Type I interferon is selectively required by dendritic cells for immune rejection of tumors

Mark S Diamond, Michelle Kinder, Hirokazu Matsushita, Mona Mashayekhi, Gavin P Dunn, Jessica M Archambault, Hsiaoju Lee, Cora D Arthur, J Michael White, Ulrich Kalinke, Kenneth M Murphy, Robert D Schreiber, Mark S Diamond, Michelle Kinder, Hirokazu Matsushita, Mona Mashayekhi, Gavin P Dunn, Jessica M Archambault, Hsiaoju Lee, Cora D Arthur, J Michael White, Ulrich Kalinke, Kenneth M Murphy, Robert D Schreiber

Abstract

Cancer immunoediting is the process whereby the immune system suppresses neoplastic growth and shapes tumor immunogenicity. We previously reported that type I interferon (IFN-α/β) plays a central role in this process and that hematopoietic cells represent critical targets of type I IFN's actions. However, the specific cells affected by IFN-α/β and the functional processes that type I IFN induces remain undefined. Herein, we show that type I IFN is required to initiate the antitumor response and that its actions are temporally distinct from IFN-γ during cancer immunoediting. Using mixed bone marrow chimeric mice, we demonstrate that type I IFN sensitivity selectively within the innate immune compartment is essential for tumor-specific T cell priming and tumor elimination. We further show that mice lacking IFNAR1 (IFN-α/β receptor 1) in dendritic cells (DCs; Itgax-Cre(+)Ifnar1(f/f) mice) cannot reject highly immunogenic tumor cells and that CD8α(+) DCs from these mice display defects in antigen cross-presentation to CD8(+) T cells. In contrast, mice depleted of NK cells or mice that lack IFNAR1 in granulocytes and macrophage populations reject these tumors normally. Thus, DCs and specifically CD8α(+) DCs are functionally relevant targets of endogenous type I IFN during lymphocyte-mediated tumor rejection.

Figures

Figure 1.
Figure 1.
Early requirement for IFN-α/β during rejection of highly immunogenic tumor cells. (A) Untreated WT and Rag2−/− mice or WT mice injected i.p. with either IFNAR1-specific MAR1-5A3 mAb or isotype control GIR-208 mAb 1 d prior were s.c. injected with 106 H31m1 tumor cells, and tumor size was measured over time. Data represent mean tumor diameter ± SEM of 12–16 mice per group from at least three independent experiments. (B–D) WT mice were injected with 106 H31m1 cells (at day 0) and treated beginning on the indicated day with MAR1-5A3 (B), IFN-γ–specific H22 mAb (C), or a mixture of anti-CD4/anti-CD8/anti–IFN-γ mAbs GK1.5/YTS-169.4/H22 (D), and tumor growth was monitored. For each time point, groups of mice were treated in parallel with the respective isotype-matched control mAb, and the data are presented as percent tumor growth over the control group. Results are from two to four experiments with 14–20 (ctrl/MAR1-5A3), 10–20 (ctrl/H22), or 6–11 (ctrl/cocktail) WT mice per group. The kinetics of tumor growth in individual mice is shown in Fig. S1.
Figure 2.
Figure 2.
Nonoverlapping host cell targets for IFN-α/β and IFN-γ during tumor rejection. (A–C) Control mice and the indicated bone marrow chimeras with selective IFN-α/β sensitivity (A and B) or IFN-γ sensitivity (C) in hematopoietic versus nonhematopoietic cells were injected s.c. with 106 H31m1 (A) or d38m2 (B and C) unedited MCA sarcoma cells, and growth was monitored. Data are presented as mean tumor diameter ± SEM over time or the percentage of tumor-positive mice per group from two to three (A and B) or five (C) independent experiments with group sizes as indicated. Hematopoietic reconstitution of all Ifnar1−/− and Ifngr1−/− bone marrow chimeras was confirmed by flow cytometry at the conclusion of each experiment.
Figure 3.
