NK cells express an array of activating and inhibitory receptors that determine NK cell responses upon triggering by cognate ligands. Although activating NK cell receptors recognize mainly ligands expressed by stressed, virus-infected, or transformed cells, most inhibitory receptors engage MHC class I, preventing NK cell activation in response to healthy cells. In this study, we provide insight into the regulation and function of additional receptors involved in mouse NK cell responses: CTLA-4 and CD28. CTLA-4 and CD28 engage the same ligands, B7-1 and B7-2, which are primarily expressed by APCs, such as dendritic cells. Our data demonstrate that activation of mouse NK cells with IL-2 induces the expression of CTLA-4 and upregulates CD28. CTLA-4 expression in IL-2–expanded NK cells was further up- or downregulated by IL-12 or TGF-β, respectively. Using gene-deficient NK cells, we show that CD28 induces, and CTLA-4 inhibits, IFN-γ release by NK cells upon engagement by the recombinant ligand, B7-1, or upon coculture with mature dendritic cells. Notably, we show that mouse NK cells infiltrating solid tumors express CD28 and CTLA-4 and respond to stimulation with recombinant B7-1, suggesting that the NK cell responses mediated by the CD28/CTLA-4:B7-1/B7-2 system could be of importance during malignant disease. Accordingly, our study might have implications for immunotherapy of cancer based on blocking anti–CTLA-4 mAbs.

Natural killer cells are innate immune effectors that play an important role during antiviral and antitumor immune responses (1, 2). NK cells express an array of activating and inhibitory surface receptors whose engagement determines the outcome of NK cell activation (3). Activating receptors recognize ligands that are upregulated by stressed, transformed, or infected cells. MHC class I molecules, expressed at high levels by healthy cells, make up the main group of ligands engaging inhibitory NK cell receptors. In addition to the engagement of NK cell receptors by respective NK cell ligands, microenvironmental signals, such as cytokines, can trigger and modulate NK cell responses. For instance, IL-12 and IL-18 synergistically induce high levels of IFN-γ production by NK cells (4, 5). On the contrary, NK cell function can be suppressed by factors such as TGF-β (6, 7). Microenvironmental factors also can regulate NK cell responses by modulating the levels of activating and inhibitory surface receptors and, thereby, determine the threshold for NK cell activation.

T cell activation also requires integration of at least two signals (8). Signal one is triggered via the TCR by the recognition of antigenic peptides in the context of MHC molecules. Signal two is delivered by the engagement of costimulatory molecules, such as CD28. Its ligands, B7-1 and B7-2, are primarily expressed by APCs and are engaged by CD28, as well as by CTLA-4 (CD152). CTLA-4 is an inhibitory receptor induced following TCR activation and represents a critical regulator of ongoing T cell responses (9, 10). In T cells, most of the CTLA-4 protein resides intracellularly; only a small fraction is exposed on the cell surface where it can engage its ligands. CTLA-4–deficient mice display extensive proliferation and accumulation of self-reactive T cells, resulting in tissue destruction and premature death of animals (11, 12). The hyperactivation of CTLA-4–deficient T cells can be prevented by interruption of the activating CD28:B7-1/B7-2 pathway (13, 14) or by the concomitant presence of CTLA-4–sufficient T cells (1517). It was reported that CTLA-4 exerts its function through both cell-intrinsic and cell-extrinsic mechanisms. In this regard, CTLA-4 was shown to compete with CD28 for their shared ligands, inhibit signal transduction downstream of the TCR, deliver inhibitory signals affecting T cell function, and remove B7-1 and B7-2 from the surface of APCs (1820).

Previous studies (2124) reported that CD28 was expressed by mouse NK cells, and its triggering induced NK cell proliferation, cytotoxicity, and cytokine secretion. In studies by Chiossone et al. (25) and Terme et al. (26), CTLA-4 transcripts were detected by whole-genome microarray analysis of mouse NK cells, indicating that CTLA-4 protein might also be expressed by NK cells. However, the function of CTLA-4 on NK cells remained unknown. In this study, we show that IL-2–activated NK cells express both receptors: CTLA-4 and CD28. We provide further insight into the regulation of their expression by cytokines. Moreover, our data show that CTLA-4 inhibits IFN-γ release by NK cells upon coculture with mature dendritic cells (mDCs). Notably, expression of both CD28 and CTLA-4 was detected in tumor-infiltrating NK cells in several mouse models of solid tumors, suggesting their involvement in NK cell antitumor responses.

Wild-type (WT) C57BL/6N (Charles River Laboratories), WT C57BL/6-Ly5.1, and Rag2- or Ly5.1-Rag2–deficient mice (both bred in the animal facility of the Deutsches Krebsforschungszentrum) were used at the age of 8–16 wk. CD28-deficient mice and respective C57BL/6J WT control mice were purchased from The Jackson Laboratory and also were used at 8–16 wk of age. WT OT-I–transgenic and OT-I–transgenic CTLA-4–deficient mice (both bred in the animal facility of the Otto-von-Guericke University) were used at the age of 4–8 wk. In all experiments, phenotype and responses of CD28- or CTLA-4–deficient NK cells were compared with their respective WT control cells. All mice were housed under specific pathogen–free conditions. For tumor experiments, mice were inoculated s.c. with 106 RMA-S lymphoma, B16 melanoma, or LL/2 lung carcinoma cells. For analysis of NK cells from blood and tumors, mice were sacrificed when the tumor diameter exceeded 1 cm (typically 1.2–1.6 cm on days 12–16 after tumor cell inoculation). All animal experiments were approved by the “Regierungspräsidium Karlsruhe.”

Single-cell suspensions from peripheral blood were prepared after RBC lysis with buffered ammonium chloride potassium phosphate solution (ACK buffer) for 2 min at room temperature. The tumor tissue was cut into small pieces and treated with digestion buffer (PBS with 0.5 mg/ml hyaluronidase V and 0.5 mg/ml DNase; Sigma-Aldrich) for 30 min at 37°C. Subsequently, viable cells were collected after centrifugation over a Lympholyte-M gradient (Cedarlane Laboratories). In experiments comparing the expression of surface molecules on NK cells from blood and tumor, both tumor tissue and single-cell suspensions from blood were treated with digestion buffer.

NK cells were obtained from Rag2-deficient mice by expansion of nonadherent splenocytes in the presence of recombinant human (rh)IL-2 (National Institutes of Health). NK cells were isolated from WT and gene-deficient mice by MACS separation (Miltenyi Biotec) comprising depletion of CD3+ cells, followed by positive selection of DX5+ cells. Cells were maintained in RPMI 1640 medium supplemented with 10% FCS, 2 mM l-glutamine, 100 U/ml penicillin/streptomycin, 1 mM sodium pyruvate, 1% nonessential amino acids, 0.25 mM 2-ME (all from Sigma-Aldrich or Invitrogen), and 1700 U/ml rhIL-2. The purity of NK cell cultures on days 7–10 of expansion was ≥99% CD3NK1.1+. Dendritic cells (DCs) were generated from bone marrow cells in medium (DMEM with 10% FCS, 2 mM l-glutamine, and 100 U/ml penicillin/streptomycin) supplemented with GM-CSF (10% supernatant of GM-CSF–producing X6310 cell line). On day 6, DCs were cultured with 200 ng/ml LPS (Sigma-Aldrich) for 18–24 h to generate mDCs. Cytokines used in NK cell cultures were purchased from PeproTech (IL-15, IL-12, and TGF-β) and MBL (IL-18).

