Tyrosine kinase 2 (TYK2) is a widely expressed receptor-associated kinase that is involved in signaling by a variety of cytokines with important immune regulatory activities. Absence of TYK2 in mice results in impaired NK cell maturation and antitumor activity, although underlying mechanisms are largely unknown. Using conditional ablation of TYK2 in NK cells we show that TYK2 is required for IFN-γ production by NK cells in response to IL-12 and for an efficient immune defense against Listeria monocytogenes. Deletion of TYK2 in NK cells did not impact NK cell maturation and IFN-γ production upon NK cell activating receptor (actR) stimulation. Similarly, NK cell–mediated tumor surveillance was unimpaired upon deletion of TYK2 in NK cells only. In line with the previously reported maturation-associated Ifng promoter demethylation, the less mature phenotype of Tyk2−/− NK cells correlated with an increased CpG methylation at the Ifng locus. Treatment with the DNA hypomethylating agent 5-aza-2-deoxycytidine restored the ability of Tyk2−/− NK cells to produce IFN-γ upon actR but not upon IL-12 stimulation. NK cell maturation was dependent on the presence of TYK2 in dendritic cells and could be rescued in Tyk2-deficient mice by treatment with exogenous IL-15/IL-15Rα complexes. IL-15 treatment also rescued the in vitro cytotoxicity defect and the impaired actR-induced IFN-γ production of Tyk2−/− NK cells. Collectively, our findings provide the first evidence, to our knowledge, for a key role of TYK2 in the host environment in promoting NK cell maturation and antitumor activity.

This article is featured in In This Issue, p.1647

Natural killer cells are effector lymphocytes of the innate immune system and are characterized by their strong cytotoxic activity against infected and transformed cells. NK cell effector functions are tightly regulated by several mechanisms, including activating and inhibitory NK cell receptor and cytokine signaling (1). Most of the cytokines that act on NK cells signal through the JAK/STAT pathway (2). All STAT family members positively or negatively regulate NK cell activities, although underlying mechanisms are just beginning to emerge (3). Little is known about the impact of the individual JAK family members (JAK1-3 and tyrosine kinase 2 [TYK2]). Jak1−/− and Jak2−/− mice die soon after birth and during embryonic development, respectively (46). Conditional deletion of JAK2 in adult mice revealed a critical role of JAK2 in the maintenance of peripheral NK cell numbers and their maturation state (7). Treatment of mice with the JAK2-specific inhibitor BSK805 or the JAK1/JAK2 inhibitor ruxolitinib mimics NK cell defects upon conditional deletion of JAK2 and results in accelerated metastasis of transplanted breast cancer cells (7). Ruxolitinib treatment of patients suffering from myeloproliferative neoplasms impairs NK cell proliferation, maturation, and cytolytic capacity (8). Jak3−/− mice and mice with a Jak3 loss-of-function mutation fail to develop NK cells (911), a phenotype that is recapitulated in patients bearing Jak3 mutations (12, 13). NK cells from Tyk2−/− mice fail to produce IFN-γ in response to IL-12 and/or IL-18 and have an impaired early control of Leishmania major infections (14, 15). Defective IFN-γ production by NK cells in response to IL-12/IL-18 cotreatment has been described in TYK2-deficient patients (16), albeit with one exception (17). TYK2 is also involved in signaling by type I IFNs, IL-23, and IL-10, which directly act on NK cells but also on other cell types that might affect NK cell development and activity, such as dendritic cells (DCs), macrophages, or stromal cells (18). The first evidence for a role of TYK2 in NK cell–mediated tumor surveillance was provided by studies using the Abelson murine leukemia virus–induced B cell tumor model (19), which was later confirmed with tumor transplants (20). NK cells from Tyk2−/− mice show reduced maturation and cytotoxicity and produce considerably less IFN-γ upon NK cell activating receptor (actR) stimulation than wild-type (WT) NK cells, although the underlying mechanisms remain unclear (19, 20). JAK/STAT signaling components are frequently altered in cancers, and considerable effort is directed toward the development of specific inhibitors for tumor therapy (2124). TYK2 inhibitors are considered for the treatment of inflammatory diseases and tumor therapy (2532). However, to realize their full potential and to identify harmful side effects, a better understanding of how TYK2 regulates NK cell function and antitumor activity is required.

In this study, we made use of mice that lack TYK2 selectively in NK cells to discriminate between direct and indirect effects of TYK2 on NK cell functionality. We demonstrate that TYK2 in CD11c+ and other non–NK cells regulates NK cell maturation, target cell cytotoxicity, and tumor surveillance, whereas TYK2 in NK cells is required for IL-12 signaling and an efficient immune defense against the intracellular bacterium Listeria monocytogenes. We furthermore provide evidence that NK cell–extrinsic, TYK2-dependent signals increase the competence of NK cells to produce high amounts of IFN-γ in response to actR stimulation and provide, to our knowledge, the first indication for an involvement of TYK2 in Ifng promoter demethylation.

All animal experiments were approved by the Ethics and Animal Welfare Committee of the University of Veterinary Medicine Vienna and the national authority (Austrian Federal Ministry of Science and Research) according to §§ 26ff. of Animal Experiments Act, Tierversuchsgesetz 2012: TVG 2012 (BMWF-68.205/0218-II/3b/2012, BMWFW-68.205/0032-WF/II/3b/2014, BMWFW-68.205/0103-WF/V/3b/2015, BMWFW-68.205/0212-WF/V/3b/2016).

WT, C57BL/6N mice were purchased from Janvier Labs. Tyk2−/− (B6.129P2-Tyk2tm1Biat and B6.129P2-Tyk2tm1.1Biat), Tyk2fl/fl (B6.129S2-Tyk2tm1(fl/fl)Biat), Tyk2ΔDC (B6.129P2-Tyk2tm1[fl/fl] Biat-Tg[Itgax-cre]1.1Reiz), and Tyk2ΔM (B6.129P2-Tyk2tm[fl/fl]Biat-Lyz2tm[cre]Ifo/J) mice were described previously (33, 34). To generate mice that lack TYK2 in NK cells (Tyk2ΔNK), Tyk2fl/fl mice were crossed to Ncr1-iCre–Tg (7BL/6N-Tg[Ncr1-icre]265Sxl) mice (35). B6.SJL-Ptprca (Ly5.1) mice were purchased from Charles River Laboratories, β2-microglobulin−/− (2m−/−]; B6.129-B2mtm1Jae) mice (36) were kindly provided by Wilfried Ellmeier (Medical University of Vienna, Austria). All mice were on C57BL/6 background and bred at University of Veterinary Medicine Vienna under specific pathogen-free conditions. Age- and sex-matched (6–12 wk) mice were used for all experiments. For in vivo experiments, male mice (age 8–12 wk) were used. Murine lymphoma cell lines RMA-S (37) and RMA-Rae1 (38) were cultured in RPMI 1640 complete medium (R8758; Sigma-Aldrich) supplemented with 10% heat-inactivated FCS (Invitrogen), 100 U/ml penicillin, 100 μg/ml streptomycin (both from Sigma-Aldrich), and 50 μM 2-ME (Life Technologies).

A total of 1 × 106 RMA-Rae1 tumor cells were s.c. injected into both flanks of mice depilated 3–4 d prior to tumor cell injection. Tumor growth was monitored every other day. After 10–13 d, the body and tumor weight were assessed.

Mice were infected i.p. with 5 × 105 CFU L. monocytogenes strain EGD in 200 μl of PBS or were mock infected with PBS. Survival of mice was monitored for 2 wk. To determine bacterial burden, spleens and livers were harvested on day 5 postinfection (p.i.) and homogenized in PBS. Serial dilutions of homogenates were plated on Oxford agar plates (Biolife), and colonies were counted after 48 h growth at 37°C.

WT and Tyk2−/− mice were injected i.p. with recombinant murine (rm) IL-15 and IL-15Rα–Fc (both R&D Systems), which were preincubated for complex formation, as previously described (39), or PBS as a control. Injections were given every 2–3 d for 2 wk (four doses). Two days after the last injection, splenic NK cells were analyzed for the expression of maturation markers, or isolated splenocytes were analyzed for IFN-γ in response to anti-NK1.1 Ab stimulation as described below.

