulate global H3K9me3 deposition as well as chromatin accessibility. Smarcad1 dependent colitis response in mice may be due to poorly characterized TM7 phylum microbiota.
- 43.Nasrallah R, Imianowski CJ, Bossini-Castillo L, Grant FM, Dogan M, Placek L, Kozhaya L, Kuo P, Sadiyah F, Whiteside SK et al. : A distal enhancer at risk locus 11q.5 promotes suppression of colitis by Treg cells. Nature 2020, 583:447–452•• An IBD risk loci in a non-coding, chromatin regulatory enhancer region is essential for suppression of intestinal inflammation by Tregs.
- 44.Fraschilla I, Jeffrey KL: The Speckled Protein (SP) family: immunity’s chromatin readers. Trends Immunol 2020, 41:572–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, Lee JC, Schumm LP, Sharma Y, Anderson CA et al. : Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491:119–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Huang H, Fang M, Jostins L, Umicevic Mirkov M, Boucher G, Anderson CA, Andersen V, Cleynen I, Cortes A, Crins F et al. : Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 2017, 547:173–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.de Lange KM, Moutsianas L, Lee JC, Lamb CA, Luo Y, Kennedy NA, Jostins L, Rice DL, Gutierrez-Achury J, Ji SG et al. : Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet 2017, 49:256–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, Lees CW, Balschun T, Lee J, Roberts R et al. : Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet 2010, 42:1118–1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang Y, Tian L, Sleiman P, Ghosh S, Hakonarson H, International IBDGC: Bayesian analysis of genome-wide inflammatory bowel disease data sets reveals new risk loci. Eur J Hum Genet 2018, 26:265–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, Park YR, Raychaudhuri S, Pouget JG, Hubenthal M et al. : Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet 2016, 48:510–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Damas OM, Gomez L, Quintero MA, Rampersaud E, Slifer S, Beecham GW, Kerman DH, Deshpande AR, Sussman DA, Abreu MT et al. : Genetic characterization and influence on inflammatory bowel disease expression in a diverse Hispanic south Florida cohort. Clin Transl Gastroenterol 2017, 8:e87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mesbah-Uddin M, Elango R, Banaganapalli B, Shaik NA, Al-Abbasi FA: In-silico analysis of inflammatory bowel disease (IBD) GWAS loci to novel connections. PLoS One 2015, 10 e0119420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ostrowski J, Paziewska A, Lazowska I, Ambrozkiewicz F, Goryca K, Kulecka M, Rawa T, Karczmarski J, Dabrowska M, Zeber-Lubecka N et al. : Genetic architecture differences between pediatric and adult-onset inflammatory bowel diseases in the Polish population. Sci Rep 2016, 6:39831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Brant SR, Okou DT, Simpson CL, Cutler DJ, Haritunians T, Bradfield JP, Chopra P, Prince J, Begum F, Kumar A et al. : Genome-wide association study identifies African-specific susceptibility loci in African Americans with inflammatory bowel disease. Gastroenterology 2017, 152:206–217 e202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, Rabaglia ME, Keller MP, Attie AD, Rey FE, Denu JM: Diet-microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol Cell 2016, 64:982–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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•• Demonstrated how the presence of microbiota induced profound DNA methylation changes at regulatory elements and microbiota dependent demethylation was mediated by the TET2/3 enzymes.
- 57.Pan WH, Sommer F, Falk-Paulsen M, Ulas T, Best P, Fazio A, Kachroo P, Luzius A, Jentzsch M, Rehman A et al. : Exposure to the gut microbiota drives distinct methylome and transcriptome changes in intestinal epithelial cells during postnatal development. Genome Med 2018, 10:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yu DH, Gadkari M, Zhou Q, Yu S, Gao N, Guan Y, Schady D, Roshan TN, Chen MH, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pan X, Gong D, Nguyen DN, Zhang X, Hu Q, Lu H, Fredholm M, Sangild PT, Gao F: Early microbial colonization affects DNA methylation of genes related to intestinal immunity and metabolism in preterm pigs. DNA Res 2018, 25:287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gury-BenAri M, Thaiss CA, Serafini N, Winter DR, Giladi A, Lara-Astiaso D, Levy M, Salame TM, Weiner A, David E et al. : The spectrum and regulatory landscape of intestinal innate lymphoid cells are shaped by the microbiome. Cell 2016, 166:1231–1246 e1213. [DOI] [PubMed] [Google Scholar]
- 61.Fellows R, Denizot J, Stellato C, Cuomo A, Jain P, Stoyanova E, Balazsi S, Hajnady Z, Liebert A, Kazakevych J et al. : Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nat Commun 2018, 9:105.• Highlights a novel histone modification– crotonylation, and its relative abundance in small intestine crypt and colon. Antibiotic cocktail treatment led to a decrease in colon histone crotonylation.