Cells were incubated for 30 min at 4°C with FcR-blocking reagent (10% supernatant of αCD16/CD32-producing hybridoma 2.4G2), followed by incubation with fluorochrome-labeled mAbs. Dead cells were excluded by labeling with 7-aminoactinomycin D (7-AAD), propidium iodide, Zombie Aqua (BioLegend, BD Biosciences), or the LIVE/DEAD Fixable Violet Dead Cell Stain Kit (Molecular Probes). CD28 expression was assessed by cell surface staining using monoclonal anti-CD28 Abs (clone 37.1 and clone E18; BioLegend). For intracellular staining of CTLA-4, cells were stained with mAbs directed against surface Ags, followed by fixation, permeabilization (Foxp3 Staining Buffer Set; eBioscience), and staining with anti–CTLA-4 mAb (clone UC10-4F10-11 from BD Bioscience and clone UC10-4B9 from BioLegend). For detection of CTLA-4 on the cell surface, fluorochrome-labeled anti–CTLA-4 mAb was added to NK cell cultures at 37°C for 4 h before cell harvesting, and cells were analyzed by flow cytometry. FACSCalibur and FACSCanto were used for FACS acquisition, and FACSDiva, FACSAria, and Influx were used for cell sorting. For the preparation of tumor-infiltrating NK cells, C57BL/6-Ly5.1 mice were inoculated with RMA-S tumor cells expressing Ly5.2. Total tumor-infiltrating leukocytes were enriched using MACS separation by positive selection of Ly5.1+ cells. NK cells were sorted as Ly5.1+CD3NK1.1+7-AAD cells by FACS (purity > 98%). Data were analyzed using CellQuest Pro (BD Bioscience) and FlowJo (Tree Star) software.

RNA was isolated from FACS-sorted NK cells (purity ≥ 99%) with the RNeasy Mini Kit (QIAGEN), according to the manufacturer’s instructions. Contaminating DNA was removed using the TURBO DNA-free kit (Ambion Life Technologies). Similar amounts of RNA were used for cDNA synthesis (ProtoScript M-MuLV First Strand Synthesis kit; New England Biolabs). As a negative control, a reaction containing all components with the exception of reverse transcriptase was carried out (−RT control). Quantitative real-time PCR (qPCR) was performed with LightCycler 480 SYBR Green I Master mix, reaction plates, LightCycler 480 instrument, and software (Roche Diagnostics). The reaction mix contained 1× LightCycler 480 SYBR Green I Master mix, 0.5 μM forward primer, and 0.5 μM reverse primer in a total volume of 10 μl. Relative expression of CTLA-4 compared with a housekeeping gene (HPRT) was calculated using the Δ-crossing point method. The following primers were used: CTLA-4-f: 5′-GCTTCCTAGATTACCCCTTCTGC-3′, CTLA-4-r: 5′-CGGGCATGGTTCTGGATCA-3′, HPRT-f: 5′-CTTTGCTGACCTGCTGGATT-3′, and HPRT-r: 5′-TATGTCCCCCGTTGACTGAT-3′.

NK cells were incubated for 8 h with plate-bound control EphA4-IgG or B7-1–IgG fusion protein (both from R&D), coated overnight at 4°C in 50 μl of 2 μg/ml fusion protein solution in PBS, or for 24 h with mDCs (1:2 NK/DC ratio), in the presence of 1700 U/ml rhIL-2. A total of 20 μg/ml control-IgG or CTLA-4–IgG fusion proteins (R&D Systems) were used in blocking experiments. Tumor-infiltrating and blood NK cells were stimulated in the presence of 200 U/ml rhIL-2 for 24 h at 37°C. IL-2–expanded NK cells were stimulated with 50 ng/ml PMA and 750 ng/ml ionomycin (Sigma-Aldrich) for 8 h or with 10 ng/ml PMA and 150 ng/ml ionomycin for 24 h at 37°C in the presence of 1700 U/ml rhIL-2. Levels of IFN-γ in culture supernatants were determined by ELISA (BD Biosciences). For intracellular staining of IFN-γ, a protein-transport inhibitor (GolgiStop; BD Biosciences) was added to the cell culture at 37°C for 4 h before harvesting. IFN-γ was detected (mAb clone XMG1.2 from BioLegend or eBioscience) after cell fixation and permeabilization using the Foxp3 Staining Buffer Set (eBioscience).

Statistical significance was assessed using the Student t test and GraphPad Prism software (GraphPad Software, La Jolla, CA). The differences between groups were considered significant when p ≤ 0.05.

To elucidate expression and regulation of CTLA-4 in mouse NK cells, we first monitored its expression by intracellular staining upon activation with IL-2. We did not detect CTLA-4 expression in freshly isolated splenic NK cells, but culture of NK cells in the presence of IL-2 resulted in an induction of CTLA-4 (Fig. 1A). We also confirmed expression of CTLA-4 mRNA in IL-2–activated, but not freshly isolated NK cells by qPCR (Fig. 1B). Expression of CTLA-4 protein increased over time during culture in IL-2 (Fig. 1C). In parallel, we observed that freshly isolated splenic NK cells expressed very low levels of CD28 (Fig. 1A). In concordance with previously published reports (21, 23, 24), culture in IL-2 led to its upregulation. The increase in CD28 surface expression and CTLA-4 intracellular levels occurred with similar kinetics (Fig. 1C) and was dependent on the dose of IL-2 present in the cultures (Fig. 1D). Of note, IL-2 activation of NK cells not only modulated expression of CD28/CTLA-4, it induced global changes in NK cell phenotype, including downregulation of activating receptors (NKp46, NK1.1, NKG2D), adhesion molecules (CD11b, ICAM-1), and B7-1 and B7-2 (Supplemental Fig. 1). As shown in Fig. 1E, CD28 expression also was upregulated on NK cells in the presence of IL-15, whereas, under our experimental conditions, the presence of IL-15 did not induce expression of CTLA-4.

FIGURE 1.

CD28 and CTLA-4 expression in NK cells is upregulated by IL-2. (A) Freshly isolated and IL-2–expanded splenic NK cells (8 d, 1700 U/ml rhIL-2) were analyzed for surface expression of CD28 by flow cytometry. Expression of CTLA-4 was assessed by intracellular staining. Representative histograms, gated on CD3NK1.1+ cells, are depicted. Bold lines represent staining with specific mAb; shaded histograms depict isotype-control staining. (B) CTLA-4 mRNA expression (relative to the housekeeping gene, HPRT) in FACS-sorted freshly isolated (day 0) and IL-2–expanded (day 7) NK cells was determined by qPCR. Data are mean (n = 2) ± SD, and values are depicted in parentheses. Data were derived from one sort experiment (n = 10 pooled mice). (C) CD28 and CTLA-4 expression was analyzed, as in (A), at the indicated time points after initiation of culture in the presence of IL-2. Representative graphs show mean fluorescence intensity (MFI) or percentage of positive NK cells, with subtracted values of corresponding isotype-control–stained NK cells. (D) NK cell cultures were supplemented with different amounts of IL-2, as indicated. CD28 and CTLA-4 expression was analyzed as in (C). (E) Freshly isolated, IL-15 (50 ng/ml)–expanded and IL-2 (1700 U/ml)–expanded splenic NK cells were analyzed for expression of CD28 and CTLA-4 at the indicated time points, as in (A). Representative graphs show MFI (for CD28) or percentage of NK cells (for CTLA-4) with subtracted values of corresponding isotype-control–stained cells. (F) NK cells were expanded in IL-2 for 7 d. For the detection of CTLA-4 on the cell surface, fluorochrome-labeled anti–CTLA-4 Ab was added to the cell culture 4 h before harvesting (surface staining, 37°C). In parallel, CTLA-4 staining was performed according to intracellular and standard surface staining protocols (4°C). All data are representative of at least two independent experiments.

FIGURE 1.