NK cells from in vitro cultures and splenic single-cell suspensions were stained with the following Abs (all from eBioscience) against: CD16/CD32 (clone 93), CD49b (DX5), NK1.1 (PK136), NKp46 (29A1.4) CD3ε (145-2C11), CD3 (17A2), TCRβ (H57-597), CD8a (53-6.7), CD11c (N418), KLRG1 (2F1), CD27 (LG.7F9), CD11b (M1/70), MHC class II (M5/114.15.2), Ly6G (1A8), Ly6C (HK1.4), F4/80 (BM8), IFN-γ (XMG1.2), Ly5.2 (clone 104), and T-bet (eBio4B10). Biotinylated Ab to IL-15Rα and the isotype control were purchased from R&D systems. Intracellular T-bet and IFN-γ levels were analyzed using Foxp3/Transcription Factor Staining Buffer Set (eBioscience) according to manufacturer’s instructions. Analyses were performed on a FACSCanto II (BD Biosciences) and analyzed using BD FACSDiva software version 8.0 or on a CytoFLEX (Beckman Coulter) and analyzed using CytExpert version 2.2.0.97.

Ly5.1 recipient mice were lethally irradiated (9 Gy) 24 h prior to transplantation. Bone marrow (BM) of donor WT, Tyk2−/−, and Ly5.1 mice was isolated and depleted of mature T and NK cells using CD5- and DX5-labeled MACS beads (Miltenyi Biotec) according to the manufacturer’s instructions. A total of 4 × 106 of WT or Tyk2−/− BM cells were mixed at a 1:1 ratio with Ly5.1 BM cells and i.v. injected into Ly5.1 recipient mice. Spleens of recipient Ly5.1 animals were isolated 6 wk after receiving the donor BM and NK cell maturation, and T-bet levels were analyzed by flow cytometry.

NK cells were isolated from splenic single-cell suspensions from Tyk2fl/fl, Tyk2ΔNK, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− mice using DX5-labeled MACS beads according to the manufacturer’s instructions (Miltenyi Biotec). NK cells were expanded in RPMI 1640 complete medium supplemented with 5000 U/ml IL-2 (Proleukin; Novartis) or 100 ng/ml IL-15 (eBioscience) for 3–7 d. Purity of NK cells was assessed by flow cytometry and was typically 70–85% CD3εNK1.1+ cells of all living cells. Expanded NK cells were stimulated with 5 ng/ml recombinant mouse IL-12 (R&D Systems) for 10 min and used for Western blot analysis. For the analysis of actR-induced IFN-γ production, test tubes were precoated with 10 μg/ml of anti-NK1.1 Ab (PK136) (BioLegend or purified from the HB-191 hybridoma cell line) 1 d prior to the experiment. Splenocytes or cultured NK cells were seeded at 5 × 106 or 5 × 105 cells per tube, respectively, in RPMI 1640 complete medium. For the analysis of IL-12–induced IFN-γ production, 5 × 106 splenocytes or 5 × 105 cultured NK cells were seeded on uncoated tubes in RPMI 1640 complete medium with 5 ng/ml IL-12 (R&D Systems) and 5000 U/ml IL-2. Brefeldin A (eBioscience) was added after 1 h of incubation at 37°C. After additional 4 h of incubation, cells were stained for CD3ε, DX5, NKp46, and IFN-γ, and the frequency of IFN-γ+ NK cells (CD3εDX5+ or CD3εNKp46+) was assessed by flow cytometry. For the analysis of IL-15Rα surface-levels, spleens were digested by collagenase D (1 mg/ml; Roche Applied Sciences) and DNase I treatment (20 μg/ml; Roche Applied Sciences).

In vitro cytotoxicity assays were performed as previously described (40) using RMA-S or RMA-Rae1 as target cells. For in vivo cytotoxicity assays, WT and β2m−/− splenocytes were isolated, stained with different concentrations of CFSE, and mixed at a 1:1 ratio (input sample). Recipient mice were i.v. injected with input cells (1 × 107 cells per mouse), and the spleens of recipient mice were isolated after 16 h. Specific lysis was assessed by flow cytometry by the analysis of the β2m−/− (CFSEhigh): WT (CFSElow) cell ratio in input sample and splenocytes of recipient mice.

RNA from DX5-depleted splenocytes (flow) and DX5 MACS-purified NK cells was isolated using peqGOLD TriFast reagent (PEQLAB), and reverse transcription was performed using iScript First Strand cDNA Synthesis Kit (Bio-Rad). The quantitative PCR was performed with a Stratagene MX3000 instrument (Agilent Technology, Boebelingen, Germany) as described previously (41) using Ube2d2 as housekeeping gene. The following primers were used: Ube2d2-forward 5′-AGG TCC TGT TGG AGA TGA TAT GTT-3′, Ube2d2-reverse 5′-TTGGGAAATGAATTG TCA AGA AA-3′, Ube2d2-probe 5′-CCA AAT GAC AGC CCC TAT CAG GGT GG-3′, Tyk2-forward 5′-GGG TCA CCT TCA GCC AGA CA-3′, Tyk2-revers 5′-GAC CTT AGC CTG TGC ATT GTA GAG T-3′, Tyk2-probe 5′-CAC ATC GCA CAC AAA GTC GGC ATC A-3′.

Cell lysis, SDS-PAGE, and Western blots were performed as described previously (41). The detection of chemiluminescence was performed using ChemiDoc Touch Imaging System (Bio-Rad) and analyzed by Image Lab software version 5.2 (Bio-Rad). For the analysis of TYK2, cells were lysed in 1× Laemmli sample buffer. The following Abs were used: anti–phospho-STAT3 (Tyr705, no. 9131), anti-STAT3 (no. 9132S), and anti-STAT4 (clone C46B10, no. 2653) from Cell Signaling Technology; anti–phospo-STAT4 (Tyr693, clone 38/pSTAT4) and anti-panERK (clone 16/ERK) from BD Transduction Laboratories. The anti-TYK2 Ab was custom made and described previously (41).

NK cells were DX5 MACS purified and FACS sorted (CD3εNK1.1+) using FACS Aria II (BD Biosciences). Genomic DNA was isolated using the QIAamp DNA Mini Kit (Qiagen). The DNA methylation status of the Ifng locus was analyzed by Varionostic GmbH, Germany, using direct pyrosequencing. The following primers were used (see Fig. 6C): 400 bp Ifng promoter fragment: forward: 5′-AGAGAATTTTATAAGAATGGTATAGGTG-3′, reverse: 5′-Biotin-CATAAAAAAAACTACAAAACCAAAATACA-3′; sequencing primer (S)1 (for CpGs −212, −198 and −178): 5′-ATGGTATAGGTGGGTA-3′; S2 (for CpGs −58, −50, −39, and +12): 5′-AAAAAAAATTAAAAAAAAATTTGTG-3′; S3 (for CpGs +91 and +114): 5′-ATGGTATAGGTGGGTA-3′. Methylation levels were analyzed with ProMarkQ24 software. Annotation of the transcriptional start site (TSS) and the translational start site is according to Ensemble genome browser (42) release 90 (ENSMUST00000068592.4) and consistent with a previous report (43).

DX5 MACS-purified IL-2–expanded Tyk2−/− and Tyk2ΔNK NK cells were treated with 5-aza-2-deoxycytidine (5-Aza) (Sigma-Aldrich) at 10 μM daily for 72 h with an untreated control, as described before (44). Cells were stimulated with anti-NK1.1 or IL-12 as described above, and frequency of IFN-γ+ NK cells was assessed by flow cytometry. Cell viability was determined with flow cytometry using the LIVE/DEAD fixable dye eFluor 780 (eBioscience).

