NOD2 regulates CXCR3-dependent CD8+ T cell accumulation in intestinal tissues with acute injury

Xingxin Wu, Amit Lahiri, G Kenneth Haines 3rd, Richard A Flavell, Clara Abraham, Xingxin Wu, Amit Lahiri, G Kenneth Haines 3rd, Richard A Flavell, Clara Abraham

Abstract

Polymorphisms in NOD2 confer risk for Crohn's disease, characterized by intestinal inflammation. How NOD2 regulates both inflammatory and regulatory intestinal T cells, which are critical to intestinal immune homeostasis, is not well understood. Anti-CD3 mAb administration is used as therapy in human autoimmune diseases, as well as a model of transient intestinal injury. The stages of T cell activation, intestinal injury, and subsequent T tolerance are dependent on migration of T cells into the small intestinal (SI) lamina propria. Upon anti-CD3 mAb treatment of mice, we found that NOD2 was required for optimal small intestinal IL-10 production, in particular from CD8(+) T cells. This requirement was associated with a critical role for NOD2 in SI CD8(+) T cell accumulation and induction of the CXCR3 ligands CXCL9 and CXCL10, which regulate T cell migration. NOD2 was required in both the hematopoietic and nonhematopoietic compartments for optimal expression of CXCR3 ligands in intestinal tissues. NOD2 synergized with IFN-γ to induce CXCL9 and CXCL10 secretion in dendritic cells, macrophages, and intestinal stromal cells in vitro. Consistent with the in vitro studies, during anti-CD3 mAb treatment in vivo, CXCR3 blockade, CD8(+) T cell depletion, or IFN-γ neutralization each inhibited SI CD8(+) T cell recruitment, and reduced chemokine expression and IL-10 expression. Thus, NOD2 synergizes with IFN-γ to promote CXCL9 and CXCL10 expression, thereby amplifying CXCR3-dependent SI CD8(+) T cell migration during T cell activation, which, in turn, contributes to induction of both inflammatory and regulatory T cell outcomes in the intestinal environment.

Figures

Figure 1. NOD2 is required for optimal…
Figure 1. NOD2 is required for optimal accumulation of IL-10-producing cells in the SI of mice during anti-CD3 mAb treatment
NOD2+/− IL-10-GFP and NOD2−/− IL-10-GFP mice were treated with 15 μg of anti-CD3 mAb or IgG isotype control at 0 and 48 h. Four hours after the last injection animals were sacrificed. (A) Representative flow cytometry plots of IL-10–producing cells in the SI LP (gated on live cells). (B) Percentage of IL-10-producing cells in the SI. (C) Number of IL-10-producing cells in the SI (mean+SEM; n=3 per group; representative of 5 independent experiments). (D) IL-10 mRNA expression in the SI. (E) Serum IL-10. D–E are shown as mean+SEM; n=6 per group from two independent experiments. Comparisons are between anti-CD3 mAb and IgG treatment in the same genotype or as indicated. *, p<0.05; **, p<0.01; ***, p<0.001.
Figure 2. CD8 + T cells are…
Figure 2. CD8+ T cells are the major source of IL-10-producing cells in the SI of mice during anti-CD3 mAb treatment
Mice were treated with anti-CD3 mAb as in Fig 1. (A) Representative flow cytometry gated on IL-10–producing cells in the SI LP. Surface CD3 is internalized with anti-CD3 mAb treatment, whereas Thy1.2 expression remains intact. Cells were stained for Thy1.2, CD4 and CD8 expression. (B) Pie chart of the proportion of IL-10-producing cell types in the SI after anti-CD3 mAb injection. (mean+SEM; n=3). (C) Percentage and (D) number of IL-10-producing CD8+ T cells (CD4−Thy1.2+) in the SI of NOD2+/− and NOD2−/− mice (mean+SEM; n=3 per group). Data are representative of at least 5 independent experiments. **, p<0.01.