CD28 and CTLA-4 expression in NK cells is upregulated by IL-2. (A) Freshly isolated and IL-2–expanded splenic NK cells (8 d, 1700 U/ml rhIL-2) were analyzed for surface expression of CD28 by flow cytometry. Expression of CTLA-4 was assessed by intracellular staining. Representative histograms, gated on CD3NK1.1+ cells, are depicted. Bold lines represent staining with specific mAb; shaded histograms depict isotype-control staining. (B) CTLA-4 mRNA expression (relative to the housekeeping gene, HPRT) in FACS-sorted freshly isolated (day 0) and IL-2–expanded (day 7) NK cells was determined by qPCR. Data are mean (n = 2) ± SD, and values are depicted in parentheses. Data were derived from one sort experiment (n = 10 pooled mice). (C) CD28 and CTLA-4 expression was analyzed, as in (A), at the indicated time points after initiation of culture in the presence of IL-2. Representative graphs show mean fluorescence intensity (MFI) or percentage of positive NK cells, with subtracted values of corresponding isotype-control–stained NK cells. (D) NK cell cultures were supplemented with different amounts of IL-2, as indicated. CD28 and CTLA-4 expression was analyzed as in (C). (E) Freshly isolated, IL-15 (50 ng/ml)–expanded and IL-2 (1700 U/ml)–expanded splenic NK cells were analyzed for expression of CD28 and CTLA-4 at the indicated time points, as in (A). Representative graphs show MFI (for CD28) or percentage of NK cells (for CTLA-4) with subtracted values of corresponding isotype-control–stained cells. (F) NK cells were expanded in IL-2 for 7 d. For the detection of CTLA-4 on the cell surface, fluorochrome-labeled anti–CTLA-4 Ab was added to the cell culture 4 h before harvesting (surface staining, 37°C). In parallel, CTLA-4 staining was performed according to intracellular and standard surface staining protocols (4°C). All data are representative of at least two independent experiments.

Close modal

In T cells, surface-exposed CTLA-4 is rapidly internalized and either degraded or recycled back to the cell surface (9). To investigate whether CTLA-4 protein can also be detected on the surface of NK cells, we stained IL-2–activated NK cells with the anti–CTLA-4 mAb at 37°C for 4 h, which enables labeling of surface-exposed CTLA-4 over the time period of incubation with mAb (27). Using this method, very small amounts of CTLA-4 protein were detectable on the cell surface of IL-2–activated NK cells (Fig. 1F).

Next, we aimed to identify additional stimuli regulating CTLA-4 and CD28 protein expression in IL-2–activated NK cells. CTLA-4 expression in NK cells was first observed 48 h after initiation of culture with IL-2 (data not shown). Upon addition of IL-12 and IL-18 to IL-2–supplemented NK cell cultures, a small percentage of CTLA-4–expressing NK cells was already detected after 24 h (Supplemental Fig. 2A). In NK cells expanded for 1 wk in IL-2, addition of IL-12 24 h before harvesting increased surface levels of CD28 and intracellular levels of CTLA-4 (Fig. 2A, 2B). Stimulation with IL-12 and IL-18 resulted in an upregulation of CTLA-4 expression, whereas CD28 was slightly reduced. On the contrary, the presence of TGF-β increased CD28 expression, while leading to a reduction in CTLA-4. Of note, addition of TGF-β significantly reduced CTLA-4 expression in NK cells, even in the presence of IL-12 and IL-18 (Fig. 2A, 2B). Upregulation of CTLA-4 and CD28 was detected 18 h after IL-12 addition and increased over time (Fig. 2C). Similarly, TGF-β–mediated downregulation of CTLA-4 was detected 18 h after cytokine addition to IL-2–supplemented cultures (Fig. 2C). An increase in CD28 and decrease in CTLA-4 expression also were observed in the presence of PMA and ionomycin, which activate protein kinase C and Ca2+ signaling, respectively (Supplemental Fig. 2B). Effects of added cytokines on CD28/CTLA-4 expression also were observed using NK cells generated from Rag2-deficient mice, ruling out the possibility that the observed changes were due to NKT or T cell contamination of our cultures. Taken together, our data show that different cytokines differentially regulate the expression of CD28 and CTLA-4 in IL-2–activated NK cells, which could potentially affect NK cell responses controlled by these receptors.

FIGURE 2.

Cytokine-mediated regulation of CD28 and CTLA-4 expression in IL-2–expanded NK cells. After culture in IL-2–supplemented medium for 6 d, NK cells were cultured for an additional 24 h in the presence of the indicated cytokines (2 ng/ml IL-12, 10 ng/ml IL-18, 2 ng/ml TGF-β). CD28 and CTLA-4 expression on gated CD3NK1.1+ cells was analyzed as in Fig. 1A and 1C. Representative dot plots (A) and quantification of triplicate cultures (B) from one representative experiment are shown. (C) Fold changes in CD28 and CTLA-4 expression after culture with IL-2/IL-12 or IL-2/TGF-β for the indicated time periods were calculated relative to the expression levels detected in cultures supplemented with IL-2. Data are mean (n = 3) ± SEM and are representative of two independent experiments. *p ≤ 0.05, **p ≤ 0.01.

FIGURE 2.

Cytokine-mediated regulation of CD28 and CTLA-4 expression in IL-2–expanded NK cells. After culture in IL-2–supplemented medium for 6 d, NK cells were cultured for an additional 24 h in the presence of the indicated cytokines (2 ng/ml IL-12, 10 ng/ml IL-18, 2 ng/ml TGF-β). CD28 and CTLA-4 expression on gated CD3NK1.1+ cells was analyzed as in Fig. 1A and 1C. Representative dot plots (A) and quantification of triplicate cultures (B) from one representative experiment are shown. (C) Fold changes in CD28 and CTLA-4 expression after culture with IL-2/IL-12 or IL-2/TGF-β for the indicated time periods were calculated relative to the expression levels detected in cultures supplemented with IL-2. Data are mean (n = 3) ± SEM and are representative of two independent experiments. *p ≤ 0.05, **p ≤ 0.01.

Close modal

Previous studies (2123) indicated that CD28 triggering supported NK cell proliferation, cytotoxicity, and cytokine secretion. Because IL-2–activated NK cells expressed both CD28 and CTLA-4, we investigated how these molecules regulate NK cell responses to their shared ligand, B7-1. Although B7-1 and B7-2 were detected on a subpopulation of freshly isolated splenic NK cells, these molecules were downregulated during expansion with IL-2 and were barely detectable on day 6 after initiation of culture (Supplemental Fig. 1B). Thus, IL-2–activated NK cells express CD28/CTLA-4, but, in parallel, B7-1/2 expression is absent/low, suggesting that it is unlikely that these molecules interact on activated NK cells under our experimental conditions. Upon stimulation with plate-bound B7-1–IgG fusion protein (B7-1–Fc), IL-2–expanded NK cells produced increased amounts of IFN-γ (Fig. 3A). As shown in Fig. 3B, high levels of intracellular IFN-γ correlated positively with high CD28 expression. In parallel, higher levels of IFN-γ were detected in CTLA-4low/− NK cells compared with the CTLA-4+ subset. Among IFN-γ–producing CD28+ NK cells, the cells expressing CTLA-4 produced less IFN-γ compared with CTLA-4low/− cells. These data suggest that IFN-γ production in response to B7-1 is positively regulated by CD28 and negatively regulated by CTLA-4. Accordingly, IFN-γ production in response to B7-1–Fc was completely abrogated in CD28-deficient NK cells, indicating that it was mediated by CD28 (Fig. 3C, left panel). We also observed that, in concordance with previously published data (22), B7-1–transduced RMA-S lymphoma cells also induced higher amounts of IFN-γ compared with a vector control–transduced cell line upon coculture with NK cells (Supplemental Fig. 3A). This increase was abrogated by preincubation of tumor cells with a blocking anti–B7-1 mAb or when CD28-deficient NK cells were used, indicating the functional interaction between B7-1 expressed by tumor cells and NK cell–expressed CD28. Of note, B7-1 expression did not influence the sensitivity of RMA-S cells to NK cell killing (Supplemental Fig. 3B). Moreover, CD28-deficient NK cells exerted similar cytotoxic responses to B7-1–expressing B16 cells and B7-1–expressing RMA-S cells, as well as to the prototypic NK cell target cell line, YAC-1 (Supplemental Fig. 3B), compared with WT NK cells.

FIGURE 3.

