Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity

K A Kuhn, H M Schulz, E H Regner, E L Severs, J D Hendrickson, G Mehta, A K Whitney, D Ir, N Ohri, C E Robertson, D N Frank, E L Campbell, S P Colgan, K A Kuhn, H M Schulz, E H Regner, E L Severs, J D Hendrickson, G Mehta, A K Whitney, D Ir, N Ohri, C E Robertson, D N Frank, E L Campbell, S P Colgan

Abstract

Interactions between the microbiota and distal gut are important for the maintenance of a healthy intestinal barrier; dysbiosis of intestinal microbial communities has emerged as a likely contributor to diseases that arise at the level of the mucosa. Intraepithelial lymphocytes (IELs) are positioned within the epithelial barrier, and in the small intestine they function to maintain epithelial homeostasis. We hypothesized that colon IELs promote epithelial barrier function through the expression of cytokines in response to interactions with commensal bacteria. Profiling of bacterial 16S ribosomal RNA revealed that candidate bacteria in the order Bacteroidales are sufficient to promote IEL presence in the colon that in turn produce interleukin-6 (IL-6) in a MyD88 (myeloid differentiation primary response 88)-dependent manner. IEL-derived IL-6 is functionally important in the maintenance of the epithelial barrier as IL-6-/- mice were noted to have increased paracellular permeability, decreased claudin-1 expression, and a thinner mucus gel layer, all of which were reversed by transfer of IL-6+/+ IELs, leading to protection of mice in response to Citrobacter rodentium infection. Therefore, we conclude that microbiota provide a homeostatic role for epithelial barrier function through regulation of IEL-derived IL-6.

Conflict of interest statement

Disclosure

The authors declare no commercial or financial conflict of interest.