Figure 3. NOD2 is required for CD8…
Figure 3. NOD2 is required for CD8+ T cell migration to and cytokine production in the SI upon anti-CD3 mAb treatment
Mice were treated with anti-CD3 mAb as in Fig 1. (A) Representative flow cytometry plots of cells from the SI gated on CD8+ T cells (CD4−Thy1.2+). (B) Percentage of IL-10-GFP+ cells in CD8+ T cells in the SI of NOD2+/− and NOD2−/− mice after anti-CD3 mAb treatment. (C) Percentage and (D) number of CD8+ T cells in the SI lamina propria. (mean+SEM; n=3 per group; representative of 5 independent experiments). (E) IFN-γ and IL-17A mRNA expression in the SI (mean+SEM; n=6–9 per group from 3 independent experiments). Comparisons are between anti-CD3 mAb and IgG treatment in the same genotype or as indicated. (F) Oral FITC-dextran was administered 4h before sacrifice (coincident with the 48h anti-CD3 mAb treatment time point) and measured in the serum at the time of sacrifice. (G–I) NOD2+/− IL-10-GFP mice were administered an antibiotic (ABX) regimen consisting of vancomycin, metronidazole, ampicillin, and neomycin for 4 weeks in the drinking water. Antibiotic-treated or –untreated (SPF, specific pathogen free) mice were then treated with anti-CD3 mAb as in Fig 1 and examined for: (G) percentage of IL-10-GFP+ cells (gated on live cells), (H) percentage of IL-10+CD8+ T cells, and (I) percentage of CD8+ T cells in the SI lamina propria. *, p<0.05; **, p<0.01; ***, p<0.001; NS, not significant.
Figure 4. NOD2 is required for optimal…
Figure 4. NOD2 is required for optimal CXCR3 ligand induction in the SI upon anti-CD3 mAb treatment
(A–D) NOD2+/− and NOD2−/− mice were treated with anti-CD3 mAb as in Fig 1. CXCL9 and CXCL10 mRNA levels in (A) SI and (B) MLN (mean+SEM; n=6 per group; representative of two independent experiments). (C–D) Percentage of CD8+ T cells expressing CXCR3 in SI, MLN and spleen was assessed. (C) Representative flow cytometry plots for CXCR3 expression on CD8+ T cells (solid black line). Low CXCR3-expressing MHCII+ cells are shown for comparison (shaded grey histogram). (D) Summary graph for CXCR3-expressing cells within CD8+ T cells (mean+SEM; n=3 per group; representative of two independent experiments). *, p<0.05; **, p<0.01; ***, p<0.001.
Figure 5. CXCR3 is essential for the…
Figure 5. CXCR3 is essential for the amplification of SI CD8+ T cell accumulation, chemokines and subsequent IL-10 induction upon anti-CD3 mAb treatment
To block CXCR3, mice were treated 100 μg anti-CXCR3 mAb (or Armenian hamster IgG isotype control) i.p. 2h before each anti-CD3 mAb injection (at 0 and 48h). Mice were harvested 4h after the 2nd anti-CD3 mAb injection. (A) Percentage and (B) number of CD8+ T cells in the SI LP (mean+SEM; n=3 per group; representative of 3 independent experiments). (C) CXCL9 and CXCL10 mRNA expression in the SI. (D) IL-10, IFN-γ, and IL-17A mRNA expression in SI. (E) Serum levels of IL-10, IFN-γ, and IL-17A (mean + SEM; n = 6/group from 2 independent experiments for C–E). *, p<0.05; **, p<0.01; ***, p<0.001.