Figure 3.
IFN-α/β sensitivity within the innate immune compartment is necessary and sufficient for tumor rejection. Mixed bone marrow chimeras with selective IFNAR1 expression in innate or adaptive immune cells were generated by reconstitution of irradiated Ifnar1−/− mice with mixtures of HSCs as described in Results. (A) Splenocytes were isolated from representative cohorts of control and mixed chimeric mice at least 12 wk after reconstitution, and IFNAR1 staining was analyzed by flow cytometry. Shown are the percentages of IFNAR1+ cells within the indicated immune cell subsets for 8–14 mice of each type. Horizontal bars represent the mean. (B–D) Control WT, Rag2−/−, and Ifnar1−/− mice and Ifnar1−/− mixed chimeric mice were injected with 106 H31m1 (B), d38m2 (C), or F515 (D) tumor cells, and growth was monitored over time. Data are presented as mean tumor diameter ± SEM or the percentage or tumor-positive mice per group from two to three independent experiments with group sizes as indicated. WT mice treated with control or IFN-γ–specific mAb were challenged with 106 F515 tumor cells, and growth was monitored (D, bottom). Mean tumor diameter ± SEM for 7–10 mice/group from two experiments is shown.
Figure 4.
Figure 4.
Normal hematopoietic reconstitution in Ifnar1−/− mixed bone marrow chimeras. (A) Spleens were harvested from WT, Ifnar1−/−, or Ifnar1−/− mixed chimeras of each type (12 wk after reconstitution), and cell density was determined. Horizontal bars represent the mean. (B) Percentages of the indicated immune cell subsets were measured by flow cytometry for WT, Ifnar1−/−, and Ifnar1−/− mixed chimeras. Mean values (as a percentage of total splenocytes) ± SEM for four to five mice/group are shown. Cell populations were defined as follows: CD4+ T cells (CD3+CD4+), CD8+ T cells (CD3+CD8+), B cells (B220+), NK cells (DX5+CD3−), DCs (CD11chi), and myeloid cells (CD11b+). (C) WT-derived 1877 tumor cells were injected at a dose of 106 cells/mouse into WT, Ifnar1−/−, Rag2−/−, and Ifnar1−/− mixed chimeras, and tumor growth was monitored over time. Data represent the mean tumor diameter ± SEM for three to eight mice/group. (A–C) Data are representative of two independent experiments.
Figure 5.
Figure 5.
Impaired tumor-specific CTL priming in Ifnar1−/− mice is restored by IFN-α/β–responsive innate immune cells. (A) Splenocytes from WT and Ifnar1−/− mice were isolated 20 d after H31m1 tumor challenge (106 cells/mouse), co-cultured with IFN-γ–treated, irradiated H31m1 cells, and 5 d later used as effectors in a cytotoxicity assay with 51Cr-labeled H31m1 targets. Specific killing activity (in percentage ± SEM) at the indicated effector/target (E:T) ratios is shown for four to five mice per group assayed in duplicate from three independent experiments. (B) Splenocytes were harvested from the indicated chimeric mice 20 d after injection of 106 H31m1 tumor cells and were treated as in A. Data include representative results from three mice per group assayed in duplicate from two independent experiments. Splenocytes harvested from a naive mouse and treated similarly served as a negative control. (C) Effector cells from H31m1-challenged innate chimeras were co-cultured at the indicated effector/target ratios with 51Cr-labeled H31m1 targets in the presence of 10 µg/ml control (PIP), anti-CD4 (GK1.5), or anti-CD8 (YTS-169.4) mAbs. Data show representative results from three mice per group assayed in duplicate from three independent experiments. Similar results were obtained when effector cells from H31m1-injected WT mice were used (not depicted). (B and C) Error bars represent SEM.
Figure 6.
Figure 6.