CD28 and CTLA-4 differentially regulate NK cell IFN-γ release in response to B7-1 and mDCs. (A) NK cells were expanded in the presence of IL-2 and incubated with plate-bound control IgG (c-Fc) or B7-1–IgG fusion protein (B7-1–Fc) for 8 h. IFN-γ production was detected by ELISA or intracellular staining. (B) Dot plots depict CD28 (cell surface staining), CTLA-4, and IFN-γ expression (both intracellular staining) in gated NK1.1+ cells. Histograms show levels of intracellular IFN-γ in the indicated NK cell subpopulations. Bold lines depict IFN-γ production upon B7-1–Fc stimulation, and shaded histograms show IFN-γ upon stimulation with c-Fc. Graph displays the percentage of IFN-γ–expressing NK cells in the indicated NK cell subpopulation upon stimulation with B7-1–Fc. Values detected upon triggering with c-Fc were subtracted. (C and D) NK cells derived from C57BL/6J WT and CD28-deficient (CD28 KO) or OT-I–transgenic WT and CTLA-4–deficient (CTLA-4 KO) mice were expanded in the presence of IL-2. IL-2–expanded NK cells were incubated with plate-bound c-Fc or B7-1–Fc for 8 h (C) or with bone marrow–derived DCs matured overnight with 200 ng/ml LPS (mDCs) for 24 h (D). mDCs were left untreated (NT) or incubated with 20 μg/ml of control IgG (c-Fc) or CTLA-4–Fc for 30 min prior to coculture with NK cells. Levels of IFN-γ released in the supernatant were assessed by ELISA. All graphs show mean (n = 3) ± SEM. Data are representative of two independent experiments. *p ≤ 0.05, **p ≤ 0.01. n.s., not significant.

FIGURE 3.

CD28 and CTLA-4 differentially regulate NK cell IFN-γ release in response to B7-1 and mDCs. (A) NK cells were expanded in the presence of IL-2 and incubated with plate-bound control IgG (c-Fc) or B7-1–IgG fusion protein (B7-1–Fc) for 8 h. IFN-γ production was detected by ELISA or intracellular staining. (B) Dot plots depict CD28 (cell surface staining), CTLA-4, and IFN-γ expression (both intracellular staining) in gated NK1.1+ cells. Histograms show levels of intracellular IFN-γ in the indicated NK cell subpopulations. Bold lines depict IFN-γ production upon B7-1–Fc stimulation, and shaded histograms show IFN-γ upon stimulation with c-Fc. Graph displays the percentage of IFN-γ–expressing NK cells in the indicated NK cell subpopulation upon stimulation with B7-1–Fc. Values detected upon triggering with c-Fc were subtracted. (C and D) NK cells derived from C57BL/6J WT and CD28-deficient (CD28 KO) or OT-I–transgenic WT and CTLA-4–deficient (CTLA-4 KO) mice were expanded in the presence of IL-2. IL-2–expanded NK cells were incubated with plate-bound c-Fc or B7-1–Fc for 8 h (C) or with bone marrow–derived DCs matured overnight with 200 ng/ml LPS (mDCs) for 24 h (D). mDCs were left untreated (NT) or incubated with 20 μg/ml of control IgG (c-Fc) or CTLA-4–Fc for 30 min prior to coculture with NK cells. Levels of IFN-γ released in the supernatant were assessed by ELISA. All graphs show mean (n = 3) ± SEM. Data are representative of two independent experiments. *p ≤ 0.05, **p ≤ 0.01. n.s., not significant.

Close modal

To further investigate the effect of CTLA-4 on NK cell IFN-γ production, IL-2–expanded NK cells derived from WT or CTLA-4–deficient OT-I–transgenic mice were stimulated with plate-bound B7-1–Fc or control Fc. CTLA-4–deficient NK cells released significantly more IFN-γ compared with the respective WT NK cells (Fig. 3C, right panel). Of note, expansion in the presence of IL-2 led to comparable induction of CTLA-4 in NK cells derived from both CD28-deficient and WT mice, and CD28 was upregulated in a similar manner in OT-I–transgenic WT and CTLA-4–deficient NK cells (Supplemental Fig. 4A). In parallel, expression of a panel of activating and inhibitory NK receptors, as well as of NK cell maturation markers, was similar between age-matched gene-deficient and respective WT NK cells (Supplemental Fig. 4B). IL-2–expanded NK cells from respective WT and CD28- or CTLA-4–deficient mice produced comparable amounts of IFN-γ after stimulation with PMA and ionomycin (Supplemental Fig. 4C), implying that, in general, NK cells derived from these mice are able to respond at levels comparable to their respective control WT NK cells. Thus, our results indicate that IFN-γ release by NK cells in response to B7-1 is mediated via CD28 and is negatively regulated by CTLA-4.

B7-1 and B7-2, the ligands of CD28 and CTLA-4, are primarily expressed by APCs, such as DCs. Upon activation, DCs undergo maturation and upregulate expression of both B7-1 and B7-2 (28). mDCs were shown to activate NK cells to secrete IFN-γ (29). Thus, we investigated whether the CD28/CTLA4:B7-1/2 axis contributes to the NK cell/mDC cross-talk. Upon coculture with LPS-matured DCs, IL-2–activated NK cells produced increased amounts of IFN-γ (Fig. 3D). IFN-γ production was partially abrogated by pretreatment of mDCs with CTLA-4–Fc, which binds to the ligands, B7-1 and B7-2, and prevents their interaction with the cognate receptors expressed on NK cells (Fig. 3D, left panel). Furthermore, upon coculture with mDCs, mouse CD28-deficient NK cells released significantly less IFN-γ, whereas CTLA-4–deficient NK cells secreted more IFN-γ compared with WT NK cells (Fig. 3D, middle and right panels). We also determined DC apoptosis in the presence of IL-2–activated NK cells, as measured by annexin V staining. In these experiments, similar frequencies of apoptotic DCs were observed, regardless of whether they were cocultured with CD28-deficient cells or with their respective WT NK cells (data not shown). These data demonstrate that the interaction of CD28, expressed by NK cells, with its ligands on mDCs contributes to the NK cell IFN-γ release induced by mDC, whereas CTLA-4 negatively regulates this response.

We further investigated whether CD28 and CTLA-4 expression in NK cells also was detectable under pathological conditions, such as in malignant disease. For that purpose, we inoculated syngeneic WT BL/6 mice s.c. with 106 RMA-S lymphoma, B16 melanoma, or LL/2 carcinoma cells, which led to progressive tumor growth. Our data demonstrate that a subset of NK cells infiltrating s.c. RMA-S lymphoma expressed CD28 and CTLA-4 (Fig. 4). CTLA-4 also was detected in B16 melanoma– and LL/2 carcinoma–infiltrating NK cells, whereas CD28 expression was mostly low or undetectable. Of note, CTLA-4 expression was detected only in tumor-infiltrating cells, but not in NK cells from blood (Fig. 4) or spleen (data not shown) of tumor-bearing mice. Moreover, our microarray analyses of NK cells from RMA-S tumor–bearing mice revealed elevated CTLA-4 transcript levels in tumor-infiltrating, compared with blood NK cells, indicating transcriptional regulation of CTLA-4 (A. Stojanovic, L. Li, N. Gretz, and A. Cerwenka, unpublished observations). CTLA-4 expression was not detected in NK cells infiltrating small tumors (data not shown), but it was observed in NK cells isolated from tumors ≥10 mm in diameter, suggesting that its expression might correlate with tumor progression. Importantly, NK cells purified from RMA-S tumor tissue responded to stimulation with plate-bound B7-1–Fc with increased IFN-γ production compared with blood NK cells (Fig. 5A). Thus, functional receptors for B7-1 on tumor-infiltrating NK cells exist that might modulate NK cell responses to B7-1–expressing cells in the tumor tissue. Analysis of tumor cells from the established s.c. tumors revealed that B7-1 and B7-2 were not detectable on RMA-S, B16, and LL/2 tumor cells (Fig. 5B). In parallel, higher levels of B7-1 and B7-2 were detected on the myeloid fraction of tumor-infiltrating immune cells, such as the CD11b+F4/80+ and CD11b+CD11c+F4/80 subsets, comprising mainly macrophages and DCs, respectively (Fig. 5B).