Figure 6. NOD2 in hematopoietic and non-hematopoietic…
Figure 6. NOD2 in hematopoietic and non-hematopoietic cells is required for optimal CD8+ T cell migration into the SI lamina propria upon anti-CD3 mAb treatment
Chimeric mice were generated and treated with anti-CD3 mAb as in Fig 1. (A–D) Transfer of NOD2+/− or NOD2−/− Thy1.1+ BM cells into irradiated Thy1.2+ WT mice. (E–H) Transfer of WT Thy1.2+ BM cells into NOD2+/− or NOD2−/− Thy1.1+ mice. (A&E) Percentage of CD8+ T cells donor cells in the SI LP. (B&F) Chemokine mRNA expression in SI. (C&G) Cytokine mRNA expression in SI. (D&H) Serum cytokine levels. Data are mean+SEM; n=3 per group; representative of 2 independent experiments. *, p<0.05; **, p<0.01; ***, p<0.001.
Figure 7. NOD2 stimulation synergistically enhances IFN-γ-induced…
Figure 7. NOD2 stimulation synergistically enhances IFN-γ-induced CXCL9 and CXCL10 expression in BMM, BMDC and intestinal stromal cells in vitro
(A) BMM, (B) BMDC or (C) intestinal stromal cells from NOD2+/− or NOD2−/− mice were stimulated for 24h with MDP (10 μg/ml for BMM & BMDC cells; 1μg/ml intestinal stromal cells), 1 ng/ml IFN-γ, or 0.1 μg/ml lipid A, alone or combination as indicated. Supernatants were assayed for CXCL9 and CXCL10. Data shown as mean+SEM, n=3 per group, and representative of 3 independent experiments. Significance is compared for conditions examining synergy as indicated (Bonferroni correction applied) and between NOD2+/− and NOD2−/− cells. **, p<0.01; ***, p<0.001.
Figure 8. Depletion of CD8 + T…
Figure 8. Depletion of CD8+ T cells or neutralization of IFN-γ attenuates intestinal outcomes upon anti-CD3 mAb treatment
(A–D) CD8+ T cells were depleted by anti-CD8 mAb injection (or treated with rat IgG2b isotype control) as per Materials & Methods. (EH) IFN-γ was neutralized by anti-IFN-γ mAb injection (or treated with rat IgG1 isotype control) as per Materials & Methods. (A&E) Percentage of CD8+ T cells in the SI LP (mean+SEM; n=3 per group; representative of 2 independent experiments). (B&F) CXCL9 and CXCL10 mRNA expression in the SI. (C&G) CXCL9 and CXCL10 protein expression per mg SI tissue. (D&H) IL-10, IFN-γ, and IL-17A mRNA expression in the SI (mean + SEM; n = 6/group from 2 independent experiments for B-D & F-H). *, p<0.05; **, p<0.01; ***, p<0.001.
Figure 9. Proposed model for mechanisms of…
Figure 9. Proposed model for mechanisms of NOD2-mediated CXCR3-dependent T cell migration into intestinal tissues
1. Acute activation of circulating and intestinal CD8+ T cells by anti-CD3 mAb; 2. IFN-γ secretion by activated T cells; 3. IFN-γ (T cell-derived) and MDP (microbiota-derived; stimulates intracellular NOD2) synergize to increase CXCL9 and CXCL10 secretion from myeloid-derived and intestinal stromal cells; 4. CXCR3-mediated chemotaxis of CD8+ T cells into the intestinal lamina propria secondary to increased CXCL9 and CXCL10 expression; 5. Newly recruited CD8+ T cells amplify recruitment of additional CD8+ T cell into intestinal tissues through the above sequence of events.

References

    1. Abraham C, Cho JH. Inflammatory bowel disease. N Eng J Med. 2009;361:2066–2078.
    1. Shaw MH, Reimer T, Sanchez-Valdepenas C, Warner N, Kim YG, Fresno M, Nunez G. T cell-intrinsic role of Nod2 in promoting type 1 immunity to Toxoplasma gondii. Nat Immunol. 2009;10:1267–U1266.
    1. Magalhaes JG, Fritz JH, Le Bourhis L, Sellge G, Travassos LH, Selvanantham T, Girardin SE, Gommerman JL, Philpott DJ. Nod2-dependent Th2 polarization of antigen-specific immunity. J Immunol. 2008;181:7925–7935.
