Regulatory T cells control NK cells in an insulitic lesion by depriving them of IL-2

Jonathan Sitrin, Aaron Ring, K Christopher Garcia, Christophe Benoist, Diane Mathis, Jonathan Sitrin, Aaron Ring, K Christopher Garcia, Christophe Benoist, Diane Mathis

Abstract

Regulatory T (T reg) cells control progression to autoimmune diabetes in the BDC2.5/NOD mouse model by reining in natural killer (NK) cells that infiltrate the pancreatic islets, inhibiting both their proliferation and production of diabetogenic interferon-γ. In this study, we have explored the molecular mechanisms underlying this NK-T reg cell axis, following leads from a kinetic exploration of gene expression changes early after punctual perturbation of T reg cells in BDC2.5/NOD mice. Results from gene signature analyses, quantification of STAT5 phosphorylation levels, cytokine neutralization experiments, cytokine supplementation studies, and evaluations of intracellular cytokine levels collectively argue for a scenario in which T reg cells regulate NK cell functions by controlling the bioavailability of limiting amounts of IL-2 in the islets, generated mainly by infiltrating CD4(+) T cells. This scenario represents a previously unappreciated intertwining of the innate and adaptive immune systems: CD4(+) T cells priming NK cells to provoke a destructive T effector cell response. Our findings highlight the need to consider potential effects on NK cells when designing therapeutic strategies based on manipulation of IL-2 levels or targets.