One-way ANOVA with Bonferroni post hoc test, two-way ANOVA with Tukey post hoc test, unpaired t test, or log-rank (Mantel–Cox) test (for Kaplan–Meier plot) were performed using GraphPad Prism version 7.0 for Mac (GraphPad Software). Statistical significances are indicated for each experiment (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001).

To analyze NK cell–intrinsic function of TYK2, we generated mice with an NK cell–specific deletion of TYK2 (Tyk2ΔNK) by crossing Tyk2fl/fl mice (34) with Ncr1-iCre–Tg mice (35). Deletion of TYK2 in NK cells from Tyk2ΔNK mice was analyzed at both protein and mRNA level in MACS-enriched NK cells (Fig. 1A, 1B). TYK2 protein was barely detectable in NK cells from Tyk2ΔNK mice (Fig. 1A), and mRNA levels were reduced to around 15% compared with NK cells from Tyk2fl/fl mice (Fig. 1B). NK cells develop in the BM from NK precursors to immature NK cells and mature NK cells before migrating to the periphery where they undergo full maturation and acquire functional competence (4547). Four distinct NK cell maturation stages are defined by the cell surface markers CD27 and CD11b: CD27CD11b, CD27+ CD11b, CD27+ CD11b+, and CD27CD11b+ (from least to most mature NK cells). Complete TYK2 deficiency does not affect NK cell development and peripheral NK cell numbers but results in a reduced frequency of the most mature NK cell population (20). We did not observe differences in the maturation of NK cells from Tyk2ΔNK and Tyk2fl/fl mice (Fig. 1C). In line with this, NK cells from Tyk2ΔNK mice had similar levels of the inhibitory lectin-like receptor KLRG1 as NK cells from Tyk2fl/fl mice (Fig. 1D, Supplemental Fig. 1A) and only modestly reduced levels of the transcription factor T-bet (Fig. 1E), both of which are highly expressed in the most mature NK cell population (48). Similar to Tyk2−/− mice (20), Tyk2ΔNK mice did not differ from control mice with respect to the total abundance of splenic NK cells (Supplemental Fig. 1B).

FIGURE 1.

NK cells from Tyk2ΔNK mice fully mature. (A and B) DX5 MACS-sorted NK cells from Tyk2fl/fl and Tyk2ΔNK mice and NK cell–depleted splenocytes (flow) were used to analyze the deletion efficiency of TYK2. Directly after isolation (flow) or after 7-d expansion of NK cells in the presence of IL-2, (A) TYK2 protein levels were assessed by Western blot, or (B) RNA was isolated and Tyk2 mRNA levels were assessed by reverse transcription quantitative PCR. Tyk2 mRNA expression levels were normalized to the housekeeping gene Ube2d2 and calculated relative to Tyk2fl/fl cells (set to 1). (A) One out of two independent experiments and (B) mean values ± SEM (n = 2–4 per genotype) are shown. (CE) Splenocytes from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were isolated and analyzed by flow cytometry for levels of (C) the maturation markers CD27 and CD11b, (D) KLRG1, and (E) T-bet in CD3εNK1.1+ cells. Mean percentages/MFIs ± SEM from two to three independent experiments are shown (n = 5–9 per genotype). *p < 0.05, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.

FIGURE 1.

NK cells from Tyk2ΔNK mice fully mature. (A and B) DX5 MACS-sorted NK cells from Tyk2fl/fl and Tyk2ΔNK mice and NK cell–depleted splenocytes (flow) were used to analyze the deletion efficiency of TYK2. Directly after isolation (flow) or after 7-d expansion of NK cells in the presence of IL-2, (A) TYK2 protein levels were assessed by Western blot, or (B) RNA was isolated and Tyk2 mRNA levels were assessed by reverse transcription quantitative PCR. Tyk2 mRNA expression levels were normalized to the housekeeping gene Ube2d2 and calculated relative to Tyk2fl/fl cells (set to 1). (A) One out of two independent experiments and (B) mean values ± SEM (n = 2–4 per genotype) are shown. (CE) Splenocytes from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were isolated and analyzed by flow cytometry for levels of (C) the maturation markers CD27 and CD11b, (D) KLRG1, and (E) T-bet in CD3εNK1.1+ cells. Mean percentages/MFIs ± SEM from two to three independent experiments are shown (n = 5–9 per genotype). *p < 0.05, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.

Close modal

Ncr1-driven Cre recombinase expression starts at the immature NK cell stage (LinCD122+DX5NK1.1+) and remains thereafter (35). To exclude the possibility that TYK2-dependent signals prior to Cre recombinase–mediated deletion impact peripheral NK cell maturation, we performed mixed BM chimera experiments. WT or Tyk2−/− Ly5.2 BM cells were mixed at a 1:1 ratio with WT Ly5.1 BM and adoptively transferred into lethally irradiated Ly5.1 mice. Splenic NK cell maturation was analyzed before (input) and 6 wk after transplantation (Fig. 2A). Tyk2−/− BM cells repopulated with the same efficiency as WT cells (Supplemental Fig. 2A). Transfer of Tyk2−/− BM into WT mice restored the frequency of Tyk2−/− CD27CD11b+ and KLRG1+ NK cells (Fig. 2B, 2C, Supplemental Fig. 2B) and T-bet levels in Tyk2−/− NK cells to those observed in WT NK cells (Fig. 2D). Thus, TYK2 in the environment rather than in NK cells themselves promotes NK cell maturation.

FIGURE 2.

TYK2-deficient NK cells fully mature in a TYK2-sufficient surrounding. (A) Scheme representing the experimental design of BM chimeras. (BD) BM from WT or Tyk2−/− mice was adoptively transferred into lethally irradiated Ly5.1 mice. Splenocytes of donor (input) and recipient mice (after 6 wk in BM chimera) were analyzed by flow cytometry for maturation markers (B) CD27, CD11b, and (C) KLRG1 and (D) T-bet in Ly5.2+ CD3εNK1.1+ NK cells. Mean percentages/MFI ± SEM from two independent experiments are shown (n = 4–5 per genotype). *p < 0.05, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.

FIGURE 2.

TYK2-deficient NK cells fully mature in a TYK2-sufficient surrounding. (A) Scheme representing the experimental design of BM chimeras. (BD) BM from WT or Tyk2−/− mice was adoptively transferred into lethally irradiated Ly5.1 mice. Splenocytes of donor (input) and recipient mice (after 6 wk in BM chimera) were analyzed by flow cytometry for maturation markers (B) CD27, CD11b, and (C) KLRG1 and (D) T-bet in Ly5.2+ CD3εNK1.1+ NK cells. Mean percentages/MFI ± SEM from two independent experiments are shown (n = 4–5 per genotype). *p < 0.05, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.

Close modal

Next, we sought to determine which cell type requires TYK2 to enable full NK cell maturation. Macrophages/monocytes (4951), CD11c+ DCs (52), and neutrophils (53) have been implicated in the regulation of NK cell maturation. Hence, we tested a potential role of TYK2 in these cell types by using mice that lack TYK2 in LysM-expressing cells (i.e., monocytes, macrophages, and neutrophils [Tyk2ΔM]) or in CD11c+ cells (i.e., mainly DCs [Tyk2ΔDC]) (34). NK cells from Tyk2ΔM mice did not display significant differences as compared with those derived from Tyk2fl/fl mice (Fig. 3A, 3B). In contrast, the frequency of CD27CD11b+ (Fig. 3A) and KLRG1+ (Fig. 3B) NK cells was significantly reduced in Tyk2ΔDC mice, albeit to a lesser extent than in Tyk2−/− mice. NK cell homeostasis and maturation critically depends on IL-15 (54, 55). As it has long been suggested that accessory cells use IL-15Rα to trans-present IL-15 to developing NK cells (50, 51, 56), we analyzed IL-15Rα surface levels in Tyk2fl/fl, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− mice. IL-15Rα levels were strongly reduced on macrophages and DCs but not on monocytes, NK cells, and CD8+ T cells from Tyk2−/− compared with Tyk2fl/fl mice (Fig. 3C–E, Supplemental Fig. 3A–D). DCs from Tyk2ΔDC mice had a similar reduction in IL-15Rα levels as Tyk2−/− mice, suggesting that DC-intrinsic TYK2 promotes IL-15Rα surface expression (Fig. 3C, Supplemental Fig. 3A, 3B). In contrast, macrophages from Tyk2ΔM mice had similar IL-15Rα surface levels as Tyk2fl/fl mice, suggesting that cell-extrinsic TYK2 signaling regulates IL-15Rα on macrophages (Fig. 3E). Having established that Tyk2−/− mice show reduced IL-15Rα surface levels on macrophages and DCs, we next tested whether treatment with IL-15 rescues the NK cell maturation defects in Tyk2−/− mice. Sustained treatment of Tyk2−/− mice with IL-15/IL-15Rα complexes restored the abundance of both CD27CD11b+ (Fig. 3F) and KLRG1+ NK cells to WT levels (Fig. 3G). Taken together, these data indicate that TYK2 in DCs and other cell types drives NK cell maturation and that this function of TYK2 can be compensated for by IL-15/IL-15Rα treatment.

