Tissues of the mucosa are lined by an epithelium that provides barrier and transport functions. It is now appreciated that inflammatory responses in inflammatory bowel diseases are accompanied by striking shifts in tissue metabolism. In this paper, we examined global metabolic consequences of mucosal inflammation using both in vitro and in vivo models of disease. Initial analysis of the metabolic signature elicited by inflammation in epithelial models and in colonic tissue isolated from murine colitis demonstrated that levels of specific metabolites associated with cellular methylation reactions are significantly altered by model inflammatory systems. Furthermore, expression of enzymes central to all cellular methylation, S-adenosylmethionine synthetase and S-adenosylhomocysteine hydrolase, are increased in response to inflammation. Subsequent studies showed that DNA methylation is substantially increased during inflammation and that epithelial NF-κB activity is significantly inhibited following treatment with a reversible S-adenosylhomocysteine hydrolase inhibitor, DZ2002. Finally, these studies demonstrated that inhibition of cellular methylation in a murine model of colitis results in disease exacerbation while folate supplementation to promote methylation partially ameliorates the severity of murine colitis. Taken together, these results identify a global change in methylation, which during inflammation, translates to an overall protective role in mucosal epithelia.

The inflammatory bowel diseases (IBD), including both Crohn’s disease and ulcerative colitis, are debilitating diseases of unknown etiology (1), but a combination of genetic and environmental factors are thought to be involved in disease pathology. Recent evidence suggests that IBD likely results from dysregulated immune responses to luminal triggers. Intestinal epithelial cells are a crucial cell type in the maintenance of colonic tissue homeostasis, and IBD is characterized by a breakdown of the intestinal epithelial barrier leading to increased exposure of the mucosal immune system to antigenic luminal material. This exposure leads to inflammation and increased breakdown of the epithelial barrier (13). It is now appreciated that ongoing inflammation leads to significant shifts in tissue metabolism. These shifts result, at least in part, from perturbation of the vasculature (4, 5) leading to the decreased supply of oxygen and nutrients (68). The expression of increased levels of cytokines and chemokines initiates the migration of innate immune cells such as neutrophils, macrophages, and dendritic cells into the mucosal tissues (9). The recruitment of inflammatory cells increases the local immune response as well as the oxygen and nutrient demand of the tissue, exacerbating the metabolic changes associated with IBD. IBD almost certainly elicits changes in a number of cellular metabolic pathways; however, the metabolic shifts induced in the colonic epithelia during the inflammatory process have not been elucidated in a systematic manner.

Studies to date indicate that alterations of cellular methylation have important implications in inflammation and immune responses (10). These studies have shown that methylation, through epigenetic mechanisms and protein modification, has important consequences for these processes, yet the molecular mechanisms of such changes remain poorly understood. Cellular methylation reactions include modification of DNA, RNA, proteins, and lipids. These reactions all require a methyl donor for the modification of the target. The methyl donor for the majority of these reactions is S-adenosylmethionine (SAM) (11). SAM is distributed throughout the cell to act as donor for the various methyltransferases. The donation of its methyl group produces S-adenosylhomocysteine (SAH) from SAM. SAH is a potent inhibitor of methyltransferase enzymes because these enzymes have a higher affinity for SAH (1214), and SAH is rapidly converted to homocysteine and adenosine by SAH hydrolase. Therefore, inhibition of SAH hydrolase represents a powerful means of inhibiting cellular methylation reactions (15). It has been known for a number of years that inhibition of methylation had immunomodulatory properties (16), but mechanisms remain poorly understood. This led to the development of more specific and less toxic SAH inhibitors for use in animal models. The SAH hydrolase inhibitor methyl 4-(adenin-9-yl)-2-hydroxybutanoate (DZ2002) has been found to have immunosuppressive actions and ameliorate disease in a number of animal models (1721). One mechanism for the immunosuppressive action of DZ2002 was shown to be the cell type-specific inhibition of NF-κB activation (19). These studies demonstrate that cellular methylation reactions play an important role in chronic inflammatory disease, such as IBD.

The hypothesis of the current study was that ongoing mucosal inflammatory responses are characterized by specific shifts in metabolic profiles. To define these principles, a nuclear magnetic resonance (NMR)-based metabolomics approach was used to elucidate the metabolic changes associated with murine models of IBD and in cellular models of mucosal inflammation. This analysis revealed that a number of metabolites associated with cellular methylation reactions are significantly impacted by ongoing inflammation. Dovetailing these findings with microarray analysis of epithelial inflammation models, we found that the expression of enzymes central to cellular methylation is significantly altered during mucosal inflammation. We confirmed these findings at the molecular level and extended these studies to a murine model of colitis. These results indicate that methylation reactions play an important role in mucosal inflammation.

Human intestinal epithelial cells (T84) and HeLa epithelial cells were grown and maintained in T175 cell culture flasks (Costar, Cambridge, MA) as described previously (22). DZ2002 (Diazyme, San Diego, CA) was prepared as described previously (19). Where indicated, DZ2002 was added at the indicated concentrations by preincubation for1 h prior to the indicated treatments.

Cells were harvested by scraping, pelleted by centrifugation, washed with ice-cold PBS, and lysed by sonication in Tris lysis buffer (150 mM NaCl, 20 mM Tris [pH 5.5], 1 mM EDTA, 1 mM EGTA, and 1% Triton X-100). Similarly, snap-frozen colon tissues samples were sonicated in Tris buffer. Protein concentration was assessed by the bicinchoninic acid assay following the manufacturer’s instructions (Thermo Scientific) to ensure equal protein loading of each preparation. Proteins were separated by SDS-PAGE electrophoresis and transferred to polyvinylidene difluoride membrane (Bio-Rad Laboratories, Hercules, CA) for immunoblotting. Abs used for this study were as follows: anti-SAM synthetase (1/1,000; Abcam), anti-SAH hydrolase (1/1,000; gift from Dr. D. Kloor, University of Tuebingen, Tuebingen, Germany), anti-IκBα (1/2,000; Cell Signaling Technologies), anti–β-actin (1/10,000; Abcam), anti–β-tubulin (1/10,000; Abcam). Proteins were visualized using the SuperSignal detection substrate (Thermo Scientific).

RNA was isolated using TRIzol (Invitrogen) and cDNA synthesized as described previously (10). Potential contaminating genomic DNA was digested using Turbo DNA-free (Ambion, Austin, TX). The mRNA profile of intestinal epithelial cells (T84) subjected to modeled inflammation (IFN-γ at 10 ng/ml for 6 or 18 h) was compared with that of control cells exposed to vehicle only using Genechip expression arrays (Affymetrix) as described previously (23). Semiquantitative and real-time PCRs were performed using increasing numbers of cycles of 95°C for 45 s, 59°C for 35 s, and 72°C for 45 s and a final extension time of 7 min. The following primers were used to quantify expression in intestinal epithelial cells: MAT2A (SAM synthetase), 5′-ATACAATCTACCACCTACAGCC-3′ (forward) and 5′-CATAAGAGACCTGAACAAGAACC-3′ (reverse); AHCY (SAH hydrolase), 5′-GGTATCGGTTGAAGAATGG-3′ (forward) and 5′-GGTACTTGTCTGGATGGGTC-3′(reverse); DNMT3B, 5′-ATAAGACACCCCCTCAAACC-3′ (forward) and 5′-TTCCCGTTCTCCCTAAAAAC-3′ (reverse); IL-8: 5′-ATGACTTCCAAGCTGGCCGTGG-3′ (forward) and 5′-CATAATTTCTGTGTTGGCGCAGTGTGG-3′ (reverse); TNF-α, 5′-TCCTTCAGACACCCTCAACC-3′ (forward) and 5′-AGGCCCCAGTTTGAATTCTT-3′ (reverse); and β-actin, 5′-GCACTCTTCCAGCCTTCCTTCC-3′ (forward) and 5′-CAGGTCTTTGCGGATGTCCACG-3′ (reverse). Transcript levels and fold change in mRNA were determined as described previously (24). Intron-spanning primer pairs for were designed by using Primer3 software (http://frodo.wi.mit.edu/). Primer properties and secondary structures including hairpins, self-dimers, and cross-dimers were evaluated in a second step using Netprimer software (http://www.premierbiosoft.com/netprimer). Samples were controlled for β-actin.