Figures

Figure 1.
Figure 1.
Gene expression changes in pancreatic NK cells soon after T reg cell perturbation. NK cells were sorted from the pancreatic infiltrate 24 h after DT injection into BDC2.5/NOD mice with or without DTR expressed in T reg cells, and were profiled by microarray. (A) Transcript changes. Differential gene-expression (FC) values for DTR+ (n = 3) and DTR− mice (n = 6) (y axis) versus their two-class mean (x axis). Induced (>2-fold) genes are highlighted in red and repressed (>2-fold) genes are highlighted in blue. (B and C) Activation features. FC/FC plots of the same NK cell data as in A versus NK cells responding to different activation stimuli. Y axis, FC values for DTR+ versus DTR− mice; x axis, FC values for splenic NK cells cultured with (n = 3) or without (n = 2) IL-12 + IL-18 (see Materials and methods for details; B) or splenic NK cells from C57/BL6 mice 24 h after being infected with MCMV or not (n = 3; C). (D) Time course of transcript changes. Carried out as in A, except additional time points at 8 and 15 h were examined. FC/FC plots comparing gene expression differentials for pancreatic NK cells from DTR+ and DTR− BDC2.5/NOD mice (y axis) versus splenic NK cells from the same animals (x axis). Multiple replicates of cellular populations were collected (usually n = 3–6) and averaged. Highlighting in A–D represents the same set of genes.
Figure 2.
Figure 2.
The role of TGF-β signaling. (A, left) Representative flow cytometry profiles of IFN-γ expression in NOD-derived splenic NK cells cultured with (red) or without (gray) IL-12 + IL-18 in the absence of TGF-β, or with both cytokines and TGF-β (blue). (right) Summary data from three independent experiments. Mean ± SD. The p-value was calculated using the two-tailed unpaired Students’ t test. (B) Reciprocal transcript changes promoted by the loss of TGF-β and T reg cells. FC/FC plot comparing cytokine-activated, cultured, splenic NK cells ± TGF-β (x axis, n = 3) versus pancreatic NK cells from BDC2.5/NOD mice ± T reg ablation (y axis, same data as in Fig. 1 A). Dashed red line, linear regression. (C) FC/FC plot comparing transcriptional profiles of cytokine-activated, cultured, splenic NK cells ± TGF-β (y axis, as in Fig. 2 B) versus NK cells ± cytokine-activation (x axis, as in Fig. 1 B). Activation-independent TGF-β–induced genes were highlighted in pink and were superimposed (D in pink) on a volcano plot (p-value vs. fold change) of NK cell transcripts from BDC2.5/NOD mice depleted or not of T reg cells (same data as in Fig. 1). The number of signature genes up-regulated (right) or down-regulated (left) 24 h after T reg cell perturbation are indicated. P-value from the χ2 test. (E and F) Summary flow cytometry data for BDC2.5/NOD mice treated for 24 h with anti–TGF-β versus either an isotype-control mAb or PBS (combined). Percentage of NK cells of lymphocytes (E) and percentage of IFN-γ+ NK cells (F) for two or three independent experiments. Mean ± SD. Dotted line (mean) and gray shading (SD) represents values achieved after T reg cell ablation (a composite of multiple independent experiments).
Figure 3.
Figure 3.
An IL-2 footprint is induced by T reg ablation. (A) IL-2–induced (orange) and –repressed (blue) gene transcripts (Marzec et al., 2008) are superimposed on volcano plots (p-value versus FC) of pancreatic NK cell transcripts up-regulated (to the right) or down-regulated (to the left) by perturbation of T reg cells (data from Fig. 1) at 8 h (top) and 24 h (bottom). P-values calculated using the χ2 test. (B) Representative flow cytometry plots for NK cells isolated from the pancreas of BDC2.5/NOD mice 24 h after T reg ablation or not. (C) Summary data for three to four independent experiments with mean ± SD. (D, left) Representative flow cytometry plots for pancreas-infiltrating NK cells after T reg cell ablation; (right) summary for the percentage of CD25-expressing versus CD25-negative NK cells simultaneously expressing IFN-γ from at least three independent experiments.
Figure 4.
Figure 4.
Neutralization of IL-2 prevents the activation of pancreatic NK cells in response to T reg cell ablation. (A–D) The pancreatic infiltrate from BDC2.5/NOD mice (DTR+ or DTR− control littermates) was analyzed 24 h after DT injection ± anti–IL-2 mAb JES6-1 (or isotype control) co-injection. (left) Representative flow cytometry plots. (right) Summary data for fraction and numbers with mean ± SD from three to four independent experiments. (A) NK cells (NKp46+CD3−CD19−) and (B) Foxp3+ T reg cells (Foxp3+CD4+CD3+CD19−) in the pancreatic infiltrate (FSC/SSC lymphocyte gated). (C) IFN-γ–producing NK cells among total NK cells in the pancreatic infiltrate. (D) CD25 expression on pancreatic NK cells.
Figure 5.
Figure 5.
Supplementation with IL-2 induces early IFN-γ production and eventual accumulation of NK cells in the pancreatic lesion, as well as clinical diabetes. Pancreatic infiltrate from BDC2.5/NOD mice was analyzed 24 h after treatment with control PBS, IL-2–S4B6 complexes, or mutant IL-2 analogue Super-2, or 4 d after injection of IL-2–S4B6 complexes. (A, left) Representative flow cytometry data. (middle) Summary data for fraction of NK cells in the FSC/SSC lymphocyte gate. (right) Cell number summary data. (B) Analogous data for IFN-γ–producing NK cells. Mean ± SD, at least three independent experiments for both panels. (C) Summary diabetes data for 2 cohorts after 3 consecutive treatments with IL-2–S4B6 complexes (n = 14 IL-2–S4B6 complex; n = 9 PBS).
Figure 6.
Figure 6.
IL-2 production in the pancreas before and after T reg ablation. (A) Representative flow cytometry plots for pancreatic infiltrate from BDC2.5/NOD mice stained and analyzed for IL-2 by intracellular flow cytometry. An extended FSC/SSC gate was taken to include both lymphocytes and leukocytes along with a live/dead stain to exclude dead cells. CD45+IL-2+ cells were analyzed for CD3 and CD4 expression. (B) Summary data for pancreatic infiltrate from mice depleted of T reg cells (8 h with DT, DTR+) or not (8 h with DT, DTR−). Mean ± SD, at least three independent experiments. (C) Summary data, as in B, gated on CD4+ T cells.