FIGURE 4.

CD28 and CTLA-4 are expressed by tumor-infiltrating NK cells. Mice were injected s.c. with 106 RMA-S lymphoma, B16 melanoma, or LL/2 carcinoma cells in 100 μl of PBS. When the tumor diameter exceeded 1 cm, mice were sacrificed, and single-cell suspensions were prepared from blood and tumors. Expression of CD28 (A) and CTLA-4 (B) was analyzed by flow cytometry. Representative dot plots show CD28 and CTLA-4 staining in RMA-S, B16, and LL/2 tumor-infiltrating NK cells, gated on CD3NK1.1+ cells. Bar graphs show quantification of CD28 and CTLA-4 expression (mean ± SEM) of two (RMA-S) or three (B16 and LL/2) animals/group. Data are representative of at least two independent experiments. *p ≤ 0.05, **p ≤ 0.01. n.d., not detectable.

FIGURE 4.

CD28 and CTLA-4 are expressed by tumor-infiltrating NK cells. Mice were injected s.c. with 106 RMA-S lymphoma, B16 melanoma, or LL/2 carcinoma cells in 100 μl of PBS. When the tumor diameter exceeded 1 cm, mice were sacrificed, and single-cell suspensions were prepared from blood and tumors. Expression of CD28 (A) and CTLA-4 (B) was analyzed by flow cytometry. Representative dot plots show CD28 and CTLA-4 staining in RMA-S, B16, and LL/2 tumor-infiltrating NK cells, gated on CD3NK1.1+ cells. Bar graphs show quantification of CD28 and CTLA-4 expression (mean ± SEM) of two (RMA-S) or three (B16 and LL/2) animals/group. Data are representative of at least two independent experiments. *p ≤ 0.05, **p ≤ 0.01. n.d., not detectable.

Close modal
FIGURE 5.

Tumor-infiltrating NK cells respond to B7-1. (A) Blood and tumor-infiltrating NK cells were FACS sorted (≥98% CD3NK1.1+), and sorted cells were incubated with plate-bound control (c-Fc) or B7-1 IgG fusion protein (B7-1–Fc). IFN-γ production was measured by ELISA after 24 h of stimulation. Three independently performed experiments (experiments 1 and 3 with 2 × 104 NK cells used for stimulation, experiment 2 with 5 × 104 NK cells used for stimulation) are depicted. (B) Single-cell suspensions were prepared from tumor tissue of RMA-S, B16, and LL/2 tumor-bearing mice (C57BL/6-Ly5.1). Expression of B7-1 and B7-2 on tumor cells and tumor-infiltrating immune cells was analyzed by flow cytometry. Depicted is the gating strategy (upper panels) and the histograms (lower panel) showing the expression of B7-1 and B7-2 (bold lines) compared with isotype-control staining (shaded histograms) on gated live tumor cells (CD45.1) and live infiltrating immune cells (CD45.1+), such as CD11b+F4/80+ and CD11b+F4/80CD11c+/high.

FIGURE 5.

Tumor-infiltrating NK cells respond to B7-1. (A) Blood and tumor-infiltrating NK cells were FACS sorted (≥98% CD3NK1.1+), and sorted cells were incubated with plate-bound control (c-Fc) or B7-1 IgG fusion protein (B7-1–Fc). IFN-γ production was measured by ELISA after 24 h of stimulation. Three independently performed experiments (experiments 1 and 3 with 2 × 104 NK cells used for stimulation, experiment 2 with 5 × 104 NK cells used for stimulation) are depicted. (B) Single-cell suspensions were prepared from tumor tissue of RMA-S, B16, and LL/2 tumor-bearing mice (C57BL/6-Ly5.1). Expression of B7-1 and B7-2 on tumor cells and tumor-infiltrating immune cells was analyzed by flow cytometry. Depicted is the gating strategy (upper panels) and the histograms (lower panel) showing the expression of B7-1 and B7-2 (bold lines) compared with isotype-control staining (shaded histograms) on gated live tumor cells (CD45.1) and live infiltrating immune cells (CD45.1+), such as CD11b+F4/80+ and CD11b+F4/80CD11c+/high.

Close modal

NK cell activation is the result of the integration of activating and inhibitory signals delivered via activating and inhibitory receptors (3). Many of these receptors and their ligands important for NK cell/tumor cell interaction and NK cell cross-talk with other immune cells have been identified. In this study, we provide evidence that additional receptors, CTLA-4 and CD28, upregulated upon activation of mouse NK cells with cytokines, are involved in the regulation of NK cell function. We show that CD28/CTLA-4 differentially modulated IFN-γ release by IL-2–activated NK cells in response to their ligand, B7-1, and upon interaction with mDCs. Because both CD28/CTLA-4–expressing NK cells and B7-1/2–expressing myeloid cells are present in the tumor tissue of different tumor entities, the NK cell responses mediated via CD28 and CTLA-4 could be of importance during malignant disease and/or therapy regimens based on CD28 and CTLA-4 targeting.

Our in vitro experiments revealed that IL-2 induced expression of CTLA-4 in mouse NK cells, whereas, in concordance with other studies (21, 23, 24), CD28 expression was upregulated. Most importantly, NK cells infiltrating certain solid tumors expressed CTLA-4 and CD28. We observed that tumor-infiltrating NK cells also expressed increased transcript and protein levels of IL-2Rα (CD25) compared with blood NK cells (A. Stojanovic et al., unpublished observations), which might result in their greater responsiveness to IL-2 and lead to CTLA-4 induction and CD28 upregulation. T cells are considered the main producers of IL-2 during ongoing immune responses (30); thus, they are likely to produce IL-2 in tumor tissue. Accordingly, we did not detect CTLA-4 expression in NK cells infiltrating RMA-S tumors in Rag2-deficient mice that lack T cells (data not shown). In concordance with a report by Hunter et al. (24), our study revealed that IL-15, closely related to IL-2 as a member of the common γ-chain cytokine family, induced CD28 upregulation on NK cells. In parallel, under our experimental conditions, we did not detect CTLA-4 expression in NK cells upon IL-15 culture. It is unknown whether IL-15 can modulate CTLA-4 expression in concert with other cytokines or microenvironmental stimuli.

Upon addition of IL-12 and IL-18 to our in vitro cultures, CTLA-4 expression increased further. IL-12 and IL-18 are mainly produced by activated macrophages and DCs and are cytokines shown to be present in the tumor microenvironment (31, 32). Thus, it is possible that IL-12 and IL-18 also might have been involved in the induction of CTLA-4 in tumor-infiltrating NK cells. IL-12 and IL-18, produced by activated DCs, are also potent inducers of IFN-γ production in NK cells. Because our data demonstrate that CTLA-4, expressed by NK cells, negatively regulated IFN-γ production in response to mDCs, the induction of CTLA-4 by IL-12 and IL-18 on NK cells could represent a negative-feedback loop limiting IFN-γ production in response to DC-expressed B7-1 and B7-2. The presence of IL-12 increased CD28 expression in IL-2–activated NK cells, whereas, upon exposure to IL-18, CD28 expression remained unchanged. Of note, addition of the combination of IL-12 and IL-18 did not result in increased CD28 expression, suggesting that signals delivered by IL-18 could counteract upregulation of CD28 mediated by IL-12. Furthermore, CD28 expression might be regulated indirectly by additional soluble factors released upon stimulation with IL-12/18 (33). Addition of TGF-β to in vitro NK cell cultures downregulated IL-2–induced CTLA-4 expression in NK cells. TGF-β was shown to have multiple effects on NK cells, including the downregulation of activating receptors (7) and the inhibition of IFN-γ release induced by IL-12 and IL-18 (6). Thus, the final outcome of NK cell exposure to TGF-β would depend on the signal integration in pathways of NK cell activation (e.g., by cytokines or via activating receptors) and inhibition (e.g., CTLA-4), which are both differentially affected by TGF-β. Our data show that TGF-β reduced CTLA-4 expression in IL-2–expanded NK cells. TGF-β is present in the microenvironment of many tumor entities, and its role in immune suppression is very well documented (3436). Whether TGF-β has a similar effect on tumor-infiltrating NK cells in concert with other microenvironmental factors in the tumor remains unknown. In the tumor tissue, the effect of TGF-β on CTLA-4 expression might depend on its concentration relative to other cytokines that might differentially accumulate during tumor progression. Our experiments revealed that CTLA-4 was detectable on NK cells in tumor tissues, despite the possible presence of TGF-β, and could play an important role in NK cell effector responses and during immunotherapy applying anti–CTLA-4–blocking Abs.