FIGURE 3.

TYK2 in CD11c+ cells is required for full NK cell maturation, and IL-15 treatment of Tyk2−/− mice rescues NK cell maturation (A and B) Splenocytes from Tyk2fl/fl, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− were isolated, and NK cells (CD3εNK1.1+) were analyzed for maturation markers (A) CD27, CD11b, and (B) KLRG1. Mean values ± SEM from two independent experiments per genotype are shown (n = 7–14, Tyk2fl/fl are combined littermate controls from Tyk2ΔM and Tyk2ΔDC mice). (CE) Splenocytes from Tyk2fl/fl, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− mice were isolated, and IL-15Rα levels determined by flow cytometry on DCs (CD11c+MHC class IIhigh), monocytes (CD11cNK1.1Ly6GLy6C+), and macrophages (CD11cNK1.1Ly6GF4/80+). Median fluorescence intensity (MdFI) ± SEM from two to three independent experiments (n = 5–10 per genotype) are shown. (F and G) WT and Tyk2−/− mice were treated with rmIL-15/IL-15Rα–Fc complexes every 2–3 d, and after four injections the expression of (F) CD27, CD11b, and (G) KLRG1 on CD3εNK1.1+ cells was assessed. Mean values ± SEM from two independent experiments are shown (n = 7 per genotype). For simplicity, significances are only given for the comparison between Tyk2fl/fl and the other genotypes in (A) and (B). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 3.

TYK2 in CD11c+ cells is required for full NK cell maturation, and IL-15 treatment of Tyk2−/− mice rescues NK cell maturation (A and B) Splenocytes from Tyk2fl/fl, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− were isolated, and NK cells (CD3εNK1.1+) were analyzed for maturation markers (A) CD27, CD11b, and (B) KLRG1. Mean values ± SEM from two independent experiments per genotype are shown (n = 7–14, Tyk2fl/fl are combined littermate controls from Tyk2ΔM and Tyk2ΔDC mice). (CE) Splenocytes from Tyk2fl/fl, Tyk2ΔM, Tyk2ΔDC, and Tyk2−/− mice were isolated, and IL-15Rα levels determined by flow cytometry on DCs (CD11c+MHC class IIhigh), monocytes (CD11cNK1.1Ly6GLy6C+), and macrophages (CD11cNK1.1Ly6GF4/80+). Median fluorescence intensity (MdFI) ± SEM from two to three independent experiments (n = 5–10 per genotype) are shown. (F and G) WT and Tyk2−/− mice were treated with rmIL-15/IL-15Rα–Fc complexes every 2–3 d, and after four injections the expression of (F) CD27, CD11b, and (G) KLRG1 on CD3εNK1.1+ cells was assessed. Mean values ± SEM from two independent experiments are shown (n = 7 per genotype). For simplicity, significances are only given for the comparison between Tyk2fl/fl and the other genotypes in (A) and (B). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Absence of TYK2 in humans and mice results in impaired IL-12 signaling and IFN-γ production in NK cells and T cells (16, 18). In vitro expanded NK cells from Tyk2ΔNK mice showed strongly reduced phosphorylation of STAT3 and STAT4 in response to IL-12 compared with NK cells from Tyk2fl/fl mice (Fig. 4A). The defect was less pronounced than in NK cells from Tyk2−/− mice (Fig. 4A), which most likely reflects the presence of minor amounts of non–NK cells in MACS-purified and IL-2–expanded NK cell cultures. Ex vivo treatment of NK cells with IL-12 showed a similar impairment of IFN-γ production of NK cells from Tyk2ΔNK and Tyk2−/− mice compared with NK cells from Tyk2fl/fl mice (Fig. 4B), confirming that IL-12 signaling requires TYK2 in NK cells themselves. To test IL-12 responsiveness of NK cells in vivo, we infected mice with L. monocytogenes and analyzed IFN-γ production at day 3 p.i. The frequency of splenic IFN-γ–producing NK cells (CD3εNK1.1+NKp46+) was profoundly reduced in Tyk2ΔNK and Tyk2−/− compared with Tyk2fl/fl mice (Fig. 4C). Tyk2−/− mice have a strongly increased susceptibility to L. monocytogenes, which has been associated with an impaired CD8+ T cell cytotoxicity and IFN-γ production (57). Tyk2ΔNK mice succumbed to L. monocytogenes significantly earlier and showed a lower survival rate than Tyk2fl/fl mice (Fig. 4D). Bacterial burden in spleen and liver was significantly higher in Tyk2ΔNK than in Tyk2fl/fl mice, albeit lower than in Tyk2−/− mice at day 5 p.i. (Fig. 4E). In line with the differences in bacterial burden, the survival rate of Tyk2−/− and Tyk2ΔNK mice was 25 and 75%, respectively, compared with Tyk2fl/fl mice at day 5 p.i. (Fig. 4F). Collectively these data indicate that TYK2 in NK cells is required for IL-12 signaling and IFN-γ production by NK cells during L. monocytogenes infection and contributes to an efficient immune defense against L. monocytogenes.

FIGURE 4.

NK cell–intrinsic TYK2 is indispensable for the IL-12–mediated response against L. monocytogenes. (A) DX5 MACS-purified NK cells were expanded for 7 d in the presence of IL-2 and stimulated with IL-12 for 10 min. Levels of phospho- and total STAT4 and STAT3 were assessed by Western blot. Data from one out of two independent experiments are shown. (B) Splenocytes from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were isolated and stimulated with IL-12. Brefeldin A was added after 1 h of incubation, and IFN-γ production by NK cells (CD3εDX5+) was assessed after additional 4 h of incubation by flow cytometry. Mean values ± SEM from three independent experiments are shown (n = 6–10 per genotype). (C) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were infected i.p. with L. monocytogenes (5 × 105 CFU/mouse). Splenocytes were isolated at day 3 p.i and incubated in the presence of brefeldin A for 4 h. IFN-γ production by NK cells (CD3εNK1.1+NKp46+) was assessed by flow cytometry. Mean percentage ± SEM of IFN-γ+ NK cells derived from two to three independent experiments are shown (n = 5–9 per genotype). (D) Tyk2fl/fl and Tyk2ΔNK mice were infected i.p. with L. monocytogenes (L.m., 5 × 105 CFU/mouse), and survival was monitored for 14 d. Data are derived from two independent experiments (n = 10–17 per genotype). (E and F) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were infected i.p. with L. monocytogenes (5 × 105 CFU/mouse). (E) Bacterial load in spleen and liver and (F) survival rate compared with Tyk2fl/fl mice (Tyk2fl/fl set to 100%) was determined 5 d p.i. (E) Mean values ± SEM from two independent experiments (n = 6–12 per genotype) are shown. Dotted line represents the detection limit. (F) Mean percentages relative to Tyk2fl/fl mice from two independent experiments are given. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 4.