Figures

Figure 1. Bacteria in the class Bacteroides…
Figure 1. Bacteria in the class Bacteroides maintain the colonic IEL population
(a–d) Groups of 3–9 C57Bl/6 male and female mice aged 8–12 weeks were untreated, treated with antibiotics for one week, or treated with antibiotics followed by recolonization for one week each. Data are from two independent experiments. (a) Immunofluorescence of methacarn-fixed, paraffin embedded colon tissue from mice was performed and representative images shown at 40X. Bar=20 µm. Dotted lines outline crypts and arrows point to IELs as identified as CD3+ (green) cells in the epithelial layer (β-catenin, red). Nuclei were stained with bis-benzimide (blue). Dashed lines outline crypts while arrows indicate IELs. (b) CD3+ epithelial cells were counted in four well-oriented high-powered fields (HPF) from immunofluorescence staining performed in 6 untreated, 7 antibiotic-treated, 6 recolonized, and 3 germ free mice and shown as the mean number of cells per HPF ± SEM. **, P<0.01 as determined by Kruskall-Wallis analysis with Dunn’s post-test. (c,d) The absolute number of epithelial CD3+ cells harvested from the colons of mice was determined by flow cytometry. Each dot represents an individual mouse and bars are the mean ± SEM. Statistical analysis for (c) was performed using a two-tailed Student’s t-test; **, P<0.01. In (d) statistical analysis was performed using a one-way ANOVA with Dunnett’s multiple comparisons test; ***, P<0.001 and ****, P<0.0001. (e) 16S rRNA sequencing from fecal DNA extracted from 5 untreated and 5 antibiotic-treated mice was performed. Order level differences in relative abundances ± SEM are shown with Wilcoxon rank test performed for statistical analysis. *, P<0.05; **, P<0.01 (f) Germ-free male and female mice aged 8–12 weeks were gavaged with PBS, Alistipes onderdonkii, Bacteroides fragilis, or Bacteroides thetaiotamicron and allowed to colonize for two weeks. Epithelial cells were harvested and CD3+ cells evaluated by flow cytometry. Each dot represents an individual mouse and bars are the mean ± SEM. ***, P<0.001 as determined by one-way ANOVA with Dunnett’s post-test.
Figure 2. IELs utilize bacterial signals for…
Figure 2. IELs utilize bacterial signals for stimulation of IL-6 secretion
IELs were harvested from colons of mice and mitogen-stimulated ex vivo. IL-6 secretion into the supernatant was measured by ELISA, and IL-6 from unstimulated IELs was subtracted from that of the mitogen-stimulated IELs from the same mouse. Data are from groups of 3–9 male and female mice aged 8–12 weeks in two independent experiments and shown as the mean ± SEM. Statistical significance was determined by one-way ANOVA with Dunnett’s test. (a) IL-6 secretion from IELs and LPLs isolated from untreated C57Bl/6 mice as well as antibiotic-treated, recolonized, and MyD88−/− mice. *, P<0.05; ****, P<0.0001. ND = not detected; NT = not tested. (b) IEL secretion of IL-6 from germ-free mice gavaged with PBS or monocolonized with bacteria. *,P<0.05
Figure 3. IL-6 signals in colon epithelia…
Figure 3. IL-6 signals in colon epithelia and enhances epithelial barrier function via induction of claudin-1 and mucin-2
(a) IL-6Rα protein in T84 and primary murine epithelial cells was determined by Western blot. (b) T84 colonic epithelial cells were cultured to confluence in the absence or presence of 50 ng/ml recombinant human IL-6. Protein was harvested after 0, 10, and 30 minutes of IL-6 exposure. Western blot confirmed phosphorylation of STAT3 after IL-6 exposure. (c) After 24 hours of IL-6 exposure, RNA from T84 cells was harvested and evaluated by qPCR for SOCS3 expression and normalized to actin. Data are the mean ± SEM fold induction of SOCS3 in IL-6 treated cells compared to untreated cells. An unpaired two-tailed Student’s t-test was used to determine statistical significance. *, PP<0.01 (e) T84 transwells were evaluated for paracellular flux of FITC-dextran. The rate of flux is shown as the mean ± SEM. An unpaired two-tailed Student’s t-test demonstrated significance. ***, P<0.0001 (f) After 24 hours of IL-6 exposure, RNA from T84 cells was extracted evaluated for CLDN1 expression by qPCR. Data are the mean expression of CLDN1 ± SEM in IL-6 treated cells relative to untreated cells. Statistical analysis using an unpaired two-tailed Student’s t-test revealed significance. *P<0.05 (g) Cellular lysates from unexposed and IL-6 exposed T84 cells at 24 hours were evaluated for claudin-1 protein by Western blot. (h) RNA from T84 cells with and without IL-6 treatment was evaluated for MUC2 expression by qPCR. Data are the mean fold induction of MUC2 ± SEM in treated cells compared to untreated cells. Statistical analysis using an unpaired two-tailed Student’s t-test revealed significance. *P<0.05