References

    1. André I., Gonzalez A., Wang B., Katz J., Benoist C., Mathis D. 1996. Checkpoints in the progression of autoimmune disease: lessons from diabetes models. Proc. Natl. Acad. Sci. USA. 93:2260–2263 10.1073/pnas.93.6.2260
    1. Barthlott T., Moncrieffe H., Veldhoen M., Atkins C.J., Christensen J., O’Garra A., Stockinger B. 2005. CD25+ CD4+ T cells compete with naive CD4+ T cells for IL-2 and exploit it for the induction of IL-10 production. Int. Immunol. 17:279–288 10.1093/intimm/dxh207
    1. Bluestone J.A., Tang Q., Sedwick C.E. 2008. T regulatory cells in autoimmune diabetes: past challenges, future prospects. J. Clin. Immunol. 28:677–684 10.1007/s10875-008-9242-z
    1. Bonifacio E. 2012. Immunotherapy in type 1 diabetes: a shorter but more winding road? Diabetes. 61:2214–2215 10.2337/db12-0648
    1. Boyman O., Kovar M., Rubinstein M.P., Surh C.D., Sprent J. 2006. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science. 311:1924–1927 10.1126/science.1122927
    1. Cooper M.A., Elliott J.M., Keyel P.A., Yang L., Carrero J.A., Yokoyama W.M. 2009. Cytokine-induced memory-like natural killer cells. Proc. Natl. Acad. Sci. USA. 106:1915–1919 10.1073/pnas.0813192106
    1. Donohue J.H., Rosenberg S.A. 1983. The fate of interleukin-2 after in vivo administration. J. Immunol. 130:2203–2208
    1. Dybkaer K., Iqbal J., Zhou G., Geng H., Xiao L., Schmitz A., d’Amore F., Chan W.C. 2007. Genome wide transcriptional analysis of resting and IL2 activated human natural killer cells: gene expression signatures indicative of novel molecular signaling pathways. BMC Genomics. 8:230 10.1186/1471-2164-8-230
    1. Fehniger T.A., Cooper M.A., Nuovo G.J., Cella M., Facchetti F., Colonna M., Caligiuri M.A. 2003. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential new link between adaptive and innate immunity. Blood. 101:3052–3057 10.1182/blood-2002-09-2876
    1. Feuerer M., Shen Y., Littman D.R., Benoist C., Mathis D. 2009. How punctual ablation of regulatory T cells unleashes an autoimmune lesion within the pancreatic islets. Immunity. 31:654–664 10.1016/j.immuni.2009.08.023
    1. Feuerer M., Hill J.A., Kretschmer K., von Boehmer H., Mathis D., Benoist C. 2010. Genomic definition of multiple ex vivo regulatory T cell subphenotypes. Proc. Natl. Acad. Sci. USA. 107:5919–5924 10.1073/pnas.1002006107
    1. Friese M.A., Wischhusen J., Wick W., Weiler M., Eisele G., Steinle A., Weller M. 2004. RNA interference targeting transforming growth factor-beta enhances NKG2D-mediated antiglioma immune response, inhibits glioma cell migration and invasiveness, and abrogates tumorigenicity in vivo. Cancer Res. 64:7596–7603 10.1158/0008-5472.CAN-04-1627
    1. Gasteiger G., Hemmers S., Bos P.D., Sun J.C., Rudensky A.Y. 2013. IL-2–dependent adaptive control of NK cell homeostasis. J. Exp. Med. 210:xxx–xxx
    1. Gasteiger G., Hemmers S., Firth M.A., Le Floc’h A., Huse M., Sun J.C., Rudensky A.Y. 2013. IL-2–dependent tuning of NK cell sensitivity for target cells is controlled by regulatory T cells. J. Exp. Med. 210:xxx–xxx
    1. Ghiringhelli F., Ménard C., Terme M., Flament C., Taieb J., Chaput N., Puig P.E., Novault S., Escudier B., Vivier E., et al. 2005. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J. Exp. Med. 202:1075–1085 10.1084/jem.20051511
    1. Ghiringhelli F., Ménard C., Martin F., Zitvogel L. 2006. The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression. Immunol. Rev. 214:229–238 10.1111/j.1600-065X.2006.00445.x
    1. Ghiringhelli F., Menard C., Puig P.E., Ladoire S., Roux S., Martin F., Solary E., Le Cesne A., Zitvogel L., Chauffert B. 2007. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 56:641–648 10.1007/s00262-006-0225-8
    1. Gonzalez A., Katz J.D., Mattei M.G., Kikutani H., Benoist C., Mathis D. 1997. Genetic control of diabetes progression. Immunity. 7:873–883 10.1016/S1074-7613(00)80405-7