NK cell–intrinsic TYK2 is indispensable for the IL-12–mediated response against L. monocytogenes. (A) DX5 MACS-purified NK cells were expanded for 7 d in the presence of IL-2 and stimulated with IL-12 for 10 min. Levels of phospho- and total STAT4 and STAT3 were assessed by Western blot. Data from one out of two independent experiments are shown. (B) Splenocytes from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were isolated and stimulated with IL-12. Brefeldin A was added after 1 h of incubation, and IFN-γ production by NK cells (CD3εDX5+) was assessed after additional 4 h of incubation by flow cytometry. Mean values ± SEM from three independent experiments are shown (n = 6–10 per genotype). (C) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were infected i.p. with L. monocytogenes (5 × 105 CFU/mouse). Splenocytes were isolated at day 3 p.i and incubated in the presence of brefeldin A for 4 h. IFN-γ production by NK cells (CD3εNK1.1+NKp46+) was assessed by flow cytometry. Mean percentage ± SEM of IFN-γ+ NK cells derived from two to three independent experiments are shown (n = 5–9 per genotype). (D) Tyk2fl/fl and Tyk2ΔNK mice were infected i.p. with L. monocytogenes (L.m., 5 × 105 CFU/mouse), and survival was monitored for 14 d. Data are derived from two independent experiments (n = 10–17 per genotype). (E and F) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice were infected i.p. with L. monocytogenes (5 × 105 CFU/mouse). (E) Bacterial load in spleen and liver and (F) survival rate compared with Tyk2fl/fl mice (Tyk2fl/fl set to 100%) was determined 5 d p.i. (E) Mean values ± SEM from two independent experiments (n = 6–12 per genotype) are shown. Dotted line represents the detection limit. (F) Mean percentages relative to Tyk2fl/fl mice from two independent experiments are given. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

We next assessed the ability of NK cells from Tyk2ΔNK mice to lyse tumor cells in vitro. IL-2–expanded NK cells from Tyk2ΔNK mice killed RMA-S and RMA-Rae1 cells as efficiently as those from Tyk2fl/fl littermate controls, whereas, consistent with previous reports (19, 20), NK cells from Tyk2−/− mice had strongly reduced cytotoxic activity (Fig. 5A, 5B). Cultivation in the presence of IL-15 restored cytotoxic activity of NK cells from Tyk2−/− mice (Fig. 5C), indicating that IL-15 not only restores maturation but also functional defects of Tyk2−/− NK cells. To test NK cell cytotoxic activity in vivo, we injected splenocytes from mice deficient for β2m−/− mice, which are recognized and lysed by NK cells because of low MHC class I (MHC I) expression (58, 59). β2m−/− cells were eliminated in Tyk2ΔNK mice as efficiently as in Tyk2fl/fl mice, whereas Tyk2−/− showed severely impaired rejection (Fig. 5D). In line with the cytotoxicity assays, Tyk2ΔNK mice controlled the growth of transplanted RMA-Rae1 tumor cells as efficiently as Tyk2fl/fl mice (Fig. 5E). Consistent with our previous report (20), tumor growth was significantly increased in Tyk2−/− mice (Fig. 5E). NK cell activity is controlled by the balance of signals derived from actR and inhibitory NK cell receptors. Although TYK2 is not involved in signaling downstream of actR (60), such as NK1.1 and Ly49H, Tyk2−/− NK cells have impaired IFN-γ production in response to anti-NK1.1 Ab stimulation (20). Unlike NK cells from Tyk2−/− mice, those from Tyk2ΔNK mice showed unimpaired anti-NK1.1 Ab–induced IFN-γ production (Fig. 5F). Thus, in contrast to IL-12, actR-induced IFN-γ production does not depend on TYK2 in NK cells themselves. Reminiscent of NK cell maturation and cytotoxicity, IFN-γ production in response to NK1.1 activation was rescued by treatment of Tyk2−/− mice with IL-15/IL-15Rα complexes (Fig. 5G). Having established that TYK2 in NK cells themselves is of little importance for NK cell–dependent target cell killing and tumor surveillance, we next assessed the ability of Tyk2ΔM and Tyk2ΔDC mice to kill target cells in vivo and control RMA-Rae1 tumor growth. Tyk2ΔM and Tyk2ΔDC mice showed comparably impaired killing of β2m−/− splenocytes, albeit the defect in Tyk2ΔDC mice was less pronounced than in Tyk2−/− mice (Fig. 5H). Tyk2ΔM mice were able to control RMA-Rae1 tumor cells as efficiently as Tyk2fl/fl controls, whereas Tyk2ΔDC mice showed an intermediate tumor growth between Tyk2fl/fl and Tyk2−/− mice (Fig. 5I). We did not observe differences in the production of IFN-γ in response to actR stimulation between NK cells from Tyk2fl/fl, Tyk2ΔM, and Tyk2ΔDC mice (Fig. 4J).

FIGURE 5.

Tyk2ΔNK cells show restored cytotoxicity, tumor surveillance, and responses to actR stimulation. DX5 MACS-purified, (A and B) IL-2–, or (C) IL-15–expanded NK cells were incubated with (A) RMA-S or (B and C) RMA-Rae1 cells at the indicated effector:target (E:T) ratios. Specific lysis was assessed after 4 h by flow cytometry. (A–C) One representative out of two independent experiments is shown (with three technical replicates per group). (D and H) Splenocytes from β2m−/− and WT mice were isolated, mixed at an 1:1 ratio, and injected i.v. into (D) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (H) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice. The specific rejection of β2m−/− (MHC Ilow) cells was analyzed after 16 h by flow cytometry, and the mean rejection ± SEM from two independent experiments is shown (n = 5–7 per genotype). (E and I) 106 RMA-Rae1 cells were s.c. injected into (E) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (I) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice, and the tumor weight was assessed after 10–13 d. Mean values ± SEM from (E) two independent experiments for Tyk2fl/fl and Tyk2ΔNK (n = 12–14 per genotype) and one experiment for Tyk2−/− mice (n = 4 per genotype) and (I) two independent experiments (n = 12–16 per genotype) are shown. (F and J) Splenocytes from (F) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (J) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice were isolated and stimulated with anti-NK1.1 Ab. Brefeldin A was added after 1 h of incubation, and IFN-γ production by (F) CD3εDX5+ or (J) CD3εNKp46+ NK cells was assessed after additional 4 h of incubation by flow cytometry. Mean percentages ± SEM from (F) two to five independent experiments (n = 10–14 per genotype) or (J) four independent experiments (n = 8–15 per genotype) are shown. (G) WT and Tyk2−/− mice were treated with rmIL-15/IL-15Rα–Fc complexes every 2–3 d, and after four injections splenocytes were isolated and stimulated with anti-NK1.1 Ab for 5 h in the presence of brefeldin A. IFN-γ production of NK cells (CD3εDX5+) was assessed by flow cytometry. Mean percentages ± SEM from two independent experiments are shown (n = 6–7 per genotype). (D, H, I, and J) Tyk2fl/fl are combined littermate controls from Tyk2ΔM and Tyk2ΔDC mice. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 5.