NK cell depletion does not abrogate IFN-α/β–dependent rejection of immunogenic sarcomas. (A and B) C57BL/6 WT, Rag2−/−, and Ifnar1−/− mice and WT mice treated with either PBS or anti-NK1.1 PK136 mAb were injected s.c. (106 cells/mouse) with 1969 (A) or 7835 (B) unedited MCA sarcoma cells, and growth was monitored over time. Data are presented as mean tumor diameter ± SEM of 4–13 (untreated) or 8 (treated) mice per group from at least two independent experiments. Error bars for Ifnar1−/− mice reflect progressive growth of 1969 and 7835 tumors in 6/9 mice. (C) WT C57BL/6 mice were treated with either PBS or PK136 mAb, and splenocytes were harvested 2 d later and analyzed by flow cytometry using the NK cell markers DX5 and NKp46. Splenocytes were gated on CD3− cells, and the percentages of DX5+NKp46+ cells are indicated. Similar results were found when harvested at day 6 (not depicted). (D) WT C57BL/6 mice were treated with PBS or PK136 followed by i.p. injection of 300 µg polyI:C 4 d later. After 24 h, splenocytes were harvested and used as effectors in a standard 4-h cytotoxicity assay with NK-sensitive YAC-1 targets. Specific lysis (in percentage ± SEM) at the indicated effector/target (E:T) ratios is shown for four mice/group assayed in duplicate from two independent experiments. (E) WT C57BL/6 mice were treated with PBS, PK136, or a mixture of anti-CD4 (GK1.5) and anti-CD8 (YTS-169.4) mAbs and injected s.c. with 105 RMA-S cells, and tumor growth was monitored over time. Mean tumor diameter ± SEM for three mice/group is shown, and data are representative of two independent experiments.
Figure 7.
Figure 7.
Granulocytes and macrophages do not require type I IFN sensitivity for tumor rejection. (A) IFNAR1 expression on peritoneal macrophages, blood monocytes, PMNs, and B cells was measured using flow cytometry in Ifnar1f/f, LysM-Cre+Ifnar1f/f, and Ifnar1−/− mice. (B) Summary of IFNAR1 levels in the indicated cellular subsets in LysM-Cre+Ifnar1f/f mice compared with Ifnar1f/f mice (expressed as a percentage of the mean fluorescence intensity [MFI]). Cells were gated using the following markers: macrophages (CD11b+F4/80+), PMNs (CD11b+Gr1+), monocytes (CD115+CD11b+), B cells (B220+), CD8α+ DCs (CD8α+Dec205+CD11chi), CD4+ DCs (CD8α−Dec205−CD11chiCD4+), pDCs (B220+PDCA+CD11cint), T cells (CD3+), and NK cells (NK1.1+). IFNAR1 expression was measured using MAR1-5A3 mAb. Data represent at least three mice from three independent experiments (**, P < 0.01). (C) Mature peritoneal macrophages from LysM-Cre+Ifnar1f/f mice were untreated (gray) or stimulated for 15 min with 10 ng/ml IFN-αv4 (black), and pSTAT1 accumulation was measured by flow cytometry. Histograms from a representative experiment are shown, with the bar graph summarizing pSTAT1 levels (as percentage of control Ifnar1f/f MFI) from two independent experiments. (B and C) Error bars represent SEM. (D) Ifnar1f/f, LysM-Cre+Ifnar1f/f, and Ifnar1−/− mice were injected s.c. with 106 1969 unedited sarcoma cells. Mean tumor diameter ± SEM from a representative experiment is shown, and the bar graph shows the percentage of tumor-positive mice per group from two independent experiments with indicated total group sizes.
Figure 8.
Figure 8.