- 62.Richards AL, Muehlbauer AL, Alazizi A, Burns MB, Findley A, Messina F, Gould TJ, Cascardo C, Pique-Regi R, Blekhman R et al. : Gut microbiota has a widespread and modifiable effect on host gene regulation. mSystems 2019, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Semenkovich NP, Planer JD, Ahern PP, Griffin NW, Lin CY, Gordon JI: Impact of the gut microbiota on enhancer accessibility in gut intraepithelial lymphocytes. Proc Natl Acad Sci U S A 2016, 113:14805–14810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chisolm DA, Weinmann AS: Connections between metabolism and epigenetics in programming cellular differentiation. Annu Rev Immunol 2018, 36:221–246. [DOI] [PubMed] [Google Scholar]
- 65.Smestad J, Erber L, Chen Y, Maher LJ 3rd: Chromatin succinylation correlates with active gene expression and is perturbed by defective TCA cycle metabolism. iScience 2018, 2:63–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gowans GJ, Bridgers JB, Zhang J, Dronamraju R, Burnetti A, King DA, Thiengmany AV, Shinsky SA, Bhanu NV, Garcia BA et al. : Recognition of histone crotonylation by Taf14 links metabolic state to gene expression. Mol Cell 2019, 76:909–921 e903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M et al. : Metabolic regulation of gene expression by histone lactylation. Nature 2019, 574:575–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G: Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A 2009, 106:3698–3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D: Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J Nutr Biochem 2008, 19:587–593. [DOI] [PubMed] [Google Scholar]
- 70.Schulthess J, Pandey S, Capitani M, Rue-Albrecht KC, Arnold I, Franchini F, Chomka A, Ilott NE, Johnston DGW, Pires E et al. : The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 2019, 50:432–445 e437•• Illustrated how butyrate treated macrophages have higher antibacterial activity against pathogenic bacterial challenges. Treatment of mice with butyrate also alleviated systemic bacterial dissemination following Citrobacter infection. Importantly, this antibacterial activity was lost in macrophages deficient in HDAC3.
- 71.Kadosh E, Snir-Alkalay I, Venkatachalam A, May S, Lasry A, Elyada E, Zinger A, Shaham M, Vaalani G, Mernberger M et al. : The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 2020. 10.1038/s41586-020-2541-0. ahead of print• Discovery that microbiota-derived gallic acid can recapitulate phenotypes of the complete microbiome and enables TCF4-chromatin association and subsequent H3K4me3 at WNT promoters.
- 72.Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ et al. : Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569:655–662•• Longitudinal multiomics study provides extensive findings on changes in transcriptomic, epigenomic, microbiota and metabolite changes in human IBD.
- 73.Sinha SR, Haileselassie Y, Nguyen LP, Tropini C, Wang M, Becker LS, Sim D, Jarr K, Spear ET, Singh G et al. : Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 2020, 27:659–670 e655.••
- 74.Wu SE, Hashimoto-Hill S, Woo V, Eshleman EM, Whitt J, Engleman L, Karns R, Denson LA, Haslam DB, Alenghat T: Microbiota-derived metabolite promotes HDAC3 activity in the gut. Nature 2020. 10.1038/s41586-020-2604-2. ahead of print•• Identification of microbial metabolites inositol phosphate 3 and phytate as promoters of HDAC activity for regulation of intestinal inflammation in mice.
- 75.Beaumont M, Paes C, Mussard E, Knudsen C, Cauquil L, Aymard P, Barilly C, Gabinaud B, Zemb O, Fourre S et al. : Gut microbiota derived metabolites contribute to intestinal barrier maturation at the suckling-to-weaning transition. Gut Microbes 2020:1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lee YS, Kim TY, Kim Y, Lee SH, Kim S, Kang SW, Yang JY, Baek IJ, Sung YH, Park YY et al. : Microbiota-derived lactate accelerates intestinal stem-cell-mediated epithelial development. Cell Host Microbe 2018, 24:833–846 e836. [DOI] [PubMed] [Google Scholar]
- 77.Davalos-Salas M, Montgomery MK, Reehorst CM, Nightingale R, Ng I, Anderton H, Al-Obaidi S, Lesmana A, Scott CM, Ioannidis P et al. : Deletion of intestinal Hdac3 remodels the lipidome of enterocytes and protects mice from diet-induced obesity. Nat Commun 2019, 10:5291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kuang Z, Wang Y, Li Y, Ye C, Ruhn KA, Behrendt CL, Olson EN, Hooper LV: The intestinal microbiota programs diurnal rhythms in host metabolism through histone deacetylase 3. Science 2019, 365:1428–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Thaiss CA, Levy M, Korem T, Dohnalova L, Shapiro H, Jaitin DA, David E, Winter DR, Gury-BenAri M, Tatirovsky E et al. : Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 2016, 167:1495–1510 e1412. [DOI] [PubMed] [Google Scholar]