    1. Granucci F., Zanoni I., Pavelka N., Van Dommelen S.L., Andoniou C.E., Belardelli F., Degli Esposti M.A., Ricciardi-Castagnoli P. 2004. A contribution of mouse dendritic cell-derived IL-2 for NK cell activation. J. Exp. Med. 200:287–295 10.1084/jem.20040370
    1. Grinberg-Bleyer Y., Baeyens A., You S., Elhage R., Fourcade G., Gregoire S., Cagnard N., Carpentier W., Tang Q., Bluestone J., et al. 2010. IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J. Exp. Med. 207:1871–1878 10.1084/jem.20100209
    1. Hulme M.A., Wasserfall C.H., Atkinson M.A., Brusko T.M. 2012. Central role for interleukin-2 in type 1 diabetes. Diabetes. 61:14–22 10.2337/db11-1213
    1. Jin G.H., Hirano T., Murakami M. 2008. Combination treatment with IL-2 and anti-IL-2 mAbs reduces tumor metastasis via NK cell activation. Int. Immunol. 20:783–789 10.1093/intimm/dxn036
    1. Josefowicz S.Z., Lu L.F., Rudensky A.Y. 2012. Regulatory T Cells: Mechanisms of Differentiation and Function. Annu. Rev. Immunol. 30:531–564
    1. Katz J.D., Wang B., Haskins K., Benoist C., Mathis D. 1993. Following a diabetogenic T cell from genesis through pathogenesis. Cell. 74:1089–1100
    1. Kim J.M., Rasmussen J.P., Rudensky A.Y. 2007. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8:191–197 10.1038/ni1428
    1. Laouar Y., Sutterwala F.S., Gorelik L., Flavell R.A. 2005. Transforming growth factor-beta controls T helper type 1 cell development through regulation of natural killer cell interferon-gamma. Nat. Immunol. 6:600–607 10.1038/ni1197
    1. Lazarski C.A., Hughson A., Sojka D.K., Fowell D.J. 2008. Regulating Treg cells at sites of inflammation. Immunity. 29:511, author reply :512 10.1016/j.immuni.2008.09.012
    1. Lee J.C., Lee K.M., Kim D.W., Heo D.S. 2004. Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J. Immunol. 172:7335–7340
    1. Lee S.H., Fragoso M.F., Biron C.A. 2012. Cutting edge: a novel mechanism bridging innate and adaptive immunity: IL-12 induction of CD25 to form high-affinity IL-2 receptors on NK cells. J. Immunol. 189:2712–2716 10.4049/jimmunol.1201528
    1. Létourneau S., van Leeuwen E.M., Krieg C., Martin C., Pantaleo G., Sprent J., Surh C.D., Boyman O. 2010. IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25. Proc. Natl. Acad. Sci. USA. 107:2171–2176 10.1073/pnas.0909384107
    1. Levin A.M., Bates D.L., Ring A.M., Krieg C., Lin J.T., Su L., Moraga I., Raeber M.E., Bowman G.R., Novick P., et al. 2012. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature. 484:529–533 10.1038/nature10975
    1. Long S.A., Rieck M., Sanda S., Bollyky J.B., Samuels P.L., Goland R., Ahmann A., Rabinovitch A., Aggarwal S., Phippard D., et al. ; Diabetes TrialNet and the Immune Tolerance Network 2012. Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function. Diabetes. 61:2340–2348 10.2337/db12-0049
    1. Marzec M., Halasa K., Kasprzycka M., Wysocka M., Liu X., Tobias J.W., Baldwin D., Zhang Q., Odum N., Rook A.H., Wasik M.A. 2008. Differential effects of interleukin-2 and interleukin-15 versus interleukin-21 on CD4+ cutaneous T-cell lymphoma cells. Cancer Res. 68:1083–1091 10.1158/0008-5472.CAN-07-2403
    1. Mostböck S., Lutsiak M.E., Milenic D.E., Baidoo K., Schlom J., Sabzevari H. 2008. IL-2/anti-IL-2 antibody complex enhances vaccine-mediated antigen-specific CD8+ T cell responses and increases the ratio of effector/memory CD8+ T cells to regulatory T cells. J. Immunol. 180:5118–5129
    1. Nikitina-Zake L., Rajalingham R., Rumba I., Sanjeevi C.B. 2004. Killer cell immunoglobulin-like receptor genes in Latvian patients with type 1 diabetes mellitus and healthy controls. Ann. N. Y. Acad. Sci. 1037:161–169 10.1196/annals.1337.027
    1. Nir T., Melton D.A., Dor Y. 2007. Recovery from diabetes in mice by beta cell regeneration. J. Clin. Invest. 117:2553–2561 10.1172/JCI32959