DCs specifically require type I IFN sensitivity for tumor immunity in vivo. (A) IFNAR1 expression on splenic CD8α+ DCs, CD4+ DCs, pDCs, LN CD103+ DCs, and dermal DCs was measured using flow cytometry in Ifnar1f/f, Itgax-Cre+Ifnar1f/f, and Ifnar1−/− mice. (B) Summary of IFNAR1 levels on the indicated cellular subsets in Itgax-Cre+Ifnar1f/f mice compared with Ifnar1f/f mice (expressed as a percentage of control mean fluorescence intensity [MFI]). Cells were gated as follows: CD8α+ DCs (CD8α+Dec205+CD11chi), CD103 DCs (CD8α−Dec205+CD11chiCD103+), CD4+ DCs (CD8α−Dec205−CD11chiCD4+), dermal DCs (CD8α−CD11chiCD103−), pDCs (B220+PDCA+CD11cint), B cells (B220+), T cells (CD3+), NK cells (NK1.1+), macrophages (CD11b+F4/80+), and blood PMNs (CD11b+Gr1+). IFNAR1 expression was measured using the MAR1-5A3 mAb. Data represent three to five mice from at least three independent experiments. (**, P < 0.01). (C) Splenocytes from Itgax-Cre+Ifnar1f/f mice were untreated (gray) or stimulated for 15 min with 10 ng/ml IFN-αv4 (black), and pSTAT1 accumulation in CD8α+ and CD4+ DCs was measured by flow cytometry. Histograms show a representative experiment, and the bar graph summarizes results from four independent experiments (**, P < 0.01). (B and C) Error bars represent SEM. (D) C57BL/6 WT, Ifnar1−/−, Ifnar1f/f, and Itgax-Cre+Ifnar1f/f mice were injected s.c. with 106 1969 unedited sarcoma cells. Mean tumor diameter ± SEM from a representative experiment is shown, and the bar graph shows a summary of the percentage of tumor-positive mice per group from three independent experiments with indicated groups sizes (P < 0.001 [WT vs. Ifnar1−/−] and P < 0.001 [Ifnar1f/f vs. Itgax-Cre+Ifnar1f/f]) using the Student’s t test at day 23. Comparisons of Ifnar1−/− versus Itgax-Cre+Ifnar1f/f or WT versus Ifnar1f/f were not significantly different.
Figure 9.
Figure 9.
Type I IFN sensitivity in CD8α+ DCs enhances antigen cross-presentation. (A) CD11c+ cells were isolated from the spleens of WT or Ifnar1−/− mice and co-cultured with the indicated number of irradiated, ovalbumin-loaded MHC class I−/− splenocytes and CFSE-labeled OT-I T cells. After a 3-d incubation, proliferation of OT-I T cells was determined by CSFE dilution. Histograms represent CFSE levels in the CD8+ T cell population, with the percentage of cells in the indicated gate noted. (B) WT and Ifnar1−/− CD11c+ cells or WT CD11c+ cells incubated with exogenous 1,000 U/ml IFN-α or 5 µg/ml IFNAR1-specific MAR1-5A3 mAb were treated as in A at a dose of 25,000 MHC class I−/− splenocytes. (C) Purified CD8α+ and CD4+ DC subsets isolated from WT or Ifnar1−/− mice were treated as in A with the indicated number of ovalbumin-loaded MHC class I−/− splenocytes. Data represent one of at least two independent experiments with similar results.
Figure 10.
Figure 10.
Impaired antigen cross-presentation in CD8α+ DCs from Itgax-Cre+Ifnar1f/f mice. CD8α+ DCs were isolated from Ifnar1f/f, Itgax-Cre+Ifnar1f/f, and Ifnar1−/− mice and incubated with OT-I T cells labeled with cell proliferation dye and 12,500 ovalbumin-loaded MHC class I−/− splenocytes. Dilution of the cell proliferation dye was measured 3 d later. Data represent one of at least two independent experiments with similar results.