Figure 4. Epithelial barrier integrity is impaired…
Figure 4. Epithelial barrier integrity is impaired in the absence of IL-6
8–12 week old male and female C57Bl/6 and IL-6−/− mice were treated as previously described. Experiments were performed using littermates in groups of 3 mice and repeated twice. (a) Untreated, antibiotic-treated (Abx), recolonized, and IL-6−/− mice were gavaged with 0.6 mg/kg body weight 4 kDa FITC-dextran. After four hours, sera were collected from the mice, the fluorescence at 492 nm measured, and the amount of dextran calculated. Data are the mean concentration of dextran ± SEM. Statistical analyses were performed by one-way ANOVA with Dunnet’s test. ***, P<0.001 (b) Claudin-1 (brown) in vivo was evaluated by immunohistochemistry. Representative photos from 1 of 5 IL-6+/+ and 1 of 4 IL-6−/− mice are shown at 400X. Bar=20 µm. (c) Fluorescent in situ hybridization using a universal bacterial probe (red) was performed on IL-6+/+ and IL-6−/− mice. Nuclei were labeled with bis-benzimide (blue) and the mucus layer labeled with wheat germ agglutinin (green). Representative images are shown at 400X. Bar=20 µm. (d) Measurement of the mucus layer was performed in 3 areas of each of 4–6 high-powered fields (400X) per mouse (6 IL-6+/+ and 6 IL-6−/− mice). Data are the mean ± SEM mucus thickness. An unpaired two-tailed Student’s t-test demonstrated significance. ***, P<0.001
Figure 5. IEL produced IL-6 repairs the…
Figure 5. IEL produced IL-6 repairs the epithelial barrier and protects from C. rodentium colitis
(a) IELs from IL-6+/+ or IL-6−/− mice were harvested, magnetically sorted, and transferred into recipient IL-6+/+ or IL-6−/− mice. After one week, intestinal barrier permeability was evaluated by FITC-dextran flux. Two experiments of 2–3 male and female mice per group were performed. Data are the mean serum concentration of dextran ± SEM. ****, P<0.0001 by one-way ANOVA with Tukey’s test. (b) Two days following IEL transfer, 4–7 mice per group were infected with C. rodentium by oral gavage and monitored by daily weights. Mice were euthanized 12 days after infection. Data are the mean percentage of starting weight ± SEM. A two-way repeated-measures ANOVA with Dunnett’s test determined statistical significance. *, P<0.05; **,P<0.01 (c) Methacarn-fixed, paraffin embedded colon tissue from 12-day infected mice in (b) were stained by H&E and evaluated in a blinded fashion for histologic damage as assessed by the number of organized inflammatory aggregates and ulcers along the entire colon. These are shown as the mean ± SEM for each treatment group. *, P<0.05 by Kruskall-Wallis test with Dunn’s multiple comparisons test. (d) Five well-oriented high-powered fields (HPF) per mouse were viewed at 200X and number of crypts counted in each section. Data are the mean crypts/HPF ± SEM. **, P<0.01 by one-way ANOVA with Tukey’s test. (e) Representative histology is shown at 200X. Bar = 50 µm.
Figure 6. Transfer of IL-6 + IELs…
Figure 6. Transfer of IL-6+ IELs into IL-6 −/− mice restores the mucus layer and claudin-1 expression
(a) FISH was performed on tissues from Figure 5 and the number of bacteria located within intestinal tissue in each of 20 HPF (at 400X) were counted and shown at the mean bacteria/HPF ± SEM. *, P<0.05; ***,P<0.001 by Kruskall-Wallis test with Dunn’s multiple comparisons test. (b) Mucus thickness in 3 areas of each of 4 HPF per mouse was measured in sections from (a) and displayed as the mean mucus thickness ± SEM. Statistical significance was determined by one-way ANOVA with Tukey’s post-test. ***,P<0.001; ****P<0.0001 (c) Representative FISH is shown at 400X. Bar=20 µm. A universal bacterial probe (red) was used to mark bacteria; nuclei were labeled with bis-benzimide (blue); and the mucus layer labeled with wheat germ agglutinin (green). Dashed white lines outline the epithelial and luminal borders of the inner mucus layer. Arrowheads point to areas of bacterial translocation. (d) Claudin-1 protein expression from the experiment in Figure 5 was evaluated by immunohistochemistry and assessed a numeric score for each mouse based on the level of staining: 0=no staining, 1=faint, 2=mild; 3=moderate; 4=intense. The mean staining intensity per group ± SEM is shown and statistical significance was assessed using a Kruskall-Wallis test with Dunn’s multiple comparisons test. *,P<0.05 (e) Representative claudin-1 immunohistochemistry (brown) is shown at 400X. Bar=20 µm.