HeLa cells were passaged into 24-well plates and allowed to attach for 24 h. Transient transfection of Hela cells and assessment of luciferase activity was carried out as described previously (22).

Dextran sodium sulfate (DSS) colitis was induced with a modification of the technique of Okayasu et al. (25). Colitis was induced on day 0 by the addition of 3% DSS (m.w. = 36,000–50,000; MP Biomedicals, Illkirch, France) solution in drinking water. Control animals received water alone. DZ2002 was delivered i.p. at a concentration of 50 mg/kg/day (19) beginning 1 d prior (day −1) to DSS administration. Folic acid (Sigma-Aldrich) was delivered i.p. at a concentration of 50 mg/kg beginning 1 d prior (day −1) to DSS administration and then every other day for the course of the experiment. BAY 11-7082 (Cayman Chemical) was administered i.p. at a concentration of 5 mg/kg beginning 1 d prior (day −1) to DSS administration and then every other day for the course of the experiment, as described previously (26).

For cytokine analysis, colonic tissue was extracted in Tris lysis buffer by sonication, and protein homogenates were stored at −80°C until use. Tissue concentrations of IFN-γ were measured in colonic protein extracts using a proinflammatory cytokine screen (Meso Scale Discovery). Assays were performed per manufacturer’s instructions and analyzed using a Sector Imager 2400 (Meso Scale Discovery). IFN-γ concentrations were normalized to total protein concentration.

5-Methyl cytidine staining was quantified using flow cytometry essentially as described by Watson et al. (27). Briefly, cells were trypsinized and fixed in 70% MeOH. Following fixation, cells were incubated in 1 M HCl for 1 h at 37°C. Cells were incubated with anti–5-methyl cytidine Ab (Abcam) or IgG control at identical concentrations, followed by application of Alexa Fluor 555 (Invitrogen) secondary Ab. Cells were analyzed using the FACSCanto system (BD Immunocytometry Systems, San Jose, CA). Postanalyses were performed using FlowJo software (Tree Star, Ashland, OR).

T84 human epithelial cells were cultured on glass coverslips and fixed in ice-cold methanol, followed by permeablization in 0.2% Triton X-100. Cells were incubated in 1 M HCl for 1 h at 37°C. Cells were incubated with blocking buffer (1% BSA) and then incubated with anti–5-methyl cytidine Ab (Abcam) or IgG-negative control, followed by Alexa Fluor 555 secondary Ab. Coverslips were mounted using ProLong Gold antifade (Invitrogen). Staining was visualized using Axioplan Zeiss microscope equipped with an AxioCam MRc5 system. Imaging software for data capture was Axiovision 4.6.

Blank sections were cut from formalin-fixed, paraffin-embedded samples. Following deparaffinization, slides were subjected to Ag retrieval by using a sodium citrate buffer. Sections were permeabilized using 2% Triton X-100, followed by blocking in 1% BSA. Sections were incubated with anti–5-methyl cytidine Ab, followed by incubation with Alexa Fluor 555 secondary Ab. Sections were counterstained with DAPI (Invitrogen). Images were visualized and captured as described above.

Collected cell pellets or frozen tumor specimens were homogenated in ice-cold 8% perchloric acid as described previously (28, 29). Briefly, after centrifugation, the supernatants (containing hydrophilic metabolites) were collected, and pH was adjusted to pH 7 by using KOH. The potassium perchlorate was removed by centrifugation, and the hydrophilic fraction was lyophilized overnight. The cell and tissue pellets (after the first centrifugation), which contained the lipophilic metabolites, were redissolved in water, and the pH was adjusted (7.0). The lipophilic fraction was lyophilized overnight. For the cell experiments only, previously collected media were lyophilized overnight after adjusting pH. The dried hydrophilic cell and tissue extracts were redissolved in 0.5 ml deuterium oxide, transferred into 5-mm NMR tubes, and used for [1H]-, [31P]-, and [13C]-NMR analysis. The medium extracts were redissolved in 1 ml deuterium oxide, transferred to 5-mm NMR tubes, and underwent [1H]- and [13C]-NMR analysis. The cell and tissue lipid extracts were redissolved in 1.2 ml deuterated chloroform/deuterated methanol mixture (2:1 v/v).

All [1H]- and [13C]-NMR spectra were obtained by the Bruker 500 MHz DRX NMR spectrometer using an inverse Bruker 5-mm TXI probe. To assist [1H]-NMR peak assignment and metabolite identification in cell, media and biopsy extracts, two-dimensional–hydrogen/carbon-heteronuclear single quantum correlation NMR techniques were used. All spectra were Fourier transformed, and lactate (Lac3, CH3) was used as an internal chemical reference for both carbon (21 ppm) and proton (1.32 ppm) axes. For metabolite quantification, one-dimensional [1H]-NMR spectra were obtained from each sample, with a standard water presaturation pulse program “zgpr”. A thin sealed glass capillary, containing trimethylsilyl proprionate, was placed in each 5-mm tube prior to [1H]-NMR experiments. The total number of acquisitions varied from 40 to 128. Conventional [1H] acquisition parameters were power level pl1 = 20 dB, power angle p1 = 6.3 ms (90° pulse), power level for water presaturation pl9 = 77 dB, water suppression at O1P = 4.76 ppm, and spectral width SW = 5000 MHz, and the pulse delay of 12.75 s (calculated as 5*T1) was applied between acquisitions. The trimethylsilyl proprionate from the reference capillary served as a chemical shift (0 ppm) and proton metabolite concentration reference. The cell and media extracts subsequently underwent [13C]-NMR analysis (with proton decoupling). The total number of scans was 24,000 and 800 for each cell and medium extract, respectively. The concentration of [3-13C] lactate at 21 ppm (calculated from two satellite peaks at the [1H]-NMR spectra) served as an internal carbon concentration reference.

Before [31P]-NMR analysis, 100 mmol/l EDTA was added to the extracts to complex divalent cations (for ATP and ADP quantification). All [31P]-NMR spectra (with proton decoupling) on cell and tissue extracts were obtained by the Bruker 300 MHz Avance NMR spectrometer using a Bruker QNP probe. The total number of scans was 8,000–16,000 per extract. A thin capillary glass containing 2.3 mmol/l methyl-diphosphoric acid served as a chemical shift (18.6 ppm) and phosphor metabolite concentration reference.

Histological examination was performed on three samples of the distal colon. Samples were fixed in 4% formalin before staining with H&E. All histological quantitation was performed in a blinded fashion, using a previously described scoring system (30). Severity of inflammation: 0, rare inflammatory cells in the lamina propria; 1, increased numbers of inflammatory cells; 2, confluence of inflammatory cells extending into the submucosa; 3, transmural extension of the inflammatory cell infiltrate. Extent of injury: 0, nil mucosal damage; 1, discrete lymphoepithelial lesions; 2, surface mucosal erosion; 3, widespread mucosal ulceration and extension through deeper bowel wall structures. The scores of the two parameters were added and the mean was calculated. Maximum possible score was 3.

Unpaired t test and/or one-way ANOVA test were used to determine differences between groups, as indicated. The significance level was set at p < 0.05 for all tests (Prism 4; GraphPad Software, San Diego, CA).