    1. van Beelen AJ, Zelinkova Z, Taanman-Kueter EW, Muller FJ, Hommes DW, Zaat SA, Kapsenberg ML, de Jong EC. Stimulation of the intracellular bacterial sensor NOD2 programs dendritic cells to promote interleukin-17 production in human memory T cells. Immunity. 2007;27:660–669.
    1. Han X, Uchida K, Jurickova I, Koch D, Willson T, Samson C, Bonkowski E, Trauernicht A, Kim MO, Tomer G, Dubinsky M, Plevy S, Kugathsan S, Trapnell BC, Denson LA. Granulocyte-macrophage colony-stimulating factor autoantibodies in murine ileitis and progressive ileal Crohn’s disease. Gastroenterology. 2009;136:1261–1271. e1261–1263.
    1. Kim YG, Shaw MH, Warner N, Park JH, Chen F, Ogura Y, Nunez G. Cutting Edge: Crohn’s Disease-Associated Nod2 Mutation Limits Production of Proinflammatory Cytokines To Protect the Host from Enterococcus faecalis-Induced Lethality. J Immunol. 2011;187:2849–2852.
    1. Zeller T, Wild P, Szymczak S, Rotival M, Schillert A, Castagne R, Maouche S, Germain M, Lackner K, Rossmann H, Eleftheriadis M, Sinning CR, Schnabel RB, Lubos E, Mennerich D, Rust W, Perret C, Proust C, Nicaud V, Loscalzo J, Hubner N, Tregouet D, Munzel T, Ziegler A, Tiret L, Blankenberg S, Cambien F. Genetics and beyond--the transcriptome of human monocytes and disease susceptibility. PLoS One. 2010;5:e10693.
    1. Zhang FR, Huang W, Chen SM, Sun LD, Liu H, Li Y, Cui Y, Yan XX, Yang HT, Yang RD, Chu TS, Zhang C, Zhang L, Han JW, Yu GQ, Quan C, Yu YX, Zhang Z, Shi BQ, Zhang LH, Cheng H, Wang CY, Lin Y, Zheng HF, Fu XA, Zuo XB, Wang Q, Long H, Sun YP, Cheng YL, Tian HQ, Zhou FS, Liu HX, Lu WS, He SM, Du WL, Shen M, Jin QY, Wang Y, Low HQ, Erwin T, Yang NH, Li JY, Zhao X, Jiao YL, Mao LG, Yin G, Jiang ZX, Wang XD, Yu JP, Hu ZH, Gong CH, Liu YQ, Liu RY, Wang DM, Wei D, Liu JX, Cao WK, Cao HZ, Li YP, Yan WG, Wei SY, Wang KJ, Hibberd ML, Yang S, Zhang XJ, Liu JJ. Genomewide association study of leprosy. N Eng J Med. 2009;361:2609–2618.
    1. Chatenoud L, Bluestone JA. CD3-specific antibodies: a portal to the treatment of autoimmunity. Nat Rev Immunol. 2007;7:622–632.
    1. Herold KC, Burton JB, Francois F, Poumian-Ruiz E, Glandt M, Bluestone JA. Activation of human T cells by FcR nonbinding anti-CD3 mAb, hOKT3gamma1(Ala-Ala) J Clin Invest. 2003;111:409–418.
    1. Merger M, Viney JL, Borojevic R, Steele-Norwood D, Zhou P, Clark DA, Riddell R, Maric D, Podack ER, Coroitoru K. Defining the roles of perforin, Fas/FasL, and tumour necrosis factor alpha in T cell induced mucosal damage in the mouse intestine. Gut. 2002;51:155–163.
    1. Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M, Galan JE, Harhaj E, Flavell RA. Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 reporter knockin tiger mouse. Immunity. 2006;25:941–952.
    1. Waldron-Lynch F, Henegariu O, Deng S, Preston-Hurlburt P, Tooley J, Flavell R, Herold KC. Teplizumab induces human gut-tropic regulatory cells in humanized mice and patients. Sci Transl Med. 2012;4:118ra112.