    1. Pandiyan P., Zheng L., Ishihara S., Reed J., Lenardo M.J. 2007. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nat. Immunol. 8:1353–1362 10.1038/ni1536
    1. Ramos-Lopez E., Scholten F., Aminkeng F., Wild C., Kalhes H., Seidl C., Tonn T., Van der Auwera B., Badenhoop K. 2009. Association of KIR2DL2 polymorphism rs2756923 with type 1 diabetes and preliminary evidence for lack of inhibition through HLA-C1 ligand binding. Tissue Antigens. 73:599–603 10.1111/j.1399-0039.2009.01252.x
    1. Reynolds L.A., Maizels R.M. 2012. Cutting edge: in the absence of TGF-β signaling in T cells, fewer CD103+ regulatory T cells develop, but exuberant IFN-γ production renders mice more susceptible to helminth infection. J. Immunol. 189:1113–1117 10.4049/jimmunol.1200991
    1. Rodacki M., Svoren B., Butty V., Besse W., Laffel L., Benoist C., Mathis D. 2007. Altered natural killer cells in type 1 diabetic patients. Diabetes. 56:177–185 10.2337/db06-0493
    1. Rogner U.C., Boitard C., Morin J., Melanitou E., Avner P. 2001. Three loci on mouse chromosome 6 influence onset and final incidence of type I diabetes in NOD.C3H congenic strains. Genomics. 74:163–171 10.1006/geno.2001.6508
    1. Rose J.W. 2012. Anti-CD25 immunotherapy: regulating the regulators. Sci Transl. Med. 4:145fs25
    1. Rudensky A.Y. 2011. Regulatory T cells and Foxp3. Immunol. Rev. 241:260–268 10.1111/j.1600-065X.2011.01018.x
    1. Scheffold A., Hühn J., Höfer T. 2005. Regulation of CD4+CD25+ regulatory T cell activity: it takes (IL-)two to tango. Eur. J. Immunol. 35:1336–1341 10.1002/eji.200425887
    1. Shevach E.M. 2012. Application of IL-2 therapy to target T regulatory cell function. Trends Immunol. 33:626–632
    1. Shimizu J., Yamazaki S., Sakaguchi S. 1999. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J. Immunol. 163:5211–5218
    1. Shoda L.K., Young D.L., Ramanujan S., Whiting C.C., Atkinson M.A., Bluestone J.A., Eisenbarth G.S., Mathis D., Rossini A.A., Campbell S.E., et al. 2005. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity. 23:115–126 10.1016/j.immuni.2005.08.002
    1. Smyth M.J., Teng M.W., Swann J., Kyparissoudis K., Godfrey D.I., Hayakawa Y. 2006. CD4+CD25+ T regulatory cells suppress NK cell-mediated immunotherapy of cancer. J. Immunol. 176:1582–1587
    1. Sojka D.K., Huang Y.H., Fowell D.J. 2008. Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunology. 124:13–22 10.1111/j.1365-2567.2008.02813.x
    1. Tang Q., Walker L., Bluestone J. 2008a. Response: Regulating Treg Cells at Sites of Inflammation. Immunity. 29:512 10.1016/j.immuni.2008.09.011
    1. Tang Q., Adams J.Y., Penaranda C., Melli K., Piaggio E., Sgouroudis E., Piccirillo C.A., Salomon B.L., Bluestone J.A. 2008b. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity. 28:687–697 10.1016/j.immuni.2008.03.016
    1. Uhl M., Aulwurm S., Wischhusen J., Weiler M., Ma J.Y., Almirez R., Mangadu R., Liu Y.W., Platten M., Herrlinger U., et al. 2004. SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo. Cancer Res. 64:7954–7961 10.1158/0008-5472.CAN-04-1013
    1. van der Slik A.R., Koeleman B.P., Verduijn W., Bruining G.J., Roep B.O., Giphart M.J. 2003. KIR in type 1 diabetes: disparate distribution of activating and inhibitory natural killer cell receptors in patients versus HLA-matched control subjects. Diabetes. 52:2639–2642 10.2337/diabetes.52.10.2639
    1. Verdeil G., Puthier D., Nguyen C., Schmitt-Verhulst A.M., Auphan-Anezin N. 2006. STAT5-mediated signals sustain a TCR-initiated gene expression program toward differentiation of CD8 T cell effectors. J. Immunol. 176:4834–4842
    1. Vignali D.A., Collison L.W., Workman C.J. 2008. How regulatory T cells work. Nat. Rev. Immunol. 8:523–532 10.1038/nri2343

Source: PubMed

Подписаться