References

    1. Ackerman A.L., Cresswell P. 2004. Cellular mechanisms governing cross-presentation of exogenous antigens. Nat. Immunol. 5:678–684 10.1038/ni1082
    1. Belardelli F., Ferrantini M., Proietti E., Kirkwood J.M. 2002. Interferon-alpha in tumor immunity and immunotherapy. Cytokine Growth Factor Rev. 13:119–134 10.1016/S1359-6101(01)00022-3
    1. Biron C.A., Nguyen K.B., Pien G.C., Cousens L.P., Salazar-Mather T.P. 1999. Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu. Rev. Immunol. 17:189–220 10.1146/annurev.immunol.17.1.189
    1. Christensen J.L., Wright D.E., Wagers A.J., Weissman I.L. 2004. Circulation and chemotaxis of fetal hematopoietic stem cells. PLoS Biol. 2:E75 10.1371/journal.pbio.0020075
    1. Clausen B.E., Burkhardt C., Reith W., Renkawitz R., Förster I. 1999. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8:265–277 10.1023/A:1008942828960
    1. Cobbold S.P., Jayasuriya A., Nash A., Prospero T.D., Waldmann H. 1984. Therapy with monoclonal antibodies by elimination of T-cell subsets in vivo. Nature. 312:548–551 10.1038/312548a0
    1. Coro E.S., Chang W.L., Baumgarth N. 2006. Type I IFN receptor signals directly stimulate local B cells early following influenza virus infection. J. Immunol. 176:4343–4351
    1. Curtsinger J.M., Valenzuela J.O., Agarwal P., Lins D., Mescher M.F. 2005. Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation. J. Immunol. 174:4465–4469
    1. Dialynas D.P., Quan Z.S., Wall K.A., Pierres A., Quintáns J., Loken M.R., Pierres M., Fitch F.W. 1983. Characterization of the murine T cell surface molecule, designated L3T4, identified by monoclonal antibody GK1.5: similarity of L3T4 to the human Leu-3/T4 molecule. J. Immunol. 131:2445–2451
    1. Dighe A.S., Richards E., Old L.J., Schreiber R.D. 1994. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity. 1:447–456 10.1016/1074-7613(94)90087-6
    1. Dudziak D., Kamphorst A.O., Heidkamp G.F., Buchholz V.R., Trumpfheller C., Yamazaki S., Cheong C., Liu K., Lee H.W., Park C.G., et al. 2007. Differential antigen processing by dendritic cell subsets in vivo. Science. 315:107–111 10.1126/science.1136080
    1. Dunn G.P., Bruce A.T., Ikeda H., Old L.J., Schreiber R.D. 2002. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3:991–998 10.1038/ni1102-991
    1. Dunn G.P., Old L.J., Schreiber R.D. 2004. The three Es of cancer immunoediting. Annu. Rev. Immunol. 22:329–360 10.1146/annurev.immunol.22.012703.104803
    1. Dunn G.P., Bruce A.T., Sheehan K.C., Shankaran V., Uppaluri R., Bui J.D., Diamond M.S., Koebel C.M., Arthur C., White J.M., Schreiber R.D. 2005. A critical function for type I interferons in cancer immunoediting. Nat. Immunol. 6:722–729 10.1038/ni1213
    1. Dunn G.P., Koebel C.M., Schreiber R.D. 2006. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 6:836–848 10.1038/nri1961
    1. Edelson B.T., Kc W., Juang R., Kohyama M., Benoit L.A., Klekotka P.A., Moon C., Albring J.C., Ise W., Michael D.G., et al. 2010. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8α+ conventional dendritic cells. J. Exp. Med. 207:823–836 10.1084/jem.20091627
    1. Gallucci S., Lolkema M., Matzinger P. 1999. Natural adjuvants: endogenous activators of dendritic cells. Nat. Med. 5:1249–1255 10.1038/15200
    1. Ginhoux F., Liu K., Helft J., Bogunovic M., Greter M., Hashimoto D., Price J., Yin N., Bromberg J., Lira S.A., et al. 2009. The origin and development of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206:3115–3130 10.1084/jem.20091756