Previous studies have shown that inflammatory lesions associated with IBD are the site of significant metabolic changes (3133). These changes in metabolism, however, have not been studied in a systematic manner. In this study, we sought to define the metabolic changes associated with modeled inflammation. Because previous reports strongly implicated intestinal epithelial cells (23, 32), we used T84 cells, a well characterized human intestinal epithelial cell line. Initial experiments involved subjecting T84 human colonic epithelial cells to IFN-γ for 48 h. In addition, cells were cultured in the presence of [1-13C]glucose for the final 4 h of culture to obtain data on the fate of glucose during inflammation. Following the incubation period, cells were immediately harvested and prepared for magnetic resonance spectroscopy (MRS) analysis. The media were also preserved and subjected to metabolite quantification. It has been previously shown that intestinal inflammation results in a state termed “inflammatory hypoxia” (32). In addition, it has been previously shown that treatment of human epithelial cells with proinflammatory cytokines leads to an increase in glycolytic rate (34, 35). The present study corroborates these studies with the finding that treatment of T84 cells with IFN-γ results in an increase in glucose uptake and lactate production as well as intracellular adenosine levels, all of which are indicative of hypoxia (Fig. 1A, Supplemental Fig. 1, Supplemental Table I). Importantly, we also found changes in a number of metabolites that participate in (betaine and cholines) or are products of (creatine, taurine, and adenosine) cellular methylation reactions (Fig. 1A, Supplemental Fig. 1, Supplemental Table I). These results strongly implicate methylation reactions as a contributing factor to mucosal inflammatory responses. Moving forward, we compared these results with those obtained from microarray analysis (Fig. 1B).

FIGURE 1.

Induction of SAM synthetase and SAH hydrolase during modeled inflammation. T84 human epithelial cells were cultured in the presence of IFN-γ (10 ng/ml) for the indicated times. A displays absolute metabolite concentrations for a subset of the measured metabolites from all treatment groups. B displays a heat-map depiction of gene expression data from microarray analysis showing both control (IFN-γ–inducible) and methylation-related genes. C displays real-time PCR and Western blot analysis for SAM synthetase, and D shows same analysis for SAH hydrolase. PCR data (n = 3; *p < 0.05, ***p < 0.005). Western blot (n = 2).

FIGURE 1.

Induction of SAM synthetase and SAH hydrolase during modeled inflammation. T84 human epithelial cells were cultured in the presence of IFN-γ (10 ng/ml) for the indicated times. A displays absolute metabolite concentrations for a subset of the measured metabolites from all treatment groups. B displays a heat-map depiction of gene expression data from microarray analysis showing both control (IFN-γ–inducible) and methylation-related genes. C displays real-time PCR and Western blot analysis for SAM synthetase, and D shows same analysis for SAH hydrolase. PCR data (n = 3; *p < 0.05, ***p < 0.005). Western blot (n = 2).

Close modal

After finding that key metabolites of methylation reactions were changed in our in vitro model of mucosal inflammation, we compared these metabolomic end points with microarray data generated in T84 intestinal epithelial cells following exposure to IFN-γ for either 6 or 18 h. As shown in Fig. 1B, a number of genes strongly implicated in IFN-γ signaling (e.g., CXL11, CXCL10, and IDO1) were highly induced by IFN-γ and thus served as positive controls for this microarray. More relevant for the current study, this microarray also revealed strong induction of several methylation-related gene transcripts, including SAM synthetase (MAT2A), SAH hydrolase (AHCY), O-6-methylguanine-DNA methyltransferase, and guanidinoacetate N-methyltransferase. MAT2A, the enzyme responsible for SAM production, was among the most highly upregulated genes in the array (Fig. 1B). Given its central role in all methylation reactions (14), we pursued the induction of MAT2A by IFN-γ. To confirm this microarray result, we performed real-time PCR using primers specific for the MAT2A and examined SAM synthetase protein levels by immunoblot analysis. These results indicate that both MAT2A expression and SAM synthetase protein are increased in epithelial cells following exposure to IFN-γ (Fig. 1C, for both p < 0.05 by ANOVA). In a similar manner, we also examined the expression of SAH hydrolase. This enzyme plays a key role in methylation reactions by catabolizing SAH, a potent methyltransferase inhibitor, to homocysteine and adenosine (14). These studies revealed that SAH hydrolase expression and protein levels are also upregulated during inflammation (Fig. 1D). Taken together, these results and that of metabolomic analysis provide strong evidence that cellular methylation pathways are important in the inflammatory process.

Having determined that key enzymes of cellular methylation are upregulated during inflammation, we next examined functional end points of these reactions, namely DNA methylation. For these studies, T84 epithelial cells were exposed to IFN-γ (10 ng/ml) for the indicated times and analyzed levels of 5′-methylcytidine, which is indicative of DNA methylation, using fluorescent microscopy and flow cytometry. As depicted in Fig. 2A, untreated T84 cells exhibit a small amount of nuclear 5′-methylcytidine staining. In stark contrast, as shown in Fig. 2B, nuclear staining for 5′-methylcytidine is substantially increased following exposure to IFN-γ for 24 h. This result was corroborated using flow cytometry. As depicted in Fig. 2C, incubation of T84 cells for 24 h with IFN-γ resulted in a 2.23 ± 0.02 increase in 5′-methylcytidine (p < 0.01). An insight into a potential mechanism for the increase in DNA methylation came when the expression of DNMT3b, a DNA methyltransferase responsible for de novo DNA methylation (14), was examined in cells exposed to IFN-γ. As shown in Fig. 2D, DNMT3b expression is significantly induced in T84 cells in an inflammatory setting (p < 0.01 by ANOVA). These data indicate that DNA methylation is increased in an in vitro model of inflammation.

FIGURE 2.

Increase in DNA methylation during inflammation. T84 human epithelial cells were cultured in the presence of IFN-γ (10 ng/ml) for the indicated times. A shows nuclear staining using fluorescent microscopy using an Ab specific for 5′-methylcytidine. B displays nuclear staining for 5′-methylcytidine in T84 cells incubated with IFN-γ for 24 h. C shows flow cytometry data for control or IFN-γ–exposed T84 cells using an anti–5′-methylcytidine Ab or an isotype-matched negative control (n = 3). D depicts real-time PCR data for DNMT3b in control and IFN-γ–exposed T84 cells for 2, 6, 12, and 18 h (n = 3; *p < 0.05, **p < 0.01).

FIGURE 2.

Increase in DNA methylation during inflammation. T84 human epithelial cells were cultured in the presence of IFN-γ (10 ng/ml) for the indicated times. A shows nuclear staining using fluorescent microscopy using an Ab specific for 5′-methylcytidine. B displays nuclear staining for 5′-methylcytidine in T84 cells incubated with IFN-γ for 24 h. C shows flow cytometry data for control or IFN-γ–exposed T84 cells using an anti–5′-methylcytidine Ab or an isotype-matched negative control (n = 3). D depicts real-time PCR data for DNMT3b in control and IFN-γ–exposed T84 cells for 2, 6, 12, and 18 h (n = 3; *p < 0.05, **p < 0.01).

Close modal

NF-κB is one of the master regulators of proinflammatory gene expression (for review, see Ref. 36). NF-κB upregulates a number of genes including proinflammatory cytokines, chemokines, and adhesion molecules and can induce specific sets of genes in response to a particular triggers (37). NF-κB has been shown by EMSA and IHC studies to be highly activated at the site of inflammation in a number of diseases, including IBD (36), as wells as activated under conditions of hypoxia (38, 39). Additionally, NF-κB has been found to be protective of the mucosal epithelium in murine disease models (40, 41). Interestingly, NF-κB has been shown to elicit epigenetic modifications of its inducible genes (42). Based on these studies and our preliminary data suggesting a link between methylation and the inflammatory response in epithelial cell models, we examined NF-κB activity using a luciferase reporter assay following inhibition of methylation by DZ2002, a potent reversible SAH hydrolase inhibitor (21). As shown in Fig. 3A, IFN-γ elicited a >8-fold increase in NF-κB activity at the highest concentrations (p < 0.01). Pretreatment with DZ2002 effectively inhibited NF-κB activity in a concentration-dependent manner (Fig. 3A). As IFN-γ does not represent a canonical activator of NF-κB, we also studied the impact of DZ2002 on NF-κB following treatment with TNF-α. As shown in Fig. 3B, although TNF-α is a much more potent NF-κB activator (maximal 58.02 ± 9.66; p < 0.001), inhibition of methylation significantly repressed such induction, likely indicating that this inhibition of NF-κB by DZ2002 is not stimulus dependent.

FIGURE 3.