Our data show that RMA-S cells overexpressing B7-1 also induced enhanced levels of IFN-γ production by NK cells compared with vector control–transduced RMA-S cells. This response was abrogated in NK cells derived from CD28-deficient mice. Preincubation of target cells with a blocking anti–B7-1 mAb reduced NK cell IFN-γ production to the levels induced by control cells, indicating that the increased IFN-γ production was dependent on the presence of B7-1 on target cells. However, expression of B7-1 on target cells or the presence of CD28 on NK cells did not influence the cytotoxic response of IL-2–activated NK cells. It is well documented that DCs also can be killed by activated NK cells (3739). Although we observed increased apoptosis of DCs in the presence of IL-2–activated NK cells, this effect was independent of CD28 expression on NK cells (data not shown). Thus, in our experimental system, the CD28:B7-1 axis controls IFN-γ release, but is not involved in NK cell cytolytic responses. Several studies investigated the NK cell responses to B7-1–expressing targets in mice. Although the involvement of CD28 in these responses was addressed in certain studies, the role of CTLA-4 remains undetermined. Kelly et al. (22) reported that NK cell proliferation, IFN-γ production, and cytotoxicity were increased in response to B7-1–expressing RMA-S cells compared with vector control cells. Similarly, P815 B7-1+ cells induced higher levels of IFN-γ in IL-2–expanded NK cells compared with control cell lines (24). Evidence exists that B7-1, expressed by the target cell, can costimulate the responses triggered through other NK cell receptors, such as Ly49D (40). Geldhof et al. (21) showed that CD28 played a crucial role in the cytotoxic response of IL-2/IL-12–expanded NK cells to B7-1+ syngenic, but not B7-1+ allogenic, tumor cells. Finally, Chambers et al. (41) reported that the enhanced lysis of B7-1–expressing RMA-S cells by syngenic NK cells compared with B7-1 RMA-S cells did not depend on CD28 or CTLA-4, implying the existence of an additional receptor for B7-1 on NK cells. Together, these data indicate that the contribution of CD28/CTLA-4 to the NK cell response to B7-1–expressing targets greatly depends on the mode of NK cell activation, expression of other activating/inhibitory ligands, and the type of tumor targets used in the studies. In human NK cells, controversial data generated with different anti-CD28 mAbs were reported with regard to the expression of CD28 (42, 43). Depending on the target cells used, either activation of human NK cells (4446) or the lack of increased responses to B7-expressing targets (47) was observed. Using different anti-CD28 mAbs, we were unable to detect CD28 on human IL-2–activated NK cells, whereas our preliminary data showed that activation with IL-2 increased expression of CTLA-4. In addition, stimulation with IL-12 and/or IL-18 for 24 h further increased CTLA-4 levels in human NK cells (data not shown). These results indicate that expression of CTLA-4 might be regulated by similar mechanisms in mouse and human NK cells. Its function in human NK cells remains to be investigated.

B7-1 is endogenously expressed only by certain tumor cells (4850), but it is often highly expressed by activated APCs. In this context, our data demonstrate that CD28/CTLA-4 differentially modulate IFN-γ release by NK cells in response to mDCs. Addition of a CTLA-4–Fc to DCs, which blocks B7-1 and B7-2 binding to corresponding receptors on NK cells, significantly inhibited NK cell IFN-γ production in response to mDCs. Of note, in our experiments, the reduction in IFN-γ production upon CTLA-4–Fc treatment was incomplete. Thus, it is likely that other factors could be involved in the NK/DC cross-talk. Several cell surface receptors and soluble factors, such as IL-12, were shown to mediate the NK/DC cross-talk in humans and mice (21, 46, 5155). These factors, in addition to CD28/CTLA-4, might also be involved in regulating NK cell IFN-γ production in response to mDCs in our experimental system. Our data demonstrate that both CTLA-4 and CD28 can be expressed by tumor-infiltrating NK cells and that their corresponding ligands were detectable on other tumor-infiltrating immune cells. Several factors might contribute to the regulation of CD28/CTLA-4 expression in tumor-infiltrating NK cells. In this context, we observed that cytokines, such as IL-2, IL-12, IL-18, and TGF-β, which might be present at different levels in the tumor microenvironment, modulated CD28/CTLA-4 expression on NK cells in vitro. Thus, the interplay of these factors and their relative amounts in the tumor microenvironment might determine the relative levels of CD28 and CTLA-4 expression in tumor-infiltrating NK cells and their response to B7-1/2. Moreover, our data indicate that, in certain tumors, CTLA-4 can be expressed in tumor-infiltrating NK cells while CD28 is absent. It is possible that CTLA-4 exerts functions in addition to controlling CD28-dependent responses in NK cells. Furthermore, by acting in a cell-extrinsic manner (20), NK cell–expressed CTLA-4 could influence the responses of other tumor-infiltrating cells, such as DCs and T cells.

Blockade of CTLA-4 by mAbs was shown to enhance T cell effector functions and to delay tumor progression in several experimental tumor models (5659). Our data demonstrate that tumor-infiltrating NK cells also represent potential targets of anti–CTLA-4 mAb-based immunotherapy and, thus, might contribute to orchestrating efficient immune responses upon CTLA-4 blockade. Combinatorial strategies incorporating CTLA-4 blockade and NK cell–based therapies might lead to clinical benefits in the treatment of cancer.

We thank the Deutsches Krebsforschungszentrum Animal Laboratory for animal care and Flow Cytometry Core Facility for cell sorting, Dr. Markus Feuerer and Dr. Jan Hettinger for providing qPCR primers, Dr. Uta E. Höpken and Dr. Michaela Kern for collaboration and helpful discussion, and Dr. Margareta Correia for critical reading of the manuscript and helpful discussion.

This work was supported in part by the Deutsche Krebshilfe (Grant 109174 to A.C.), the Cooperation Program in Cancer Research of the Deutsches Krebsforschungszentrum, Israel’s Ministry of Science and Technology (to A.C.), and the German Research Foundation (SFB 854 TP14 to M.B.-W.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

7-AAD

7-aminoactinomycin D

DC

dendritic cell

mDC

mature DC

qPCR

quantitative real-time PCR

rh

recombinant human

WT

wild-type.