    1. Gogas H., Ioannovich J., Dafni U., Stavropoulou-Giokas C., Frangia K., Tsoutsos D., Panagiotou P., Polyzos A., Papadopoulos O., Stratigos A., et al. 2006. Prognostic significance of autoimmunity during treatment of melanoma with interferon. N. Engl. J. Med. 354:709–718 10.1056/NEJMoa053007
    1. Havenar-Daughton C., Kolumam G.A., Murali-Krishna K. 2006. Cutting Edge: The direct action of type I IFN on CD4 T cells is critical for sustaining clonal expansion in response to a viral but not a bacterial infection. J. Immunol. 176:3315–3319
    1. Heath W.R., Belz G.T., Behrens G.M., Smith C.M., Forehan S.P., Parish I.A., Davey G.M., Wilson N.S., Carbone F.R., Villadangos J.A. 2004. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol. Rev. 199:9–26 10.1111/j.0105-2896.2004.00142.x
    1. Hildner K., Edelson B.T., Purtha W.E., Diamond M., Matsushita H., Kohyama M., Calderon B., Schraml B.U., Unanue E.R., Diamond M.S., et al. 2008. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science. 322:1097–1100 10.1126/science.1164206
    1. Hogquist K.A., Jameson S.C., Heath W.R., Howard J.L., Bevan M.J., Carbone F.R. 1994. T cell receptor antagonist peptides induce positive selection. Cell. 76:17–27 10.1016/0092-8674(94)90169-4
    1. Huang A.Y., Golumbek P., Ahmadzadeh M., Jaffee E., Pardoll D., Levitsky H. 1994. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 264:961–965 10.1126/science.7513904
    1. Jablonska J., Leschner S., Westphal K., Lienenklaus S., Weiss S. 2010. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J. Clin. Invest. 120:1151–1164 10.1172/JCI37223
    1. Kamphuis E., Junt T., Waibler Z., Forster R., Kalinke U. 2006. Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia. Blood. 108:3253–3261 10.1182/blood-2006-06-027599
    1. Kaplan D.H., Shankaran V., Dighe A.S., Stockert E., Aguet M., Old L.J., Schreiber R.D. 1998. Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc. Natl. Acad. Sci. USA. 95:7556–7561 10.1073/pnas.95.13.7556
    1. Kärre K., Ljunggren H.G., Piontek G., Kiessling R. 1986. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 319:675–678 10.1038/319675a0
    1. Koebel C.M., Vermi W., Swann J.B., Zerafa N., Rodig S.J., Old L.J., Smyth M.J., Schreiber R.D. 2007. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 450:903–907 10.1038/nature06309
    1. Kolumam G.A., Thomas S., Thompson L.J., Sprent J., Murali-Krishna K. 2005. Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J. Exp. Med. 202:637–650 10.1084/jem.20050821
    1. Koo G.C., Peppard J.R. 1984. Establishment of monoclonal anti-Nk-1.1 antibody. Hybridoma. 3:301–303 10.1089/hyb.1984.3.301
    1. Le Bon A., Schiavoni G., D’Agostino G., Gresser I., Belardelli F., Tough D.F. 2001. Type i interferons potently enhance humoral immunity and can promote isotype switching by stimulating dendritic cells in vivo. Immunity. 14:461–470 10.1016/S1074-7613(01)00126-1
    1. Le Bon A., Etchart N., Rossmann C., Ashton M., Hou S., Gewert D., Borrow P., Tough D.F. 2003. Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat. Immunol. 4:1009–1015 10.1038/ni978
    1. Le Bon A., Durand V., Kamphuis E., Thompson C., Bulfone-Paus S., Rossmann C., Kalinke U., Tough D.F. 2006a. Direct stimulation of T cells by type I IFN enhances the CD8+ T cell response during cross-priming. J. Immunol. 176:4682–4689