NF-κB activity is inhibited by DZ2002. A and B, Cells were cultured in 24-well plates for 24 h and then transfected with an NF-κB luciferase reporter construct. Cells were then treated with DZ2002 or vehicle for 1 h at the indicated concentrations, followed by addition of IFN-γ at the indicated concentrations (A) or TNF-α (10 ng/ml) (B). Cells were incubated for 12 h and immediately prepared for analysis of luciferase expression (n = 3, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). C, Western blot analysis of IKBα following exposure to IFN-γ (10 ng/ml) or DZ2002 (100 μM) and IFN-γ for the indicated time periods (n = 3). D and E, Real-time PCR analysis of TNF-α (D) and IL-8 (E) expression in T84 cells following IFN-γ treatment (10 ng/ml) or DZ2002 and IFN-γ for the indicated time periods. Values are expressed as fold increase over vehicle-treated control (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001).

FIGURE 3.

NF-κB activity is inhibited by DZ2002. A and B, Cells were cultured in 24-well plates for 24 h and then transfected with an NF-κB luciferase reporter construct. Cells were then treated with DZ2002 or vehicle for 1 h at the indicated concentrations, followed by addition of IFN-γ at the indicated concentrations (A) or TNF-α (10 ng/ml) (B). Cells were incubated for 12 h and immediately prepared for analysis of luciferase expression (n = 3, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). C, Western blot analysis of IKBα following exposure to IFN-γ (10 ng/ml) or DZ2002 (100 μM) and IFN-γ for the indicated time periods (n = 3). D and E, Real-time PCR analysis of TNF-α (D) and IL-8 (E) expression in T84 cells following IFN-γ treatment (10 ng/ml) or DZ2002 and IFN-γ for the indicated time periods. Values are expressed as fold increase over vehicle-treated control (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001).

Close modal

To confirm that DZ2002 directly impacts NF-κB, we next examined the fate of the NF-κB inhibitory protein, IκBα, in response to DZ2002. As shown in Fig. 3C, IκBα levels are decreased in response to IFN-γ treatment. This response is abrogated with pretreatment with DZ2002 indicating that the inhibition of NF-κB is mediated, at least in part, through modulation of the inhibitory protein IκBα. To confirm these results at a functional level, we examined the expression of TNF-α and IL-8, two known NF-κB target genes. As depicted in Fig. 3D and 3E, expression of both TNF-α and IL-8 is increased in response to IFN-γ treatment. In support of our hypothesis, expression of both of these targets was diminished upon pretreatment with DZ2002 (p < 0.05). These results strongly implicate methylation in NF-κB activation, at least in part, through the regulation of IκBα.

Previous studies have shown that inflammatory lesions in mucosal inflammation (e.g., IBD) result in substantial metabolic changes and major shifts in metabolite supply and demand (10). In this study, we sought to define the metabolic changes induced during inflammation in an epithelial-driven in vivo model of experimental colitis. For these purposes, we selected the DSS model. C57BL/6 mice were administered DSS or vehicle (water) alone for 5 d, after which mice were euthanized, and colon tissue was immediately removed and frozen. Colons were extracted and prepared for MRS analysis. Fig. 4 displays representative proton spectra for both control (Fig. 4A) and DSS-treated (Fig. 4B) colonic tissue. These experiments revealed alterations in a number of metabolites associated with methylation reactions, including adenosine, betaine, choline, creatine, glutathione (p < 0.05; n = 6) (Fig. 4C, Supplemental Table II). In addition to an impact on metabolites involved in methylation, changes in several metabolites are indicative of hypoxia (glucose, lactate, and adenosine), consistent with previous work in experimental colitis models (23, 32, 33, 43). To confirm our in vitro experiments, we examined the parallel expression of SAM synthetase/SAH hydrolase and IFN-γ in colonic tissue extracts from control and DSS-treated mice harvested at 2 and 4 d of treatment. As shown in Fig. 4D, both SAM synthetase and SAH hydrolase expression were increased after 2 and 4 d of DSS administration. Likewise, tissue concentrations of IFN-γ were increased at day 4 of DSS treatment (Fig. 4E).

FIGURE 4.

Metabolic analysis of murine colonic inflammation. Wild-type mice received DSS for 5 d. Tissue was extracted and used for MRS analysis of endogenous metabolites. A and B, Representative proton spectra from control (A) and DSS-treated (3%, 6-d treatment) colonic tissue extracts. Numbered peaks are as follows: 1, glucose; 2, inositol; 3, betaine; 4, total cholines; 5, total creatine; 6, total glutathione; 7, succinate; 8, lactate; and 9, leucine, isoleucine, and valine. C displays absolute metabolite concentrations for a subset of the measured metabolites for control and DSS-treated tissue. D, Western blot analysis of SAM synthetase and SAH hydrolase in colonic tissue extracts from control and DSS-treated animals (three animals per treatment). E, MesoScale analysis of IFN-γ concentrations in colonic tissue from control (day 0) and DSS-treated animals (2 and 4 d of DSS) (n = 4 animals/group).

FIGURE 4.

Metabolic analysis of murine colonic inflammation. Wild-type mice received DSS for 5 d. Tissue was extracted and used for MRS analysis of endogenous metabolites. A and B, Representative proton spectra from control (A) and DSS-treated (3%, 6-d treatment) colonic tissue extracts. Numbered peaks are as follows: 1, glucose; 2, inositol; 3, betaine; 4, total cholines; 5, total creatine; 6, total glutathione; 7, succinate; 8, lactate; and 9, leucine, isoleucine, and valine. C displays absolute metabolite concentrations for a subset of the measured metabolites for control and DSS-treated tissue. D, Western blot analysis of SAM synthetase and SAH hydrolase in colonic tissue extracts from control and DSS-treated animals (three animals per treatment). E, MesoScale analysis of IFN-γ concentrations in colonic tissue from control (day 0) and DSS-treated animals (2 and 4 d of DSS) (n = 4 animals/group).

Close modal

We next examined the impact of methylation-inhibition on DSS colitis outcomes. Given that the SAH hydrolase (AHCY)-null mouse is embryonic lethal (44) and that SAM synthetase is not easily inhibited pharmacologically (15), we reverted to a pharmacological approach using DZ2002. Our in vitro data revealed that modeled inflammation results in an increase in DNA methylation in epithelial models, as evidenced by an increase in 5′-methylcytidine staining detected both by immunocytochemistry and flow cytometry (Fig. 2). On the basis of these findings, we initially examined the extent of DNA methylation in response to DSS in the presence and absence of DZ2002 in vivo. As shown in Fig. 5, treatment with DSS resulted in a significant increase in colonic DNA methylation. Colonic tissue from vehicle-treated animals displayed low levels of basal 5-methylcytidine staining (Fig. 5, column 1). By contrast, DSS induction resulted in a substantial increase in DNA methylation as indicated by the increase in 5′-methylcytidine and DAPI/5′-methylcytidine costaining (see arrows in merged image, column 2). Treatment of animals with DZ2002 in combination with DSS significantly dampened DNA methylation as indicated by the decrease in both 5′-methylcytidine staining as well as DAPI/5′-methylcytidine colocalization (see arrows in merged image, column 3).

FIGURE 5.

DSS treatment increases DNA methylation in vivo. Twelve-week-old C57BL/6 mice were treated as described above. Tissue sections were prepared as previously described (32), followed by labeling with anti–5′-methylcytidine Ab (Abcam) and detection with anti-rabbit Alexa Fluor 555 secondary Ab. Tissues were counterstained with DAPI. Arrows in merged image indicate representative cells displaying staining for both 5′-methylcytidine and DAPI.

FIGURE 5.

DSS treatment increases DNA methylation in vivo. Twelve-week-old C57BL/6 mice were treated as described above. Tissue sections were prepared as previously described (32), followed by labeling with anti–5′-methylcytidine Ab (Abcam) and detection with anti-rabbit Alexa Fluor 555 secondary Ab. Tissues were counterstained with DAPI. Arrows in merged image indicate representative cells displaying staining for both 5′-methylcytidine and DAPI.