Tyk2ΔNK cells show restored cytotoxicity, tumor surveillance, and responses to actR stimulation. DX5 MACS-purified, (A and B) IL-2–, or (C) IL-15–expanded NK cells were incubated with (A) RMA-S or (B and C) RMA-Rae1 cells at the indicated effector:target (E:T) ratios. Specific lysis was assessed after 4 h by flow cytometry. (A–C) One representative out of two independent experiments is shown (with three technical replicates per group). (D and H) Splenocytes from β2m−/− and WT mice were isolated, mixed at an 1:1 ratio, and injected i.v. into (D) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (H) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice. The specific rejection of β2m−/− (MHC Ilow) cells was analyzed after 16 h by flow cytometry, and the mean rejection ± SEM from two independent experiments is shown (n = 5–7 per genotype). (E and I) 106 RMA-Rae1 cells were s.c. injected into (E) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (I) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice, and the tumor weight was assessed after 10–13 d. Mean values ± SEM from (E) two independent experiments for Tyk2fl/fl and Tyk2ΔNK (n = 12–14 per genotype) and one experiment for Tyk2−/− mice (n = 4 per genotype) and (I) two independent experiments (n = 12–16 per genotype) are shown. (F and J) Splenocytes from (F) Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice or (J) Tyk2fl/fl, Tyk2ΔM, Tyk2DC, and Tyk2−/− mice were isolated and stimulated with anti-NK1.1 Ab. Brefeldin A was added after 1 h of incubation, and IFN-γ production by (F) CD3εDX5+ or (J) CD3εNKp46+ NK cells was assessed after additional 4 h of incubation by flow cytometry. Mean percentages ± SEM from (F) two to five independent experiments (n = 10–14 per genotype) or (J) four independent experiments (n = 8–15 per genotype) are shown. (G) WT and Tyk2−/− mice were treated with rmIL-15/IL-15Rα–Fc complexes every 2–3 d, and after four injections splenocytes were isolated and stimulated with anti-NK1.1 Ab for 5 h in the presence of brefeldin A. IFN-γ production of NK cells (CD3εDX5+) was assessed by flow cytometry. Mean percentages ± SEM from two independent experiments are shown (n = 6–7 per genotype). (D, H, I, and J) Tyk2fl/fl are combined littermate controls from Tyk2ΔM and Tyk2ΔDC mice. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Taken together, these data show that cell-extrinsic TYK2 promotes NK cell cytotoxicity, tumor surveillance, and actR-induced IFN-γ production and that TYK2 signaling in myeloid cells and DCs augments NK cell–dependent target cell killing in vivo.

Human NK cells acquire the competence to produce IFN-γ in response to actR engagement during maturation through progressive demethylation at the Ifng locus (61). To test whether differences in DNA methylation cause the difference in IFN-γ production upon actR stimulation between WT and Tyk2−/− NK cells, we expanded NK cells for 72 h in the presence of 5-Aza, a drug that blocks DNA methylation during cell proliferation. WT and Tyk2−/− NK cells showed similar survival under these conditions (Supplemental Fig. 4). 5-Aza treatment restored anti-NK1.1 Ab–induced IFN-γ production by Tyk2−/− NK cells to levels produced by WT NK cells (Fig. 6A). Importantly, IL-12–induced IFN-γ production remained strongly reduced (Fig. 6B), arguing against a general enhancement of Ifng gene induction upon global demethylation. We next analyzed the methylation status of nine CpGs located around the TSS of the Ifng gene in NK cells from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice (Fig. 6C). We found a significant increase in methylation of four CpGs (−212, −58, +91, and +114) in NK cells from Tyk2−/− compared with those from Tyk2fl/fl and Tyk2ΔNK mice (Fig. 6D). Although not conserved in humans, CpGs +91 and +114 are in the same region as CpGs +122 and +128 at human Ifng promoter, which undergo demethylation during NK cell maturation (61). Consistent with the maturation-independent low level of methylation of the CpG at position −53 observed in human NK cells (61), CpGs in the proximal Ifng promoter (−198, −178, −58, −50, −39) were mostly demethylated in NK cells from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice (Fig. 6D). Collectively, these data suggest that impaired actR-induced IFN-γ production by NK cells from Tyk2−/− mice is due to hypermethylation at the Ifng gene locus and most likely reflects defective terminal maturation.

FIGURE 6.

DNA demethylation restores IFN-γ production by Tyk2−/− NK cells in response to actR but not IL-12, and Tyk2−/− NK cells have increased methylation at three CpG sites in the Ifng gene locus compared with WT cells. (A and B) DX5 MACS-purified NK cells were expanded in the presence of IL-2 for 3 d in the absence (Ctrl) or presence of 5-Aza, stimulated with (A) anti-NK1.1 Ab, or (B) IL-2 and IL-12. Brefeldin A was added after 1 h incubation, and IFN-γ production by CD3εDX5+ NK cells was assessed after additional 4 h of incubation by flow cytometry. Mean values ± SEM from (A) two independent experiments (n = 3–4 per genotype and treatment) and (B) two independent experiments (n = 4 per genotype and treatment) are shown. (C) Alignment of the mouse and the human Ifng promoter sequence around TSS. CpGs are highlighted in gray boxes with the relative position to the TSS indicated in brackets. Underlined sequences indicate the position of the primers used for the amplification of the region of interest. The underlined ATG indicates the translational start site. (D) DNA was isolated from FACS-sorted splenic NK cells (CD3εNK1.1+) from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice. CpG methylation was analyzed by bisulfite pyrosequencing. Data are shown as mean percentage of methylation ± SEM from three independent experiments (n = 3 per genotype). *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 6.

DNA demethylation restores IFN-γ production by Tyk2−/− NK cells in response to actR but not IL-12, and Tyk2−/− NK cells have increased methylation at three CpG sites in the Ifng gene locus compared with WT cells. (A and B) DX5 MACS-purified NK cells were expanded in the presence of IL-2 for 3 d in the absence (Ctrl) or presence of 5-Aza, stimulated with (A) anti-NK1.1 Ab, or (B) IL-2 and IL-12. Brefeldin A was added after 1 h incubation, and IFN-γ production by CD3εDX5+ NK cells was assessed after additional 4 h of incubation by flow cytometry. Mean values ± SEM from (A) two independent experiments (n = 3–4 per genotype and treatment) and (B) two independent experiments (n = 4 per genotype and treatment) are shown. (C) Alignment of the mouse and the human Ifng promoter sequence around TSS. CpGs are highlighted in gray boxes with the relative position to the TSS indicated in brackets. Underlined sequences indicate the position of the primers used for the amplification of the region of interest. The underlined ATG indicates the translational start site. (D) DNA was isolated from FACS-sorted splenic NK cells (CD3εNK1.1+) from Tyk2fl/fl, Tyk2ΔNK, and Tyk2−/− mice. CpG methylation was analyzed by bisulfite pyrosequencing. Data are shown as mean percentage of methylation ± SEM from three independent experiments (n = 3 per genotype). *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

In this study, we provide evidence that NK cells require at least two distinct TYK2-dependent signaling inputs to be fully functional in both tumor surveillance and antibacterial immunity: an NK cell–extrinsic input that drives NK cell maturation, cytotoxicity, and IFN-γ production in response to actR stimulation and an NK cell–intrinsic input that drives IFN-γ production in response to IL-12 and contributes to the immune defense against bacterial infection.

NK cells from Tyk2−/− mice develop normally in the BM but show impaired terminal maturation in the spleen (20). Using mice that lack TYK2 specifically in NK cells (Tyk2ΔNK) and mixed BM chimera experiments, we show that TYK2 in accessory cells rather than in NK cells themselves drives splenic NK cell maturation. Several lines of evidence support the idea that IL-15 trans-presentation by CD11c+ DCs and LysM+ cells (i.e., mainly monocytes, macrophages, and neutrophils) is required for peripheral NK cell homeostasis and terminal maturation into the CD27CD11b+ NK cell stage (4952). Our finding that lack of TYK2 in CD11c+ cells results in a reduction of CD27CD11b+ splenic NK cells further underscores the importance of DCs for peripheral NK cell maturation. In contrast, we did not find evidence for a role of TYK2 in LysM+ cells in this process. It is currently unclear which signals drive IL-15/IL-15Rα expression under homeostatic conditions. We show that the absence of TYK2 results in reduced IL-15Rα levels on macrophages and DCs in naive mice. In response to immune challenge, type I IFNs induce IL-15 production and trans-presentation from DCs to NK cells (62). It is generally accepted that low levels of type I IFNs are produced under homeostatic conditions (63), prompting the hypothesis that IL-15 trans-presentation to NK cells may be driven by type I IFNs even in the absence of immune challenge. This idea is supported by the findings that naive Stat1−/− and Ifnar1−/− mice, but not Ifnar1ΔNK mice, have less CD27CD11b+ NK cells and that Stat1−/− NK cells fully mature upon transfer into WT mice (40, 64). However, further studies will be required to test this possibility and to confirm that TYK2 drives NK cell maturation through its role in the type I IFN signaling cascade.