    1. Le Bon A., Thompson C., Kamphuis E., Durand V., Rossmann C., Kalinke U., Tough D.F. 2006b. Cutting edge: enhancement of antibody responses through direct stimulation of B and T cells by type I IFN. J. Immunol. 176:2074–2078
    1. Longhi M.P., Trumpfheller C., Idoyaga J., Caskey M., Matos I., Kluger C., Salazar A.M., Colonna M., Steinman R.M. 2009. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J. Exp. Med. 206:1589–1602 10.1084/jem.20090247
    1. Lorenzi S., Mattei F., Sistigu A., Bracci L., Spadaro F., Sanchez M., Spada M., Belardelli F., Gabriele L., Schiavoni G. 2011. Type I IFNs control antigen retention and survival of CD8α(+) dendritic cells after uptake of tumor apoptotic cells leading to cross-priming. J. Immunol. 186:5142–5150 10.4049/jimmunol.1004163
    1. Luft T., Pang K.C., Thomas E., Hertzog P., Hart D.N., Trapani J., Cebon J. 1998. Type I IFNs enhance the terminal differentiation of dendritic cells. J. Immunol. 161:1947–1953
    1. Lybarger L., Wang X., Harris M.R., Virgin H.W., IV, Hansen T.H. 2003. Virus subversion of the MHC class I peptide-loading complex. Immunity. 18:121–130 10.1016/S1074-7613(02)00509-5
    1. Marrack P., Kappler J., Mitchell T. 1999. Type I interferons keep activated T cells alive. J. Exp. Med. 189:521–530 10.1084/jem.189.3.521
    1. Melief C.J. 2008. Cancer immunotherapy by dendritic cells. Immunity. 29:372–383 10.1016/j.immuni.2008.08.004
    1. Mombaerts P., Iacomini J., Johnson R.S., Herrup K., Tonegawa S., Papaioannou V.E. 1992. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 68:869–877 10.1016/0092-8674(92)90030-G
    1. Montoya M., Schiavoni G., Mattei F., Gresser I., Belardelli F., Borrow P., Tough D.F. 2002. Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood. 99:3263–3271 10.1182/blood.V99.9.3263
    1. Mumberg D., Monach P.A., Wanderling S., Philip M., Toledano A.Y., Schreiber R.D., Schreiber H. 1999. CD4(+) T cells eliminate MHC class II-negative cancer cells in vivo by indirect effects of IFN-gamma. Proc. Natl. Acad. Sci. USA. 96:8633–8638 10.1073/pnas.96.15.8633
    1. Nguyen K.B., Salazar-Mather T.P., Dalod M.Y., Van Deusen J.B., Wei X.Q., Liew F.Y., Caligiuri M.A., Durbin J.E., Biron C.A. 2002. Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 169:4279–4287
    1. Ochsenbein A.F., Sierro S., Odermatt B., Pericin M., Karrer U., Hermans J., Hemmi S., Hengartner H., Zinkernagel R.M. 2001. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature. 411:1058–1064 10.1038/35082583
    1. Pilz A., Kratky W., Stockinger S., Simma O., Kalinke U., Lingnau K., von Gabain A., Stoiber D., Sexl V., Kolbe T., et al. 2009. Dendritic cells require STAT-1 phosphorylated at its transactivating domain for the induction of peptide-specific CTL. J. Immunol. 183:2286–2293 10.4049/jimmunol.0901383
    1. Prinz M., Schmidt H., Mildner A., Knobeloch K.P., Hanisch U.K., Raasch J., Merkler D., Detje C., Gutcher I., Mages J., et al. 2008. Distinct and nonredundant in vivo functions of IFNAR on myeloid cells limit autoimmunity in the central nervous system. Immunity. 28:675–686 10.1016/j.immuni.2008.03.011
    1. Qin Z., Blankenstein T. 2000. CD4+ T cell—mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN gamma receptor expression by nonhematopoietic cells. Immunity. 12:677–686 10.1016/S1074-7613(00)80218-6
    1. Qin Z., Schwartzkopff J., Pradera F., Kammertoens T., Seliger B., Pircher H., Blankenstein T. 2003. A critical requirement of interferon gamma-mediated angiostasis for tumor rejection by CD8+ T cells. Cancer Res. 63:4095–4100