Close modal

We next demonstrated that DZ2002, administered in combination with administration of DSS, exacerbated disease progression. As shown in Fig. 6A–C, inhibition of methylation in combination with DSS treatment resulted in significant decrease in weight (p < 0.05; Fig. 6A), increased colon shortening (p < 0.001; Fig. 6B), and an increase in disease activity as indicated by histological score (p < 0.001; Fig. 6C). Importantly, DZ2002 treatment alone resulted in no weight loss or colon shortening (Fig. 6A, 6B), indicating that this inhibitor is not toxic at the doses administered. In addition, these indicators of disease severity were confirmed by histology. As shown in Fig. 6D, vehicle-treated mice display normal tissue morphology, including intact epithelium and crypt structure. DSS-treated animals exhibited a deterioration in normal crypt architecture and epithelial cell depletion (Fig. 6E), and animals treated with DZ2002 and DSS display an almost complete lack of normal tissue architecture and loss of the epithelium (Fig. 6F). These results confirm that inhibition of methylation worsens disease progression in a murine model of colitis and strongly implicate that inflammation-associated methylation functions as an endogenous protective mechanism. Importantly, the finding that NF-κB inhibition exacerbates epithelial-driven colitis is consistent with recent literature (45, 46).

FIGURE 6.

DSS colitis is enhanced by DZ2002. Twelve- to 18-wk-old C57BL/6 mice were i.p. injected daily with vehicle (0.4% DMSO in PBS) or DZ2002 (50 mg/kg) beginning at day 0. From day 1, mice were administered water (control) or 3% DSS ad libitum for 6 d. A displays percent weight loss, B displays colon length; and C represents histological score (n = 5, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). Histological score determined as described previously (33). D and E, H&E staining of tissue isolated from vehicle (D), DSS (E), and DSS + DZ2002 (F) treated animals.

FIGURE 6.

DSS colitis is enhanced by DZ2002. Twelve- to 18-wk-old C57BL/6 mice were i.p. injected daily with vehicle (0.4% DMSO in PBS) or DZ2002 (50 mg/kg) beginning at day 0. From day 1, mice were administered water (control) or 3% DSS ad libitum for 6 d. A displays percent weight loss, B displays colon length; and C represents histological score (n = 5, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). Histological score determined as described previously (33). D and E, H&E staining of tissue isolated from vehicle (D), DSS (E), and DSS + DZ2002 (F) treated animals.

Close modal

In an attempt to clarify the role of the NF-κB pathway in the context of our findings, we administered the NF-κB inhibitor BAY 11-7082 to mice, alone or in combination with DSS. This inhibitor selectively and irreversibly inhibits NF-κB activation by blocking cytokine-induced phosphorylation of IκB-α without influencing constitutive IκB-α phosphorylation (47). Although animals receiving BAY 11-7082 alone showed no symptomology compared with vehicle controls, mice receiving both BAY 11-7082 and DSS developed significantly more severe disease, to the extent that it was necessary that we humanely sacrifice animals prior to completion of the DSS time course (data not shown). These experiments do not provide direct evidence of a role for NF-κB activation in the protection of the mucosa in a DSS model of colitis. However, the results do corroborate our findings and those of others indicating that NF-κB is protective within the intestinal epithelium (40, 41, 48) and suggest that the endogenously protective effect of methylation may be due, at least in part, to epithelial NF-κB activation.

Having shown that inhibition of methylation using DZ2002 in combination with administration of DSS worsens colitis in mice, we next determined whether augmentation of methylation would be protective in the DSS model. For these experiments, we administered folate in the same manner as the methylation inhibitor (i.e., systemic via i.p. injection) to make the experiments as comparable as possible. As shown in Fig. 7A–C, administration of folate in combination with DSS treatment resulted in an increase in weight (p < 0.05; Fig. 7A), significantly decreased colon shortening (p < 0.05; Fig. 7B), and a significant decrease in disease activity as indicated by histological score (p < 0.01; Fig. 7C). Administration of folate alone had no effect on weight loss or colon length (Fig. 7A, 7B). Once again, these indicators of disease severity were confirmed by histology. As shown in Fig. 7D, vehicle-treated mice display normal tissue morphology, including intact epithelium and crypt structure. DSS-treated animals exhibited deterioration in normal crypt architecture and epithelial cell depletion (Fig. 7E). Importantly, animals administered folate in combination with DSS display improved tissue architecture, diminished loss of the epithelium, and less inflammatory cell migration than DSS alone (Fig. 6F). These results confirm that augmentation of methylation through the administration of folic acid ameliorates disease progression and provides further evidence that inflammation-associated methylation is protective in a murine colitic model. These data are in agreement with clinical studies demonstrating vitamin B12 and folate deficiencies are common in IBD patients (4951) and suggest folate supplementation in IBD treatment. In addition, a recent study demonstrated that a methyl-deficient diet exacerbates disease in an animal model of colitis (52).

FIGURE 7.

DSS colitis is reduced in folic acid-treated animals. Eight- to 10-wk-old C57BL/6 mice were i.p. injected every other day with vehicle (PBS) or folic acid (50 mg/kg) beginning at day 0. From day 1, mice were administered water (control) or 3% DSS ad libitum for 6 d. A displays percent weight loss; B displays colon length; and C represents histological score (n = 4, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). Histological score determined as described previously (33). D and E, H&E staining of tissue isolated from vehicle (D), DSS (E), and DSS + folic acid (F)-treated animals.

FIGURE 7.

DSS colitis is reduced in folic acid-treated animals. Eight- to 10-wk-old C57BL/6 mice were i.p. injected every other day with vehicle (PBS) or folic acid (50 mg/kg) beginning at day 0. From day 1, mice were administered water (control) or 3% DSS ad libitum for 6 d. A displays percent weight loss; B displays colon length; and C represents histological score (n = 4, data are expressed as mean ± SD; *p < 0.05, **p < 0.01, ***p < 0.005). Histological score determined as described previously (33). D and E, H&E staining of tissue isolated from vehicle (D), DSS (E), and DSS + folic acid (F)-treated animals.

Close modal

This study aimed to identify metabolic changes associated with modeled intestinal inflammation, particularly related to the epithelium. As previous work had demonstrated specific shifts in metabolism during inflammation, we reasoned that such changes could be reflected on a more global basis. An amalgamated approach using NMR-based metabolomics and transcriptional arrays identified shifts in methylation-dependent pathways as a major metabolic fingerprint. These studies significantly extend previous work related to inflammation-associated metabolism and identify changes in methylation as a target signature within the epithelium during mucosal inflammation. Evidence is provided that shifts in methylation associated with inflammation serve an endogenously protective role in murine colitis.

Ongoing mucosal inflammatory responses are characterized by significant shifts in tissue metabolism (10). These changes include a shift toward a glycolytic phenotype and careful analysis has shown the development of significant hypoxia, termed inflammatory hypoxia, particularly prominent within the epithelium (53). Coinciding with inflammation-associated hypoxia is the stabilization of hypoxia inducible factor (32, 53), and given the central role of hypoxia inducible factor in most metabolic processes (54, 55), we reasoned that overall metabolism would be changed. In support of this hypothesis, global analysis of metabolism using MRS identified significant changes in a number of cellular metabolites. Many of these alterations are in agreement with results from our laboratory indicating the presence of inflammatory hypoxia. These include increased intracellular adenosine, increased glucose uptake and intracellular glucose, increased intracellular lactate and lactate export, and increased turnover of ATP (Figs. 1, 4, Supplemental Fig. 1, Supplemental Tables I, II). In addition, significant changes were observed in a number of lipid metabolites including monounsaturated fatty acids, triacylglycerol, polyunsaturated fatty acids, phosphatidyl choline, and phosphatidyl ethanolamine (Supplemental Tables I, II), which may be indicative of dynamic alterations in cellular membranes in response to inflammation. Importantly, this analysis revealed a distinct and specific methylation fingerprint. Cellular methylation reactions include modification of DNA, RNA, proteins, and lipids (56, 57). These reactions all require a methyl donor for the modification of the target. The methyl donor for the majority of these reactions is SAM (11). SAM is distributed within all cell and tissue compartments and functions as a methyl donor for a number of different methyltransferases. The donation of methyl groups results in the generation of SAH. Methyltransferase enzymes have a higher affinity for SAH than SAM, and thus, SAH functions as a potent feedback inhibitor (14). SAH is rapidly converted to homocysteine and adenosine by SAH hydrolase. On the basis of this methylation fingerprint, we performed parallel microarray analysis of modeled inflammation using a Th1 cytokine (IFN-γ) as a stimulus and discovered that SAM synthetase (MAT2A) and SAH hydrolase (AHCY) were among the top 20 highest IFN-γ–induced transcripts in T84 intestinal epithelia. The mammalian MAT2A gene promoter is relatively well-characterized. Previous studies have shown that the human MAT2A promoter is basally controlled by three tandem Sp1 sites (58). Under modeled inflammatory conditions (TNF-α activation), MAT2A is strongly induced through activation of AP-1 and NF-κB (59). These studies support our findings of MAT2A induction by IFN-γ, a known activator of both NF-κB and AP-1 (60). At present, essentially nothing is known about the regulation of the mammalian AHCY gene.