1
Vidal
S. M.
,
Khakoo
S. I.
,
Biron
C. A.
.
2011
.
Natural killer cell responses during viral infections: flexibility and conditioning of innate immunity by experience.
Curr. Opin. Virol.
1
:
497
512
.
2
Ljunggren
H. G.
,
Malmberg
K. J.
.
2007
.
Prospects for the use of NK cells in immunotherapy of human cancer.
Nat. Rev. Immunol.
7
:
329
339
.
3
Long
E. O.
,
Kim
H. S.
,
Liu
D.
,
Peterson
M. E.
,
Rajagopalan
S.
.
2013
.
Controlling natural killer cell responses: integration of signals for activation and inhibition.
Annu. Rev. Immunol.
31
:
227
258
.
4
Lauwerys
B. R.
,
Renauld
J. C.
,
Houssiau
F. A.
.
1999
.
Synergistic proliferation and activation of natural killer cells by interleukin 12 and interleukin 18.
Cytokine
11
:
822
830
.
5
Okamura
H.
,
Kashiwamura
S.
,
Tsutsui
H.
,
Yoshimoto
T.
,
Nakanishi
K.
.
1998
.
Regulation of interferon-gamma production by IL-12 and IL-18.
Curr. Opin. Immunol.
10
:
259
264
.
6
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
.
7
Castriconi
R.
,
Cantoni
C.
,
Della Chiesa
M.
,
Vitale
M.
,
Marcenaro
E.
,
Conte
R.
,
Biassoni
R.
,
Bottino
C.
,
Moretta
L.
,
Moretta
A.
.
2003
.
Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells.
Proc. Natl. Acad. Sci. USA
100
:
4120
4125
.
8
Chen
L.
,
Flies
D. B.
.
2013
.
Molecular mechanisms of T cell co-stimulation and co-inhibition.
Nat. Rev. Immunol.
13
:
227
242
.
9
Rudd
C. E.
,
Taylor
A.
,
Schneider
H.
.
2009
.
CD28 and CTLA-4 coreceptor expression and signal transduction.
Immunol. Rev.
229
:
12
26
.
10
Pentcheva-Hoang
T.
,
Corse
E.
,
Allison
J. P.
.
2009
.
Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections.
Immunol. Rev.
229
:
67
87
.
11
Tivol
E. A.
,
Borriello
F.
,
Schweitzer
A. N.
,
Lynch
W. P.
,
Bluestone
J. A.
,
Sharpe
A. H.
.
1995
.
Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4.
Immunity
3
:
541
547
.
12
Waterhouse
P.
,
Penninger
J. M.
,
Timms
E.
,
Wakeham
A.
,
Shahinian
A.
,
Lee
K. P.
,
Thompson
C. B.
,
Griesser
H.
,
Mak
T. W.
.
1995
.
Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4.
Science
270
:
985
988
.
13
Mandelbrot
D. A.
,
McAdam
A. J.
,
Sharpe
A. H.
.
1999
.
B7-1 or B7-2 is required to produce the lymphoproliferative phenotype in mice lacking cytotoxic T lymphocyte-associated antigen 4 (CTLA-4).
J. Exp. Med.
189
:
435
440
.
14
Tivol
E. A.
,
Boyd
S. D.
,
McKeon
S.
,
Borriello
F.
,
Nickerson
P.
,
Strom
T. B.
,
Sharpe
A. H.
.
1997
.
CTLA4Ig prevents lymphoproliferation and fatal multiorgan tissue destruction in CTLA-4-deficient mice.
J. Immunol.
158
:
5091
5094
.
15
Bachmann
M. F.
,
Köhler
G.
,
Ecabert
B.
,
Mak
T. W.
,
Kopf
M.
.
1999
.
Cutting edge: lymphoproliferative disease in the absence of CTLA-4 is not T cell autonomous.
J. Immunol.
163
:
1128
1131
.
16
Corse
E.
,
Allison
J. P.
.
2012
.
Cutting edge: CTLA-4 on effector T cells inhibits in trans.
J. Immunol.
189
:
1123
1127
.
17
Wang
C. J.
,
Kenefeck
R.
,
Wardzinski
L.
,
Attridge
K.
,
Manzotti
C.
,
Schmidt
E. M.
,
Qureshi
O. S.
,
Sansom
D. M.
,
Walker
L. S.
.
2012
.
Cutting edge: cell-extrinsic immune regulation by CTLA-4 expressed on conventional T cells.
J. Immunol.
189
:
1118
1122
.
18
Qureshi
O. S.
,
Zheng
Y.
,
Nakamura
K.
,
Attridge
K.
,
Manzotti
C.
,
Schmidt
E. M.
,
Baker
J.
,
Jeffery
L. E.
,
Kaur
S.
,
Briggs
Z.
, et al
.
2011
.
Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4.
Science
332
:
600
603
.
19
Bour-Jordan
H.
,
Esensten
J. H.
,
Martinez-Llordella
M.
,
Penaranda
C.
,
Stumpf
M.
,
Bluestone
J. A.
.
2011
.
Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family.
Immunol. Rev.
241
:
180
205
.
20
Wing
K.
,
Yamaguchi
T.
,
Sakaguchi
S.
.
2011
.
Cell-autonomous and -non-autonomous roles of CTLA-4 in immune regulation.
Trends Immunol.
32
:
428
433
.
21
Geldhof
A. B.
,
Moser
M.
,
Lespagnard
L.
,
Thielemans
K.
,
De Baetselier
P.
.
1998
.
Interleukin-12-activated natural killer cells recognize B7 costimulatory molecules on tumor cells and autologous dendritic cells.
Blood
91
:
196
206
.
22
Kelly
J. M.
,
Takeda
K.
,
Darcy
P. K.
,
Yagita
H.
,
Smyth
M. J.
.
2002
.
A role for IFN-gamma in primary and secondary immunity generated by NK cell-sensitive tumor-expressing CD80 in vivo.
J. Immunol.
168
:
4472
4479
.
23
Nandi
D.
,
Gross
J. A.
,
Allison
J. P.
.
1994
.
CD28-mediated costimulation is necessary for optimal proliferation of murine NK cells.
J. Immunol.
152
:
3361
3369
.
24
Hunter
C. A.
,
Ellis-Neyer
L.
,
Gabriel
K. E.
,
Kennedy
M. K.
,
Grabstein
K. H.
,
Linsley
P. S.
,
Remington
J. S.
.
1997
.
The role of the CD28/B7 interaction in the regulation of NK cell responses during infection with Toxoplasma gondii.
J. Immunol.
158
:
2285
2293
.
25
Chiossone
L.
,
Chaix
J.
,
Fuseri
N.
,
Roth
C.
,
Vivier
E.
,
Walzer
T.
.
2009
.
Maturation of mouse NK cells is a 4-stage developmental program.
Blood
113
:
5488
5496
.
26
Terme
M.
,
Ullrich
E.
,
Aymeric
L.
,
Meinhardt
K.
,
Coudert
J. D.
,
Desbois
M.
,
Ghiringhelli
F.
,
Viaud
S.
,
Ryffel
B.
,
Yagita
H.
, et al
.
2012
.
Cancer-induced immunosuppression: IL-18-elicited immunoablative NK cells.
Cancer Res.
72
:
2757
2767
.
27
Alegre
M. L.
,
Noel
P. J.
,
Eisfelder
B. J.
,
Chuang
E.
,
Clark
M. R.
,
Reiner
S. L.
,
Thompson
C. B.
.
1996
.
Regulation of surface and intracellular expression of CTLA4 on mouse T cells.
J. Immunol.
157
:
4762
4770
.
28
Bhatia
S.
,
Edidin
M.
,
Almo
S. C.
,
Nathenson
S. G.
.
2006
.
B7-1 and B7-2: similar costimulatory ligands with different biochemical, oligomeric and signaling properties.
Immunol. Lett.
104
:
70
75
.
29
Fernandez
N. C.
,
Lozier
A.
,
Flament
C.
,
Ricciardi-Castagnoli
P.
,
Bellet
D.
,
Suter
M.
,
Perricaudet
M.
,
Tursz
T.
,
Maraskovsky
E.
,
Zitvogel
L.
.
1999
.
Dendritic cells directly trigger NK cell functions: cross-talk relevant in innate anti-tumor immune responses in vivo.
Nat. Med.
5
:
405
411
.
30
Malek
T. R.
2008
.
The biology of interleukin-2.
Annu. Rev. Immunol.
26
:
453
479
.
31
Arend
W. P.
,
Palmer
G.
,
Gabay
C.
.
2008
.
IL-1, IL-18, and IL-33 families of cytokines.
Immunol. Rev.
223
:
20
38
.
32
Langrish
C. L.
,
McKenzie
B. S.
,
Wilson
N. J.
,
de Waal Malefyt
R.
,