IFN-γ is a major NK cell effector molecule that is critical for tumor surveillance and immunity against infections. IFN-γ production by NK cells can be triggered by cytokines, such as IL-12 and IL-18, or actR stimulation (65). In support of the well-established role of TYK2 in the IL-12 signaling cascade (18), IFN-γ production in response to IL-12 was similarly impaired in NK cells from Tyk2ΔNK and Tyk2−/− mice. IFN-γ production by NK cells was also severely impaired in Tyk2ΔNK mice upon infection with L. monocytogenes. Our previous study revealed defects in anti-NK1.1 and anti-Ly49D Ab–induced IFN-γ production in Tyk2−/− NK cells, although TYK2 is not involved in the signaling cascade downstream of NK cell actR (60). Unlike NK cells from Tyk2−/− mice, splenic NK cells from Tyk2ΔNK mice showed similar IFN-γ production as those from Tyk2fl/fl mice upon stimulation with anti-NK1.1 Ab, indicating that the impact of TYK2 is indirect. Recently the most mature CD27CD11b+ NK cells have been identified as the main producers of IFN-γ in response to actR stimulation (52), suggesting that the impaired anti-NK1.1–induced IFN-γ production in the absence of TYK2 may reflect defects in terminal maturation. However, our finding that NK cells from Tyk2ΔDC mice have a lower frequency of the CD27CD11b+ cells but show unimpaired actR-induced IFN-γ production argues against this possibility. In human NK cells, terminal differentiation of peripheral NK cells is accompanied with demethylation of the Ifng promoter at three CpG sites in the transcribed region (+122, +128, +171), which enables high IFN-γ production in response to actR engagement (61). Our study suggests that such a mechanism is also important in mice, as the impaired maturation of Tyk2−/− NK cells correlated with an increased methylation at CpGs +91 and +114 and impaired IFN-γ production upon actR stimulation. Moreover, treatment of NK cells from Tyk2−/− mice with 5-Aza restored anti-NK1.1 Ab– but not IL-12–induced IFN-γ production. However, more detailed studies will be required to test whether these CpGs are indeed demethylated during murine NK cell maturation and whether this increased methylation in Tyk2−/− NK cells reflects the more immature phenotype.

Furthermore, we show that TYK2 in NK cells is not required for target cell cytotoxicity and NK cell–dependent tumor surveillance. In vitro expanded NK cells from Tyk2ΔNK mice showed unimpaired cytotoxic activity against RMA-S and RMA-Rae1 target cells, whereas NK cells from Tyk2−/− mice had strongly reduced cytotoxic activity. Similar to what has been shown for NK cells from Stat1−/− mice (66), NK cells from Tyk2−/− mice had restored cytotoxicity against RMA-Rae1 cells when grown in the presence of IL-15 instead of IL-2. It remains to be investigated whether this reflects differences in cellular responses to IL-2 and IL-15 (67) or in signal strength. However, these data further underscore that IL-15 not only restores maturation but also functional competence of NK cells from Tyk2−/− mice. Tyk2ΔNK mice rejected cells that lack MHC I molecules (β2m−/− splenocytes), and controlled s.c. transplanted RMA-Rae1 cells as efficiently as littermate controls. We show that TYK2 in LysM+ cells and DCs is required for in vivo killing of β2m−/− splenocytes and that TYK2 in DCs contributes to RMA-Rae1 growth control. It remains an open question which external TYK2-dependent signal augments NK cell cytotoxicity and tumor surveillance. Defects in cytotoxicity may, at least in part, be causally linked to defects in terminal maturation or may result from impaired MHC I molecule–dependent NK cell education, although these two processes might not be completely independent (61, 68, 69). Further studies are required to define how TYK2 regulates the interplay between NK cells and other immune and nonimmune cells in both the differentiation of a functionally balanced NK cell pool and in antitumor immunity.

Although not in the focus of our study, we found that TYK2 signaling in NK cells is required for an efficient immune defense against L. monocytogenes. This is surprising, as several studies established that NK cells act deleterious rather than protective during systemic L. monocytogenes infections (7072). It seems possible that despite their predominant immunosuppressive phenotype, NK cells have low level of antibacterial activity. This idea is supported by the findings that NK cells produce considerable amounts of IFN-γ upon i.p. L. monocytogenes infection (7376) and modestly reduce bacterial burden upon transfer into Ifng−/− mice (77) and upon local infections (62, 78). Increased sensitivity of Tyk2ΔNK mice to L. monocytogenes infection correlated with an impaired IFN-γ production by NK cells, supporting the notion that the balance between IFN-γ– and IL-10–producing NK cells critically determines their role in antibacterial immunity (79, 80), thus arguing against a previously suggested negative impact of NK cell–derived IFN-γ (72). However, further investigations are required to characterize the impact of TYK2 on NK cell responses other than the production of IFN-γ to understand how NK cell–intrinsic TYK2 improves survival upon L. monocytogenes infection.

In summary, we provide evidence that NK cell–extrinsic TYK2 promotes NK cell maturation, cytotoxicity, and tumor surveillance, whereas NK cell–intrinsic TYK2 is needed for an efficient immune defense against L. monocytogenes infection. Moreover, we show that TYK2 not only facilitates IFN-γ production through its role in the IL-12 signaling cascade but also through NK cell–extrinsic mechanisms that specifically impact on IFN-γ production in response to NK cell actR stimulation and may involve Ifng promoter demethylation. Our findings that cytotoxic defects of NK cells from Tyk2−/− mice can be rescued by IL-15 treatment suggest that unwanted effects of TYK2 inhibitors in tumor therapy may be overcome by boosting NK cell activity. However, more detailed experimentation will be required to better understand how TYK2 regulates NK cell–dependent immunity in vivo and to identify signaling cascades and cellular networks involved.

We thank Claus Vogl for help with the statistical analysis and Therese Lederer, Sabine Fajmann, Sabine Macho-Maschler, Daniela Prinz, and Dominika Gogová for excellent technical support. We are grateful to Michaela Prchal-Murphy, Dagmar Gotthardt, and Eva Maria Putz for valuable discussions and, additionally, Eva Maria Putz for critically reading of the manuscript.

This work was supported by Austrian Science Fund Project P-25642 (to B.S.) and Special Research Programmes SFB-F6101 and SFB-F6106 (to M.M. and B.S.), SFB-F6102 (to C.B. and M.F.), and SFB-F6107 (to V.S.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

actR

activating receptor

5-Aza

5-aza-2-deoxycytidine

BM

bone marrow

DC

dendritic cell

Ly5.1

B6.SJL-Ptprca

β2m−/−

β2-microglobulin−/−

MHC I

MHC class I

p.i.

postinfection

rm

recombinant murine

S

sequencing primer

TSS

transcriptional start site

TYK2

tyrosine kinase 2

WT

wild-type.