As a proof of principle, we extended these findings to a mucosal inflammation model. Given our understanding of inflammation-associated metabolic changes within the epithelium, we selected DSS colitis as an appropriate animal model to study colitis. DSS functions primarily as an epithelial irritant to drive permeability-induced colonic inflammation (61). Therefore, inhibition of SAH hydrolase represents a powerful means of inhibiting cellular methylation reactions (15). It has been known for several years that inhibition of methylation had immunosuppressive influences (16). This led to the development of more specific, reversible, and less toxic SAH inhibitors for use in animal models of inflammation. Using these compounds, it was demonstrated that SAH hydrolase inhibition particularly downregulates T cell activation and adaptive immune responses. One of these SAH hydrolase inhibitors, DZ2002, has been found to have potent immunosuppressive effects and ameliorates disease in a number of animal models including delayed-type hypersensitivity (20), arthritis (20), and experimental autoimmune encephalomyelitis (19). Although we do not know the exact mechanism by which DZ2002, we have shown that inhibition of methylation by DZ2002 inhibits NF-κB in vitro. NF-κB is a master regulator of the proinflammatory response, upregulating a number of genes including cytokines, chemokines, and adhesion molecules, as well as activated under conditions of hypoxia (62). A number of studies have shown that inhibition of NF-κB may be protective in animal models of IBD (6365). Paradoxically, however, NF-κB activation has also been found to be protective in the context of the mucosal epithelium in murine disease models (40, 41). Our findings with DZ20002 support this hypothesis and demonstrate that parenteral administration of DZ2002 significantly inhibits tissue methylation and that such inhibition is associated with exacerbated DSS colitic responses. In addition, augmentation of methylation through the administration of folate ameliorates disease in this colitic model. Whether methylation inhibition would impact other models of intestinal inflammation in the same manner is not currently known.

Taken together, these studies provide a new and compelling role for methylation as an endogenously protective mechanism for mucosal inflammation. In addition, the findings in this study provide a tractable and potentially new therapeutic opportunities for methylation-dependent targets in mucosal diseases such as IBD.

The online version of this article contains supplemental material.

Abbreviations used in this article:

DSS

dextran sodium sulfate

DZ2002

methyl 4-(adenin-9-yl)-2-hydroxybutanoate

IBD

inflammatory bowel disease

MRS

magnetic resonance spectroscopy

NMR

nuclear magnetic resonance

SAH

S-adenosylhomocysteine

SAM

S-adenosylmethionine.