Kastelein
R. A.
,
Cua
D. J.
.
2004
.
IL-12 and IL-23: master regulators of innate and adaptive immunity.
Immunol. Rev.
202
:
96
105
.
33
Brady
J.
,
Carotta
S.
,
Thong
R. P.
,
Chan
C. J.
,
Hayakawa
Y.
,
Smyth
M. J.
,
Nutt
S. L.
.
2010
.
The interactions of multiple cytokines control NK cell maturation.
J. Immunol.
185
:
6679
6688
.
34
Yang
L.
,
Pang
Y.
,
Moses
H. L.
.
2010
.
TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression.
Trends Immunol.
31
:
220
227
.
35
Ikushima
H.
,
Miyazono
K.
.
2010
.
TGFbeta signalling: a complex web in cancer progression.
Nat. Rev. Cancer
10
:
415
424
.
36
Connolly
E. C.
,
Freimuth
J.
,
Akhurst
R. J.
.
2012
.
Complexities of TGF-β targeted cancer therapy.
Int. J. Biol. Sci.
8
:
964
978
.
37
Carbone
E.
,
Terrazzano
G.
,
Ruggiero
G.
,
Zanzi
D.
,
Ottaiano
A.
,
Manzo
C.
,
Kärre
K.
,
Zappacosta
S.
.
1999
.
Recognition of autologous dendritic cells by human NK cells.
Eur. J. Immunol.
29
:
4022
4029
.
38
Piccioli
D.
,
Sbrana
S.
,
Melandri
E.
,
Valiante
N. M.
.
2002
.
Contact-dependent stimulation and inhibition of dendritic cells by natural killer cells.
J. Exp. Med.
195
:
335
341
.
39
Moretta
L.
,
Ferlazzo
G.
,
Bottino
C.
,
Vitale
M.
,
Pende
D.
,
Mingari
M. C.
,
Moretta
A.
.
2006
.
Effector and regulatory events during natural killer-dendritic cell interactions.
Immunol. Rev.
214
:
219
228
.
40
Gao
J. X.
,
Liu
X.
,
Wen
J.
,
Caligiuri
M. A.
,
Stroynowski
I.
,
Zheng
P.
,
Liu
Y.
.
2003
.
Two-signal requirement for activation and effector function of natural killer cell response to allogeneic tumor cells.
Blood
102
:
4456
4463
.
41
Chambers
B. J.
,
Salcedo
M.
,
Ljunggren
H. G.
.
1996
.
Triggering of natural killer cells by the costimulatory molecule CD80 (B7-1)
.
Immunity
5
:
311
317
.
42
Galea-Lauri
J.
,
Darling
D.
,
Gan
S. U.
,
Krivochtchapov
L.
,
Kuiper
M.
,
Gäken
J.
,
Souberbielle
B.
,
Farzaneh
F.
.
1999
.
Expression of a variant of CD28 on a subpopulation of human NK cells: implications for B7-mediated stimulation of NK cells.
J. Immunol.
163
:
62
70
.
43
Goodier
M. R.
,
Londei
M.
.
2004
.
CD28 is not directly involved in the response of human CD3- CD56+ natural killer cells to lipopolysaccharide: a role for T cells.
Immunology
111
:
384
390
.
44
Luque
I.
,
Reyburn
H.
,
Strominger
J. L.
.
2000
.
Expression of the CD80 and CD86 molecules enhances cytotoxicity by human natural killer cells.
Hum. Immunol.
61
:
721
728
.
45
Terrazzano
G.
,
Zanzi
D.
,
Palomba
C.
,
Carbone
E.
,
Grimaldi
S.
,
Pisanti
S.
,
Fontana
S.
,
Zappacosta
S.
,
Ruggiero
G.
.
2002
.
Differential involvement of CD40, CD80, and major histocompatibility complex class I molecules in cytotoxicity induction and interferon-gamma production by human natural killer effectors.
J. Leukoc. Biol.
72
:
305
311
.
46
Terrazzano
G.
,
Pisanti
S.
,
Grimaldi
S.
,
Sica
M.
,
Fontana
S.
,
Carbone
E.
,
Zappacosta
S.
,
Ruggiero
G.
.
2004
.
Interaction between natural killer and dendritic cells: the role of CD40, CD80 and major histocompatibility complex class i molecules in cytotoxicity induction and interferon-gamma production.
Scand. J. Immunol.
59
:
356
362
.
47
Lang
S.
,
Vujanovic
N. L.
,
Wollenberg
B.
,
Whiteside
T. L.
.
1998
.
Absence of B7.1-CD28/CTLA-4-mediated co-stimulation in human NK cells.
Eur. J. Immunol.
28
:
780
786
.
48
Li
J.
,
Yang
Y.
,
Inoue
H.
,
Mori
M.
,
Akiyoshi
T.
.
1996
.
The expression of costimulatory molecules CD80 and CD86 in human carcinoma cell lines: its regulation by interferon gamma and interleukin-10.
Cancer Immunol. Immunother.
43
:
213
219
.
49
Tsukada
N.
,
Aoki
S.
,
Maruyama
S.
,
Kishi
K.
,
Takahashi
M.
,
Aizawa
Y.
.
1997
.
The heterogeneous expression of CD80, CD86 and other adhesion molecules on leukemia and lymphoma cells and their induction by interferon
.
J. Exp. Clin. Cancer Res.
16
:
171
176
.
50
Whiteway
A.
,
Corbett
T.
,
Anderson
R.
,
Macdonald
I.
,
Prentice
H. G.
.
2003
.
Expression of co-stimulatory molecules on acute myeloid leukaemia blasts may effect duration of first remission.
Br. J. Haematol.
120
:
442
451
.
51
Amakata
Y.
,
Fujiyama
Y.
,
Andoh
A.
,
Hodohara
K.
,
Bamba
T.
.
2001
.
Mechanism of NK cell activation induced by coculture with dendritic cells derived from peripheral blood monocytes.
Clin. Exp. Immunol.
124
:
214
222
.
52
Degli-Esposti
M. A.
,
Smyth
M. J.
.
2005
.
Close encounters of different kinds: dendritic cells and NK cells take centre stage.
Nat. Rev. Immunol.
5
:
112
124
.
53
Draghi
M.
,
Pashine
A.
,
Sanjanwala
B.
,
Gendzekhadze
K.
,
Cantoni
C.
,
Cosman
D.
,
Moretta
A.
,
Valiante
N. M.
,
Parham
P.
.
2007
.
NKp46 and NKG2D recognition of infected dendritic cells is necessary for NK cell activation in the human response to influenza infection.
J. Immunol.
178
:
2688
2698
.
54
Ferlazzo
G.
,
Tsang
M. L.
,
Moretta
L.
,
Melioli
G.
,
Steinman
R. M.
,
Münz
C.
.
2002
.
Human dendritic cells activate resting natural killer (NK) cells and are recognized via the NKp30 receptor by activated NK cells.
J. Exp. Med.
195
:
343
351
.
55
Jinushi
M.
,
Takehara
T.
,
Kanto
T.
,
Tatsumi
T.
,
Groh
V.
,
Spies
T.
,
Miyagi
T.
,
Suzuki
T.
,
Sasaki
Y.
,
Hayashi
N.
.
2003
.
Critical role of MHC class I-related chain A and B expression on IFN-alpha-stimulated dendritic cells in NK cell activation: impairment in chronic hepatitis C virus infection.
J. Immunol.
170
:
1249
1256
.
56
Demaria
S.
,
Kawashima
N.
,
Yang
A. M.
,
Devitt
M. L.
,
Babb
J. S.
,
Allison
J. P.
,
Formenti
S. C.
.
2005
.
Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer.
Clin. Cancer Res.
11
:
728
734
.
57
Hurwitz
A. A.
,
Foster
B. A.
,
Kwon
E. D.
,
Truong
T.
,
Choi
E. M.
,
Greenberg
N. M.
,
Burg
M. B.
,
Allison
J. P.
.
2000
.
Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade.
Cancer Res.
60
:
2444
2448
.
58
Leach
D. R.
,
Krummel
M. F.
,
Allison
J. P.
.
1996
.
Enhancement of antitumor immunity by CTLA-4 blockade.
Science
271
:
1734
1736
.
59
van Elsas
A.
,
Hurwitz
A. A.
,
Allison
J. P.
.
1999
.
Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation.
J. Exp. Med.
190
:
355
366
.

The authors have no conflicts of interest.

Supplementary data