    1. Esplugues E, Huber S, Gagliani N, Hauser AE, Town T, Wan YY, O’Connor W, Jr, Rongvaux A, Van Rooijen N, Haberman AM, Iwakura Y, Kuchroo VK, Kolls JK, Bluestone JA, Herold KC, Flavell RA. Control of TH17 cells occurs in the small intestine. Nature. 2011;475:514–518.
    1. Huber S, Gagliani N, Esplugues E, O’Connor W, Jr, Huber FJ, Chaudhry A, Kamanaka M, Kobayashi Y, Booth CJ, Rudensky AY, Roncarolo MG, Battaglia M, Flavell RA. Th17 cells express interleukin-10 receptor and are controlled by Foxp3(−) and Foxp3+ regulatory CD4+ T cells in an interleukin-10-dependent manner. Immunity. 2011;34:554–565.
    1. Blazquez AB, Knight AK, Getachew H, Bromberg JS, Lira SA, Mayer L, Berin MC. A Functional Role for CCR6 on Proallergic T Cells in the Gastrointestinal Tract. Gastroenterology. 2010;138:275–284.
    1. Zhang JL, Sun DJ, Hou CM, Wei YL, Li XY, Yu ZY, Feng JN, Shen BF, Li Y, Xiao H. CD3 mAb treatment ameliorated the severity of the cGVHD-induced lupus nephritis in mice by up-regulation of Foxp3(+) regulatory T cells in the target tissue: Kidney. Transpl Immunol. 2010;24:17–25.
    1. Waldron-Lynch F, Deng S, Preston-Hurlburt P, Henegariu O, Herold KC. Analysis of human biologics with a mouse skin transplant model in humanized mice. Am J Transplant. 2012;12:2652–2662.
    1. Daifotis AG, Koenig S, Chatenoud L, Herold KC. Anti-CD3 clinical trials in type 1 diabetes mellitus. Clin Immunol 2013
    1. Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso AS, Wu H, Chen ML, Gandhi R, Miller A, Maron R, Weiner HL. Oral CD3-specific antibody suppresses autoimmune encephalomyelitis by inducing CD4+ CD25- LAP+ T cells. Nat Med. 2006;12:627–635.
    1. Rahman MK, Midtling EH, Svingen PA, Xiong YN, Bell MP, Tung J, Smyrk T, Egan LJ, Faubion WA. The Pathogen Recognition Receptor NOD2 Regulates Human FOXP3(+) T Cell Survival. J Immunol. 2010;184:7247–7256.
    1. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–241.
    1. Marski M, Kandula S, Turner JR, Abraham C. CD18 is required for optimal development and function of CD4(+) CD25(+) T regulatory cells. J Immunol. 2005;175:7889–7897.
    1. Wang C, McDonough JS, McDonald KG, Huang C, Newberry RD. Alpha4beta7/MAdCAM-1 interactions play an essential role in transitioning cryptopatches into isolated lymphoid follicles and a nonessential role in cryptopatch formation. J Immunol. 2008;181:4052–4061.
    1. Bisikirska B, Colgan J, Luban J, Bluestone JA, Herold KC. TCR stimulation with modified anti-CD3 mAb expands CD8+ T cell population and induces CD8+CD25+ Tregs. J Clin Invest. 2005;115:2904–2913.
    1. Clayburgh DR, Musch MW, Leitges M, Fu YX, Turner JR. Coordinated epithelial NHE3 inhibition and barrier dysfunction are required for TNF-mediated diarrhea in vivo. J Clin Invest. 2006;116:2682–2694.
    1. Villablanca EJ, Cassani B, von Andrian UH, Mora JR. Blocking Lymphocyte Localization to the Gastrointestinal Mucosa as a Therapeutic Strategy for Inflammatory Bowel Diseases. Gastroenterology. 2011;140:1776–U1104.