1
Xavier
R. J.
,
Podolsky
D. K.
.
2007
.
Unravelling the pathogenesis of inflammatory bowel disease.
Nature
448
:
427
434
.
2
Baumgart
D. C.
,
Carding
S. R.
.
2007
.
Inflammatory bowel disease: cause and immunobiology.
Lancet
369
:
1627
1640
.
3
Sartor
R. B.
1995
.
Current concepts of the etiology and pathogenesis of ulcerative colitis and Crohn’s disease.
Gastroenterol. Clin. North Am.
24
:
475
507
.
4
Danese
S.
,
Dejana
E.
,
Fiocchi
C.
.
2007
.
Immune regulation by microvascular endothelial cells: directing innate and adaptive immunity, coagulation, and inflammation.
J. Immunol.
178
:
6017
6022
.
5
Hatoum
O. A.
,
Binion
D. G.
,
Gutterman
D. D.
.
2005
.
Paradox of simultaneous intestinal ischaemia and hyperaemia in inflammatory bowel disease.
Eur. J. Clin. Invest.
35
:
599
609
.
6
Haddad
J. J.
2003
.
Science review: redox and oxygen-sensitive transcription factors in the regulation of oxidant-mediated lung injury: role for hypoxia-inducible factor-1α.
Crit. Care
7
:
47
54
.
7
Kokura
S.
,
Yoshida
N.
,
Yoshikawa
T.
.
2002
.
Anoxia/reoxygenation-induced leukocyte-endothelial cell interactions.
Free Radic. Biol. Med.
33
:
427
432
.
8
Saadi
S.
,
Wrenshall
L. E.
,
Platt
J. L.
.
2002
.
Regional manifestations and control of the immune system.
FASEB J.
16
:
849
856
.
9
Lewis
J. S.
,
Lee
J. A.
,
Underwood
J. C.
,
Harris
A. L.
,
Lewis
C. E.
.
1999
.
Macrophage responses to hypoxia: relevance to disease mechanisms.
J. Leukoc. Biol.
66
:
889
900
.
10
Kominsky
D. J.
,
Campbell
E. L.
,
Colgan
S. P.
.
2010
.
Metabolic shifts in immunity and inflammation.
J. Immunol.
184
:
4062
4068
.
11
Baldessarini
R. J.
1987
.
Neuropharmacology of S-adenosyl-l-methionine.
Am. J. Med.
83
(
5A
):
95
103
.
12
Cantoni
G.
,
Chiang
P.
.
1980
.
The role of S-adenosylhomocysteine and S-adenosylhomocysteine hydrolase in the control of biological methylations.
In
Natural Sulfur Compounds: Novel Biochemical and Structural Aspects.
Cavallini
D.
,
Gaull
G. E.
,
Zappia
V.
, eds.
New York
,
Plenum Press
, p.
67
80
.
13
Cantoni
G. L.
,
Scarano
E.
.
1954
.
The formation of S-adenosylhomocysteine in enzymatic transmethylation reactions.
J. Am. Chem. Soc.
76
:
4744
.
14
Clarke
S.
,
Banfield
K.
.
2001
.
S-Adenosylmethionine-dependent methyltransferases.
In
Homocysteine in Health and Disease
,
Carmel
R.
,
Jacobsen
D. W.
, eds.
Cambridge University Press
,
New York
, p.
63
78
.
15
Chiang
P. K.
1998
.
Biological effects of inhibitors of S-adenosylhomocysteine hydrolase.
Pharmacol. Ther.
77
:
115
134
.
16
Wolos
J. A.
,
Frondorf
K. A.
,
Babcock
G. F.
,
Stripp
S. A.
,
Bowlin
T. L.
.
1993
.
Immunomodulation by an inhibitor of S-adenosyl-l-homocysteine hydrolase: inhibition of in vitro and in vivo allogeneic responses.
Cell. Immunol.
149
:
402
408
.
17
Fu
Y. F.
,
Wang
J. X.
,
Zhao
Y.
,
Yang
Y.
,
Tang
W.
,
Ni
J.
,
Zhu
Y. N.
,
Zhou
R.
,
He
P. L.
,
Li
C.
, et al
.
2006
.
S-Adenosyl-l-homocysteine hydrolase inactivation curtails ovalbumin-induced immune responses.
J. Pharmacol. Exp. Ther.
316
:
1229
1237
.
18
Fu
Y. F.
,
Zhu
Y. N.
,
Ni
J.
,
Zhong
X. G.
,
Tang
W.
,
Re
Y. D.
,
Shi
L. P.
,
Wan
J.
,
Yang
Y. F.
,
Yuan
C.
, et al
.
2006
.
A reversible S-adenosyl-l-homocysteine hydrolase inhibitor ameliorates experimental autoimmune encephalomyelitis by inhibiting T cell activation.
J. Pharmacol. Exp. Ther.
319
:
799
808
.
19
Lawson
B. R.
,
Manenkova
Y.
,
Ahamed
J.
,
Chen
X.
,
Zou
J. P.
,
Baccala
R.
,
Theofilopoulos
A. N.
,
Yuan
C.
.
2007
.
Inhibition of transmethylation down-regulates CD4 T cell activation and curtails development of autoimmunity in a model system.
J. Immunol.
178
:
5366
5374
.
20
Saso
Y.
,
Conner
E. M.
,
Teegarden
B. R.
,
Yuan
C. S.
.
2001
.
S-Adenosyl-l-homocysteine hydrolase inhibitor mediates immunosuppressive effects in vivo: suppression of delayed type hypersensitivity ear swelling and peptidoglycan polysaccharide-induced arthritis.
J. Pharmacol. Exp. Ther.
296
:
106
112
.
21
Wu
Q. L.
,
Fu
Y. F.
,
Zhou
W. L.
,
Wang
J. X.
,
Feng
Y. H.
,
Liu
J.
,
Xu
J. Y.
,
He
P. L.
,
Zhou
R.
,
Tang
W.
, et al
.
2005
.
Inhibition of S-adenosyl-l-homocysteine hydrolase induces immunosuppression.
J. Pharmacol. Exp. Ther.
313
:
705
711
.
22
Khoury
J.
,
Ibla
J. C.
,
Neish
A. S.
,
Colgan
S. P.
.
2007
.
Antiinflammatory adaptation to hypoxia through adenosine-mediated cullin-1 deneddylation.
J. Clin. Invest.
117
:
703
711
.
23
Synnestvedt
K.
,
Furuta
G. T.
,
Comerford
K. M.
,
Louis
N.
,
Karhausen
J.
,
Eltzschig
H. K.
,
Hansen
K. R.
,
Thompson
L. F.
,
Colgan
S. P.
.
2002
.
Ecto-5′-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia.
J. Clin. Invest.
110
:
993
1002
.
24
Eltzschig
H. K.
,
Ibla
J. C.
,
Furuta
G. T.
,
Leonard
M. O.
,
Jacobson
K. A.
,
Enjyoji
K.
,
Robson
S. C.
,
Colgan
S. P.
.
2003
.
Coordinated adenine nucleotide phosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role of ectonucleotidases and adenosine A2B receptors.
J. Exp. Med.
198
:
783
796
.
25
Okayasu
I.
,
Hatakeyama
S.
,
Yamada
M.
,
Ohkusa
T.
,
Inagaki
Y.
,
Nakaya
R.
.
1990
.
A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice.
Gastroenterology
98
:
694
702
.
26
Magness
S. T.
,
Jijon
H.
,
Van Houten Fisher
N.
,
Sharpless
N. E.
,
Brenner
D. A.
,
Jobin
C.
.
2004
.
In vivo pattern of lipopolysaccharide and anti–CD3-induced NF-κB activation using a novel gene-targeted enhanced GFP reporter gene mouse.
J. Immunol.
173
:
1561
1570
.
27
Watson
J. A.
,
Watson
C. J.
,
McCrohan
A. M.
,
Woodfine
K.
,
Tosetto
M.
,
McDaid
J.
,
Gallagher
E.
,
Betts
D.
,
Baugh
J.
,
O’Sullivan
J.
, et al
.
2009
.
Generation of an epigenetic signature by chronic hypoxia in prostate cells.
Hum. Mol. Genet.
18
:
3594
3604
.
28
Gottschalk
S.
,
Anderson
N.
,
Hainz
C.
,
Eckhardt
S. G.
,
Serkova
N. J.
.
2004
.
Imatinib (STI571)-mediated changes in glucose metabolism in human leukemia BCR-ABL–positive cells.
Clin. Cancer Res.
10
:
6661
6668
.
29
Serkova
N.
,
Fuller
T. F.
,
Klawitter
J.
,
Freise
C. E.
,
Niemann
C. U.
.
2005
.
H-NMR–based metabolic signatures of mild and severe ischemia/reperfusion injury in rat kidney transplants.
Kidney Int.
67
:
1142
1151
.
30
Hartmann
G.
,
Bidlingmaier
C.
,
Siegmund
B.
,
Albrich
S.
,
Schulze
J.
,
Tschoep
K.
,
Eigler
A.
,
Lehr
H. A.
,
Endres
S.
.
2000
.
Specific type IV phosphodiesterase inhibitor rolipram mitigates experimental colitis in mice.
J. Pharmacol. Exp. Ther.
292
:
22
30
.
31
Furuta
G. T.
,
Turner
J. R.
,
Taylor
C. T.
,
Hershberg
R. M.
,
Comerford
K.
,
Narravula
S.
,
Podolsky
D. K.
,
Colgan
S. P.
.
2001
.
Hypoxia-inducible factor 1-dependent induction of intestinal trefoil factor protects barrier function during hypoxia.
J. Exp. Med.
193
:
1027
1034
.
32
Karhausen
J.
,
Furuta
G. T.
,
Tomaszewski
J. E.
,
Johnson
R. S.
,
Colgan
S. P.
,
Haase
V. H.
.
2004
.
Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis.
J. Clin. Invest.
114
:
1098
1106
.
33
Robinson
A.
,
Keely
S.
,
Karhausen
J.
,
Gerich
M. E.
,
Furuta
G. T.
,
Colgan
S. P.
.
2008
.
Mucosal protection by hypoxia-inducible factor prolyl hydroxylase inhibition.
Gastroenterology
134
:
145
155
.
34
Berg
S.
,
Sappington
P. L.
,
Guzik
L. J.
,
Delude
R. L.
,
Fink
M. P.
.
2003
.