- 80.Woo V, Eshleman EM, Rice T, Whitt J, Vallance BA, Alenghat T: Microbiota inhibit epithelial pathogen adherence by epigenetically regulating C-type lectin expression. Front Immunol 2019, 10:928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kang B, Alvarado LJ, Kim T, Lehmann ML, Cho H, He J, Li P, Kim BH, Larochelle A, Kelsall BL: Commensal microbiota drive the functional diversification of colon macrophages. Mucosal Immunol 2020, 13:216–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Fernando MR, Saxena A, Reyes JL, McKay DM: Butyrate enhances antibacterial effects while suppressing other features of alternative activation in IL-4-induced macrophages. Am J Physiol Gastrointest Liver Physiol 2016, 310: G822–831. [DOI] [PubMed] [Google Scholar]
- 83.Chang PV, Hao L, Offermanns S, Medzhitov R: The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A 2014, 111:2247–2252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Park JW, Kim HY, Kim MG, Jeong S, Yun CH, Han SH: Short-chain fatty acids inhibit staphylococcal lipoprotein-induced nitric oxide production in murine macrophages. Immune Netw 2019, 19:e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Scott NA, Andrusaite A, Andersen P, Lawson M, Alcon-Giner C, Leclaire C, Caim S, Le Gall G, Shaw T, Connolly JPR et al. : Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci Transl Med 2018, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Roth D, Chiang AJ, Hu W, Gugiu GB, Morra CN, Versalovic J, Kalkum M: Two-carbon folate cycle of commensal Lactobacillus reuteri 6475 gives rise to immunomodulatory ethionine, a source for histone ethylation. FASEB J 2019, 33:3536–3548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Heim CE, Bosch ME, Yamada KJ, Aldrich AL, Chaudhari SS, Klinkebiel D, Gries CM, Alqarzaee AA, Li Y, Thomas VC et al. : Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection. Nat Microbiol 2020. 10.1038/s41564-020-0756-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Burgess SL, Leslie JL, Uddin MJ, Oakland DN, Gilchrist CA, Moreau GB, Watanabe K, Saleh MM, Simpson M, Thompson BA et al. : Gut microbiome communication with bone marrow regulates susceptibility to amebiasis. J Clin Invest 2020, 130:4019–4024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ganal SC, Sanos SL, Kallfass C, Oberle K, Johner C, Kirschning C, Lienenklaus S, Weiss S, Staeheli P, Aichele P et al. : Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity 2012, 37:171–186. [DOI] [PubMed] [Google Scholar]
- 90.Schaupp L, Muth S, Rogell L, Kofoed-Branzk M, Melchior F, Lienenklaus S, Ganal-Vonarburg SC, Klein M, Guendel F, Hain T et al. : Microbiota-induced Type I interferons instruct a poised basal state of dendritic cells. Cell 2020•• Expands on how microbiota programs the chromatin landscape of dendritic cells in their ability to respond to pathogens and prime T cells.
- 91.Furusawa Y, Obata Y, Fukuda S, Endo TA, 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. [DOI] [PubMed] [Google Scholar]
- 92.Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ et al. : Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504:451–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS: The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341:569–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhang M, Zhou L, Wang Y, Dorfman RG, Tang D, Xu L, Pan Y, Zhou Q, Li Y, Yin Y et al. : Faecalibacterium prausnitzii produces butyrate to decrease c-Myc-related metabolism and Th17 differentiation by inhibiting histone deacetylase 3. Int Immunol 2019, 31:499–514. [DOI] [PubMed] [Google Scholar]
- 96.Zhou L, Zhang M, Wang Y, Dorfman RG, Liu H, Yu T, Chen X, Tang D, Xu L, Yin Y et al. : 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. [DOI] [PubMed] [Google Scholar]
- 97.Simeoli R, Mattace Raso G, Pirozzi C, Lama A, Santoro A, Russo R, Montero-Melendez T, Berni Canani R, Calignano A, Perretti M et al. : 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Luu M, Pautz S, Kohl V, Singh R, Romero R, Lucas S, Hofmann J, Raifer H, Vachharajani N, Carrascosa LC et al. : The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat Commun 2019, 10:760.• Expands on a lesser studied SCFA, pentanoate, and its role in regulating Th17 cell differentiation. Pentanoate is present in SPF mice but not GF mice and has potent HDAC inhibitory activity. It is also cell type specific as it additionally impacts B regulatory cells but not T regulatory cells.
- 99.Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, Geva-Zatorsky N, Jupp R, Mathis D, Benoist C et al. : Microbial bile acid metabolites modulate gut RORgamma(+) regulatory T cell homeostasis. Nature 2020, 577:410–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hang S, Paik D, Ya L, Ki E, Trinat J, L J, H S, Nelso BN, Kell SP, W L et al.: Bile acid metabolites control TH17 and Treg cell differentiation. Nature 2019, 576:143–148• Demonstrated how microbial processed bile acid metabolites shape T cell differentiation and modulate intestinal inflammation.
- 101.Winston JA, Rivera AJ, Cai J, Thanissery R, Montgomery SA, Patterson AD, Theriot CM: Ursodeoxycholic acid (UDCA) mitigates the host inflammatory response during Clostridioides difficile infection by altering gut bile acids. Infect Immun 2020, 88:e00045–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Akare S, Jean-Louis S, Chen W, Wood DJ, Powell AA, Martinez JD: Ursodeoxycholic acid modulates histone acetylation and induces differentiation and senescence. Int J Cancer 2006, 119:2958–2969. [DOI] [PubMed] [Google Scholar]