    1. Groom JR, Luster AD. CXCR3 ligands: redundant, collaborative and antagonistic functions. Immunol Cell Biol. 2011;89:207–215.
    1. Abraham C, Cho JH. Functional consequences of NOD2 (CARD15) mutations. Inflamm Bowel Dis. 2006;12:641–650.
    1. Singh UP, Singh S, Taub DD, Lillard JW. Inhibition of IFN-gamma-inducible protein-10 abrogates colitis in IL-10(−/−) mice. J Immunol. 2003;171:1401–1406.
    1. Berg DJ, Zhang J, Weinstock JV, Ismail HF, Earle KA, Alila H, Pamukcu R, Moore S, Lynch RG. Rapid development of colitis in NSAID-treated IL-10-deficient mice. Gastroenterology. 2002;123:1527–1542.
    1. Hyun JG, Lee G, Brown JB, Grimm GR, Tang YM, Mittal N, Dirisina R, Zhang Z, Fryer JP, Weinstock JV, Luster AD, Barrett TA. Anti-interferon-inducible chemokine, CXCL10, reduces colitis by impairing T helper-1 induction and recruitment in mice. Inflamm Bowel Dis. 2005;11:799–805.
    1. Penack O, Smith OM, Cunningham-Bussel A, Liu X, Rao U, Yim N, Na IK, Holland AM, Ghosh A, Lu SX, Jenq RR, Liu C, Murphy GF, Brandl K, van den Brink MR. NOD2 regulates hematopoietic cell function during graft-versus-host disease. J Exp Med. 2009;206:2101–2110.
    1. Biswas A, Liu YJ, Hao L, Mizoguchi A, Salzman NH, Bevins CL, Kobayashi KS. Induction and rescue of Nod2-dependent Th1-driven granulomatous inflammation of the ileum. Proc Natl Acad Sci U S A. 2010;107:14739–14744.
    1. Jamontt J, Petit S, Clark N, Parkinson SJ, Smith P. Nucleotide-Binding Oligomerization Domain 2 Signaling Promotes Hyperresponsive Macrophages and Colitis in IL-10-Deficient Mice. J Immunol. 2013;190:2948–2958.
    1. Kim YG, Kamada N, Shaw MH, Warner N, Chen GY, Franchi L, Nunez G. The Nod2 Sensor Promotes Intestinal Pathogen Eradication via the Chemokine CCL2-Dependent Recruitment of Inflammatory Monocytes. Immunity. 2011;34:769–780.
    1. Magalhaes JG, Rubino SJ, Travassos LH, Le Bourhis L, Duan W, Sellge G, Geddes K, Reardon C, Lechmann M, Carneiro LA, Selvanantham T, Fritz JH, Taylor BC, Artis D, Mak TW, Comeau MR, Croft M, Girardin SE, Philpott DJ. Nucleotide oligomerization domain-containing proteins instruct T cell helper type 2 immunity through stromal activation. Proc Natl Acad Sci U S A. 2011;108:14896–14901.
    1. Sasaki S, Yoneyama H, Suzuki K, Suriki H, Aiba T, Watanabe S, Kawauchi Y, Kawachi H, Shimizu F, Matsushima K, Asakura H, Narumi S. Blockade of CXCL10 protects mice from acute colitis and enhances crypt cell survival. Eur J Immunol. 2002;32:3197–3205.
    1. Hardi R, Mayer L, Targan SR, Yellin M, Cardarelli PM, Das KM. A phase 1 open-label, single-dose, dose-escalation study of Mdx-1100, a high-affinity, neutralizing, fully human Igg1 kappa anti-CXCL10 (Ip10) monoclonal antibody, in ulcerative colitis. Gastroenterology. 2008;134:A99–A100.
    1. Masopust D, Vezys V, Marzo AL, Lefrancois L. Preferential localization of effector memory cells in nonlymphoid tissue. Science. 2001;291:2413–2417.

Source: PubMed

3
Iratkozz fel