Proinflammatory cytokines increase the rate of glycolysis and adenosine-5′-triphosphate turnover in cultured rat enterocytes.
Crit. Care Med.
31
:
1203
1212
.
35
Scharte
M.
,
Han
X.
,
Bertges
D. J.
,
Fink
M. P.
,
Delude
R. L.
.
2003
.
Cytokines induce HIF-1 DNA binding and the expression of HIF-1–dependent genes in cultured rat enterocytes.
Am. J. Physiol. Gastrointest. Liver Physiol.
284
:
G373
G384
.
36
Li
X.
,
Stark
G. R.
.
2002
.
NFκB-dependent signaling pathways.
Exp. Hematol.
30
:
285
296
.
37
Werner
S. L.
,
Barken
D.
,
Hoffmann
A.
.
2005
.
Stimulus specificity of gene expression programs determined by temporal control of IKK activity.
Science
309
:
1857
1861
.
38
Cockman
M. E.
,
Lancaster
D. E.
,
Stolze
I. P.
,
Hewitson
K. S.
,
McDonough
M. A.
,
Coleman
M. L.
,
Coles
C. H.
,
Yu
X.
,
Hay
R. T.
,
Ley
S. C.
, et al
.
2006
.
Posttranslational hydroxylation of ankyrin repeats in IκB proteins by the hypoxia-inducible factor (HIF) asparaginyl hydroxylase, factor inhibiting HIF (FIH).
Proc. Natl. Acad. Sci. USA
103
:
14767
14772
.
39
Cummins
E. P.
,
Berra
E.
,
Comerford
K. M.
,
Ginouves
A.
,
Fitzgerald
K. T.
,
Seeballuck
F.
,
Godson
C.
,
Nielsen
J. E.
,
Moynagh
P.
,
Pouyssegur
J.
,
Taylor
C. T.
.
2006
.
Prolyl hydroxylase-1 negatively regulates IκB kinase-β, giving insight into hypoxia-induced NFκB activity.
Proc. Natl. Acad. Sci. USA
103
:
18154
18159
.
40
Chen
L. W.
,
Egan
L.
,
Li
Z. W.
,
Greten
F. R.
,
Kagnoff
M. F.
,
Karin
M.
.
2003
.
The two faces of IKK and NF-κB inhibition: prevention of systemic inflammation but increased local injury following intestinal ischemia-reperfusion.
Nat. Med.
9
:
575
581
.
41
Greten
F. R.
,
Eckmann
L.
,
Greten
T. F.
,
Park
J. M.
,
Li
Z. W.
,
Egan
L. J.
,
Kagnoff
M. F.
,
Karin
M.
.
2004
.
IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer.
Cell
118
:
285
296
.
42
Vanden Berghe
W.
,
Ndlovu
M. N.
,
Hoya-Arias
R.
,
Dijsselbloem
N.
,
Gerlo
S.
,
Haegeman
G.
.
2006
.
Keeping up NF-κB appearances: epigenetic control of immunity or inflammation-triggered epigenetics.
Biochem. Pharmacol.
72
:
1114
1131
.
43
Cummins
E. P.
,
Seeballuck
F.
,
Keely
S. J.
,
Mangan
N. E.
,
Callanan
J. J.
,
Fallon
P. G.
,
Taylor
C. T.
.
2008
.
The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis.
Gastroenterology
134
:
156
165
.
44
Miller
M. W.
,
Duhl
D. M.
,
Winkes
B. M.
,
Arredondo-Vega
F.
,
Saxon
P. J.
,
Wolff
G. L.
,
Epstein
C. J.
,
Hershfield
M. S.
,
Barsh
G. S.
.
1994
.
The mouse lethal nonagouti (a(x)) mutation deletes the S-adenosylhomocysteine hydrolase (Ahcy) gene.
EMBO J.
13
:
1806
1816
.
45
Eckmann
L.
,
Nebelsiek
T.
,
Fingerle
A. A.
,
Dann
S. M.
,
Mages
J.
,
Lang
R.
,
Robine
S.
,
Kagnoff
M. F.
,
Schmid
R. M.
,
Karin
M.
, et al
.
2008
.
Opposing functions of IKKβ during acute and chronic intestinal inflammation.
Proc. Natl. Acad. Sci. USA
105
:
15058
15063
.
46
Zaph
C.
,
Troy
A. E.
,
Taylor
B. C.
,
Berman-Booty
L. D.
,
Guild
K. J.
,
Du
Y.
,
Yost
E. A.
,
Gruber
A. D.
,
May
M. J.
,
Greten
F. R.
, et al
.
2007
.
Epithelial-cell-intrinsic IKK-β expression regulates intestinal immune homeostasis.
Nature
446
:
552
556
.
47
Pierce
J. W.
,
Schoenleber
R.
,
Jesmok
G.
,
Best
J.
,
Moore
S. A.
,
Collins
T.
,
Gerritsen
M. E.
.
1997
.
Novel inhibitors of cytokine-induced IκBα phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo.
J. Biol. Chem.
272
:
21096
21103
.
48
Cummins
E. P.
,
Seeballuck
F.
,
Keely
S. J.
,
Mangan
N. E.
,
Callanan
J. J.
,
Fallon
P. G.
,
Taylor
C. T.
.
2008
.
The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis.
Gastroenterology
134
:
156
165
.
49
Erzin
Y.
,
Uzun
H.
,
Celik
A. F.
,
Aydin
S.
,
Dirican
A.
,
Uzunismail
H.
.
2008
.
Hyperhomocysteinemia in inflammatory bowel disease patients without past intestinal resections: correlations with cobalamin, pyridoxine, folate concentrations, acute phase reactants, disease activity, and prior thromboembolic complications.
J. Clin. Gastroenterol.
42
:
481
486
.
50
Yakut
M.
,
Ustün
Y.
,
Kabaçam
G.
,
Soykan
I.
.
2010
.
Serum vitamin B12 and folate status in patients with inflammatory bowel diseases.
Eur. J. Intern. Med.
21
:
320
323
.
51
Zezos
P.
,
Papaioannou
G.
,
Nikolaidis
N.
,
Vasiliadis
T.
,
Giouleme
O.
,
Evgenidis
N.
.
2005
.
Hyperhomocysteinemia in ulcerative colitis is related to folate levels.
World J. Gastroenterol.
11
:
6038
6042
.
52
Chen
M.
,
Peyrin-Biroulet
L.
,
George
A.
,
Coste
F.
,
Bressenot
A.
,
Bossenmeyer-Pourie
C.
,
Alberto
J. M.
,
Xia
B.
,
Namour
B.
,
Gueant
J. L.
.
Methyl-deficient diet aggravates experimental colitis in rats.
J. Cell. Mol. Med.
In press.
53
Taylor
C. T.
,
Colgan
S. P.
.
2007
.
Hypoxia and gastrointestinal disease.
J. Mol. Med.
85
:
1295
1300
.
54
Semenza
G. L.
2009
.
Regulation of oxygen homeostasis by hypoxia-inducible factor 1.
Physiology
24
:
97
106
.
55
Tormos
K. V.
,
Chandel
N. S.
.
2010
.
Inter-connection between mitochondria and HIFs.
J. Cell. Mol. Med.
14
:
795
804
.
56
Morgan
H. D.
,
Santos
F.
,
Green
K.
,
Dean
W.
,
Reik
W.
.
2005
.
Epigenetic reprogramming in mammals.
Hum. Mol. Genet.
14
(
Spec No 1
):
R47
R58
.
57
Trojer
P.
,
Reinberg
D.
.
2006
.
Histone lysine demethylases and their impact on epigenetics.
Cell
125
:
213
217
.
58
Halim
A. B.
,
LeGros
L.
,
Chamberlin
M. E.
,
Geller
A.
,
Kotb
M.
.
2001
.
Regulation of the human MAT2A gene encoding the catalytic α2 subunit of methionine adenosyltransferase, MAT II: gene organization, promoter characterization, and identification of a site in the proximal promoter that is essential for its activity.
J. Biol. Chem.
276
:
9784
9791
.
59
Yang
H.
,
Sadda
M. R.
,
Yu
V.
,
Zeng
Y.
,
Lee
T. D.
,
Ou
X.
,
Chen
L.
,
Lu
S. C.
.
2003
.
Induction of human methionine adenosyltransferase 2A expression by tumor necrosis factor α: role of NF-κB and AP-1.
J. Biol. Chem.
278
:
50887
50896
.
60
Gough
D. J.
,
Levy
D. E.
,
Johnstone
R. W.
,
Clarke
C. J.
.
2008
.
IFNγ signaling-does it mean JAK-STAT?
Cytokine Growth Factor Rev.
19
:
383
394
.
61
Strober
W.
,
Fuss
I. J.
,
Blumberg
R. S.
.
2002
.
The immunology of mucosal models of inflammation.
Annu. Rev. Immunol.
20
:
495
549
.
62
Taylor
C. T.
,
Cummins
E. P.
.
2009
.
The role of NF-κB in hypoxia-induced gene expression.
Ann. N. Y. Acad. Sci.
1177
:
178
184
.
63
Hirata
I.
,
Yasumoto
S.
,
Toshina
K.
,
Inoue
T.
,
Nishikawa
T.
,
Murano
N.
,
Murano
M.
,
Wang
F. Y.
,
Katsu
K.
.
2007
.
Evaluation of the effect of pyrrolidine dithiocarbamate in suppressing inflammation in mice with dextran sodium sulfate-induced colitis.
World J. Gastroenterol.
13
:
1666
1671
.
64
Schmidt
N.
,
Gonzalez
E.
,
Visekruna
A.
,
Kühl
A. A.
,
Loddenkemper
C.
,
Mollenkopf
H.
,
Kaufmann
S. H.
,
Steinhoff
U.
,
Joeris
T.
.
2010
.
Targeting the proteasome: partial inhibition of the proteasome by bortezomib or deletion of the immunosubunit LMP7 attenuates experimental colitis.
Gut
59
:
896
906
.
65
Zhang
D. K.
,
Cheng
L. N.
,
Huang
X. L.
,
Shi
W.
,
Xiang
J. Y.
,
Gan
H. T.
.
2009
.
Tetrandrine ameliorates dextran-sulfate-sodium-induced colitis in mice through inhibition of nuclear factor-κB activation.
Int. J. Colorectal Dis.
24
:
5
12
.

The authors have no financial conflicts of interest.