    1. Schreiber R.D., Hicks L.J., Celada A., Buchmeier N.A., Gray P.W. 1985. Monoclonal antibodies to murine gamma-interferon which differentially modulate macrophage activation and antiviral activity. J. Immunol. 134:1609–1618
    1. Schreiber R.D., Old L.J., Smyth M.J. 2011. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 331:1565–1570 10.1126/science.1203486
    1. Shankaran V., Ikeda H., Bruce A.T., White J.M., Swanson P.E., Old L.J., Schreiber R.D. 2001. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 410:1107–1111 10.1038/35074122
    1. Sheehan K.C., Lai K.S., Dunn G.P., Bruce A.T., Diamond M.S., Heutel J.D., Dungo-Arthur C., Carrero J.A., White J.M., Hertzog P.J., Schreiber R.D. 2006. Blocking monoclonal antibodies specific for mouse IFN-alpha/beta receptor subunit 1 (IFNAR-1) from mice immunized by in vivo hydrodynamic transfection. J. Interferon Cytokine Res. 26:804–819 10.1089/jir.2006.26.804
    1. Smyth M.J., Dunn G.P., Schreiber R.D. 2006. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv. Immunol. 90:1–50 10.1016/S0065-2776(06)90001-7
    1. Spiotto M.T., Rowley D.A., Schreiber H. 2004. Bystander elimination of antigen loss variants in established tumors. Nat. Med. 10:294–298 10.1038/nm999
    1. Steinman R.M., Banchereau J. 2007. Taking dendritic cells into medicine. Nature. 449:419–426 10.1038/nature06175
    1. Steinman R.M., Hawiger D., Nussenzweig M.C. 2003. Tolerogenic dendritic cells. Annu. Rev. Immunol. 21:685–711 10.1146/annurev.immunol.21.120601.141040
    1. Stetson D.B., Medzhitov R. 2006. Type I interferons in host defense. Immunity. 25:373–381 10.1016/j.immuni.2006.08.007
    1. Stranges P.B., Watson J., Cooper C.J., Choisy-Rossi C.M., Stonebraker A.C., Beighton R.A., Hartig H., Sundberg J.P., Servick S., Kaufmann G., et al. 2007. Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity. 26:629–641 10.1016/j.immuni.2007.03.016
    1. Street S.E., Cretney E., Smyth M.J. 2001. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood. 97:192–197 10.1182/blood.V97.1.192
    1. Street S.E., Trapani J.A., MacGregor D., Smyth M.J. 2002. Suppression of lymphoma and epithelial malignancies effected by interferon gamma. J. Exp. Med. 196:129–134 10.1084/jem.20020063
    1. Swann J.B., Hayakawa Y., Zerafa N., Sheehan K.C., Scott B., Schreiber R.D., Hertzog P., Smyth M.J. 2007. Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J. Immunol. 178:7540–7549
    1. Thompson L.J., Kolumam G.A., Thomas S., Murali-Krishna K. 2006. Innate inflammatory signals induced by various pathogens differentially dictate the IFN-I dependence of CD8 T cells for clonal expansion and memory formation. J. Immunol. 177:1746–1754
    1. Vesely M.D., Kershaw M.H., Schreiber R.D., Smyth M.J. 2011. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 29:235–271 10.1146/annurev-immunol-031210-101324
    1. Wolkers M.C., Stoetter G., Vyth-Dreese F.A., Schumacher T.N. 2001. Redundancy of direct priming and cross-priming in tumor-specific CD8+ T cell responses. J. Immunol. 167:3577–3584
    1. Zietara N., Łyszkiewicz M., Gekara N., Puchałka J., Dos Santos V.A., Hunt C.R., Pandita T.K., Lienenklaus S., Weiss S. 2009. Absence of IFN-beta impairs antigen presentation capacity of splenic dendritic cells via down-regulation of heat shock protein 70. J. Immunol. 183:1099–1109

Source: PubMed

3
Předplatit