References

    1. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–230.
    1. Hooper LV, Macpherson AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nature reviews. Immunology. 2010;10:159–169.
    1. Cheroutre H, Lambolez F, Mucida D. The light and dark sides of intestinal intraepithelial lymphocytes. Nat Rev Immunol. 2011;11:445–456.
    1. Higgins JM, et al. Direct and regulated interaction of integrin alphaEbeta7 with E-cadherin. J Cell Biol. 1998;140:197–210.
    1. Yu Q, et al. MyD88-dependent signaling for IL-15 production plays an important role in maintenance of CD8 alpha TCR alpha beta and TCR gamma delta intestinal intraepithelial lymphocytes. Journal of immunology. 2006;176:6180–6185.
    1. Jiang W, et al. Recognition of gut microbiota by NOD2 is essential for the homeostasis of intestinal intraepithelial lymphocytes. The Journal of experimental medicine. 2013;210:2465–2476.
    1. Ismail AS, et al. Gammadelta intraepithelial lymphocytes are essential mediators of host-microbial homeostasis at the intestinal mucosal surface. Proc Natl Acad Sci U S A. 2011;108:8743–8748.
    1. Kuhn KA, Manieri NA, Liu TC, Stappenbeck TS. IL-6 stimulates intestinal epithelial proliferation and repair after injury. PloS one. 2014;9:e114195.
    1. Rose-John S, Scheller J, Elson G, Jones SA. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. Journal of leukocyte biology. 2006;80:227–236.
    1. Tebbutt NC, et al. Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nature medicine. 2002;8:1089–1097.
    1. Bollrath J, et al. gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer cell. 2009;15:91–102.
    1. Dann SM, et al. IL-6-dependent mucosal protection prevents establishment of a microbial niche for attaching/effacing lesion-forming enteric bacterial pathogens. J Immunol. 2008;180:6816–6826.
    1. Grivennikov S, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15:103–113.
    1. Helgeland L, Vaage JT, Rolstad B, Midtvedt T, Brandtzaeg P. Microbial colonization influences composition and T-cell receptor V beta repertoire of intraepithelial lymphocytes in rat intestine. Immunology. 1996;89:494–501.
    1. Umesaki Y, Setoyama H, Matsumoto S, Okada Y. Expansion of alpha beta T-cell receptor-bearing intestinal intraepithelial lymphocytes after microbial colonization in germ-free mice and its independence from thymus. Immunology. 1993;79:32–37.
    1. Song Y, et al. Alistipes onderdonkii sp nov, Alistipes shahii sp nov., of human origin. Int J Syst Evol Microbiol. 2006;56:1985–1990.
    1. Rautio M, et al. Reclassification of Bacteroides putredinis (Weinberg , 1937) in a new genus Alistipes gen nov, as Alistipes putredinis comb nov, description of Alistipes finegoldii sp nov., from human sources. Syst Appl Microbiol. 2003;26:182–188.
    1. Madara JL, Stafford J, Dharmsathaphorn K, Carlson S. Structural analysis of a human intestinal epithelial cell line. Gastroenterology. 1987;92:1133–1145.
    1. Saeedi BJ, et al. HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol Biol Cell. 2015;26:2252–2262.
    1. Inai T, Kobayashi J, Shibata Y. Claudin-1 contributes to the epithelial barrier function in MDCK cells. Eur J Cell Biol. 1999;78:849–855.
    1. Johansson ME, et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:15064–15069.
    1. Hasnain SZ, et al. IL-10 promotes production of intestinal mucus by suppressing protein misfolding and endoplasmic reticulum stress in goblet cells. Gastroenterology. 2013;144:368–357. e359.
    1. Sugimoto K, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. The Journal of clinical investigation. 2008;118:534–544.
    1. Smith PM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–573.
    1. Ishizuka S, Tanaka S. Modulation of CD8+ intraepithelial lymphocyte distribution by dietary fiber in the rat large intestine. Experimental biology and medicine. 2002;227:1017–1021.
    1. Al-Sadi R, et al. Interleukin-6 modulation of intestinal epithelial tight junction permeability is mediated by JNK pathway activation of claudin-2 gene. PloS one. 2014;9:e85345.
    1. Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. The Journal of biological chemistry. 2011;286:31263–31271.
    1. Madara JL, Stafford J. Interferon-gamma directly affects barrier function of cultured intestinal epithelial monolayers. The Journal of clinical investigation. 1989;83:724–727.
    1. Kominsky DJ, et al. IFN-gamma-mediated induction of an apical IL-10 receptor on polarized intestinal epithelia. J Immunol. 2014;192:1267–1276.
    1. Birchenough GM, Johansson ME, Gustafsson JK, Bergstrom JH, Hansson GC. New developments in goblet cell mucus secretion and function. Mucosal immunology. 2015;8:712–719.
    1. Enss ML, et al. Proinflammatory cytokines trigger MUC gene expression and mucin release in the intestinal cancer cell line LS180. Inflamm Res. 2000;49:162–169.
    1. Guttman JA, Samji FN, Li Y, Vogl AW, Finlay BB. Evidence that tight junctions are disrupted due to intimate bacterial contact and not inflammation during attaching and effacing pathogen infection in vivo. Infect Immun. 2006;74:6075–6084.
    1. Koroleva EP, et al. Citrobacter rodentium-induced colitis: A robust model to study mucosal immune responses in the gut. J Immunol Methods. 2015;421:61–72.
    1. Zheng Y, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nature medicine. 2008;14:282–289.
    1. Ishigame H, et al. Differential roles of interleukin-17A and −17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 2009;30:108–119.
    1. Song X, et al. IL-17RE is the functional receptor for IL-17C and mediates mucosal immunity to infection with intestinal pathogens. Nature immunology. 2011;12:1151–1158.
    1. Bhinder G, et al. The Citrobacter rodentium mouse model: studying pathogen and host contributions to infectious colitis. Journal of visualized experiments : JoVE. 2013:e50222.
    1. Son JS, et al. Altered Interactions between the Gut Microbiome and Colonic Mucosa Precede Polyposis in APCMin/+ Mice. PloS one. 2015;10:pe0127985.
    1. Frank DN, et al. Perilipin-2 Modulates Lipid Absorption and Microbiome Responses in the Mouse Intestine. PloS one. 2015;10:e0131944.
    1. Alkanani AK, et al. Induction of diabetes in the RIP-B7.1 mouse model is critically dependent on TLR3 and MyD88 pathways and is associated with alterations in the intestinal microbiome. Diabetes. 2014;63:619–631.
    1. Pruesse E, Peplies J, Glockner FO. SINA: accurate high-throughput multiple sequence alignment of ribosomal RNA genes. Bioinformatics. 2012;28:1823–1829.
    1. Pruesse E, et al. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic acids research. 2007;35:7188–7196.
    1. Robertson CE, et al. Explicet: graphical user interface software for metadata-driven management, analysis and visualization of microbiome data. Bioinformatics. 2013;29:3100–3101.
    1. Amann RI, et al. Combination of 16S rRNA-targeted oligonucleotide probes with flow cytometry for analyzing mixed microbial populations. Applied and environmental microbiology. 1990;56:1919–1925.
    1. Malo MS, et al. Intestinal alkaline phosphatase preserves the normal homeostasis of gut microbiota. Gut. 2010;59:1476–1484.
    1. Lee SM, et al. Bacterial colonization factors control specificity and stability of the gut microbiota. Nature. 2013;501:426–429.
    1. Saeedi BJ, et al. HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol Biol Cell. 2015;26:2252–2262.

Source: PubMed

3
Abonnere