1
Vivier
,
E.
,
E.
Tomasello
,
M.
Baratin
,
T.
Walzer
,
S.
Ugolini
.
2008
.
Functions of natural killer cells.
Nat. Immunol.
9
:
503
510
.
2
Marçais
,
A.
,
S.
Viel
,
M.
Grau
,
T.
Henry
,
J.
Marvel
,
T.
Walzer
.
2013
.
Regulation of mouse NK cell development and function by cytokines.
Front. Immunol.
4
:
450
.
3
Gotthardt
,
D.
,
V.
Sexl
.
2017
.
STATs in NK-cells: the good, the bad, and the ugly.
Front. Immunol.
7
:
694
.
4
Neubauer
,
H.
,
A.
Cumano
,
M.
Müller
,
H.
Wu
,
U.
Huffstadt
,
K.
Pfeffer
.
1998
.
Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis.
Cell
93
:
397
409
.
5
Parganas
,
E.
,
D.
Wang
,
D.
Stravopodis
,
D. J.
Topham
,
J. C.
Marine
,
S.
Teglund
,
E. F.
Vanin
,
S.
Bodner
,
O. R.
Colamonici
,
J. M.
van Deursen
, et al
.
1998
.
Jak2 is essential for signaling through a variety of cytokine receptors.
Cell
93
:
385
395
.
6
Rodig
,
S. J.
,
M. A.
Meraz
,
J. M.
White
,
P. A.
Lampe
,
J. K.
Riley
,
C. D.
Arthur
,
K. L.
King
,
K. C.
Sheehan
,
L.
Yin
,
D.
Pennica
, et al
.
1998
.
Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses.
Cell
93
:
373
383
.
7
Bottos
,
A.
,
D.
Gotthardt
,
J. W.
Gill
,
A.
Gattelli
,
A.
Frei
,
A.
Tzankov
,
V.
Sexl
,
A.
Wodnar-Filipowicz
,
N. E.
Hynes
.
2016
.
Decreased NK-cell tumour immunosurveillance consequent to JAK inhibition enhances metastasis in breast cancer models.
Nat. Commun.
7
:
12258
.
8
Schönberg
,
K.
,
J.
Rudolph
,
M.
Vonnahme
,
S.
Parampalli Yajnanarayana
,
I.
Cornez
,
M.
Hejazi
,
A. R.
Manser
,
M.
Uhrberg
,
W.
Verbeek
,
S.
Koschmieder
, et al
.
2015
.
JAK inhibition impairs NK cell function in myeloproliferative neoplasms.
Cancer Res.
75
:
2187
2199
.
9
Bongfen
,
S. E.
,
I. G.
Rodrigue-Gervais
,
J.
Berghout
,
S.
Torre
,
P.
Cingolani
,
S. A.
Wiltshire
,
G. A.
Leiva-Torres
,
L.
Letourneau
,
R.
Sladek
,
M.
Blanchette
, et al
.
2012
.
An N-ethyl-N-nitrosourea (ENU)-induced dominant negative mutation in the JAK3 kinase protects against cerebral malaria.
PLoS One
7
:
e31012
.
10
Park
,
S. Y.
,
K.
Saijo
,
T.
Takahashi
,
M.
Osawa
,
H.
Arase
,
N.
Hirayama
,
K.
Miyake
,
H.
Nakauchi
,
T.
Shirasawa
,
T.
Saito
.
1995
.
Developmental defects of lymphoid cells in Jak3 kinase-deficient mice.
Immunity
3
:
771
782
.
11
Robinette
,
M. L.
,
M.
Cella
,
J. B.
Telliez
,
T. K.
Ulland
,
A. D.
Barrow
,
K.
Capuder
,
S.
Gilfillan
,
L. L.
Lin
,
L. D.
Notarangelo
,
M.
Colonna
.
2018
.
Jak3 deficiency blocks innate lymphoid cell development.
Mucosal Immunol.
11
:
50
60
.
12
Roberts
,
J. L.
,
A.
Lengi
,
S. M.
Brown
,
M.
Chen
,
Y. J.
Zhou
,
J. J.
O’Shea
,
R. H.
Buckley
.
2004
.
Janus kinase 3 (JAK3) deficiency: clinical, immunologic, and molecular analyses of 10 patients and outcomes of stem cell transplantation.
Blood
103
:
2009
2018
.
13
Vély
,
F.
,
V.
Barlogis
,
B.
Vallentin
,
B.
Neven
,
C.
Piperoglou
,
M.
Ebbo
,
T.
Perchet
,
M.
Petit
,
N.
Yessaad
,
F.
Touzot
, et al
.
2016
.
Evidence of innate lymphoid cell redundancy in humans.
Nat. Immunol.
17
:
1291
1299
.
14
Schleicher
,
U.
,
J.
Mattner
,
M.
Blos
,
H.
Schindler
,
M.
Röllinghoff
,
M.
Karaghiosoff
,
M.
Müller
,
G.
Werner-Felmayer
,
C.
Bogdan
.
2004
.
Control of Leishmania major in the absence of Tyk2 kinase.
Eur. J. Immunol.
34
:
519
529
.
15
Shimoda
,
K.
,
H.
Tsutsui
,
K.
Aoki
,
K.
Kato
,
T.
Matsuda
,
A.
Numata
,
K.
Takase
,
T.
Yamamoto
,
H.
Nukina
,
T.
Hoshino
, et al
.
2002
.
Partial impairment of interleukin-12 (IL-12) and IL-18 signaling in Tyk2-deficient mice.
Blood
99
:
2094
2099
.
16
Kreins
,
A. Y.
,
M. J.
Ciancanelli
,
S.
Okada
,
X. F.
Kong
,
N.
Ramírez-Alejo
,
S. S.
Kilic
,
J.
El Baghdadi
,
S.
Nonoyama
,
S. A.
Mahdaviani
,
F.
Ailal
, et al
.
2015
.
Human TYK2 deficiency: mycobacterial and viral infections without hyper-IgE syndrome.
J. Exp. Med.
212
:
1641
1662
.
17
Fuchs
,
S.
,
P.
Kaiser-Labusch
,
J.
Bank
,
S.
Ammann
,
A.
Kolb-Kokocinski
,
C.
Edelbusch
,
H.
Omran
,
S.
Ehl
.
2016
.
Tyrosine kinase 2 is not limiting human antiviral type III interferon responses.
Eur. J. Immunol.
46
:
2639
2649
.
18
Strobl
,
B.
,
D.
Stoiber
,
V.
Sexl
,
M.
Mueller
.
2011
.
Tyrosine kinase 2 (TYK2) in cytokine signalling and host immunity.
Front. Biosci. (Landmark Ed)
16
:
3214
3232
.
19
Stoiber
,
D.
,
B.
Kovacic
,
C.
Schuster
,
C.
Schellack
,
M.
Karaghiosoff
,
R.
Kreibich
,
E.
Weisz
,
M.
Artwohl
,
O. C.
Kleine
,
M.
Muller
, et al
.
2004
.
TYK2 is a key regulator of the surveillance of B lymphoid tumors.
J. Clin. Invest.
114
:
1650
1658
.
20
Prchal-Murphy
,
M.
,
A.
Witalisz-Siepracka
,
K. T.
Bednarik
,
E. M.
Putz
,
D.
Gotthardt
,
K.
Meissl
,
V.
Sexl
,
M.
Müller
,
B.
Strobl
.
2015
.
In vivo tumor surveillance by NK cells requires TYK2 but not TYK2 kinase activity.
OncoImmunology
4
:
e1047579
.
21
Buchert
,
M.
,
C. J.
Burns
,
M.
Ernst
.
2016
.
Targeting JAK kinase in solid tumors: emerging opportunities and challenges.
Oncogene
35
:
939
951
.
22
Kontzias
,
A.
,
A.
Kotlyar
,
A.
Laurence
,
P.
Changelian
,
J. J.
O’Shea
.
2012
.
Jakinibs: a new class of kinase inhibitors in cancer and autoimmune disease.
Curr. Opin. Pharmacol.
12
:
464
470
.
23
Lai
,
P. S.
,
D. A.
Rosa
,
A.
Magdy Ali
,
R. F.
Gómez-Biagi
,
D. P.
Ball
,
A. E.
Shouksmith
,
P. T.
Gunning
.
2015
.
A STAT inhibitor patent review: progress since 2011. [Published erratum appears in 2015 Expert Opin. Ther. Pat. 25: 1495.]
Expert Opin. Ther. Pat.
25
:
1397
1421
.
24
Villarino
,
A. V.
,
Y.
Kanno
,
J. J.
O’Shea
.
2017
.
Mechanisms and consequences of Jak-STAT signaling in the immune system.
Nat. Immunol.
18
:
374
384
.
25
Akahane
,
K.
,
Z.
Li
,
J.
Etchin
,
A.
Berezovskaya
,
E.
Gjini
,
C. E.
Masse
,
W.
Miao
,
J.
Rocnik
,
R.
Kapeller
,
J. R.
Greenwood
, et al
.
2017
.
Anti-leukaemic activity of the TYK2 selective inhibitor NDI-031301 in T-cell acute lymphoblastic leukaemia.
Br. J. Haematol.
177
:
271
282
.
26
Jang
,
W. D.
,
J. T.
Kim
,
H. Y.
Son
,
S. Y.
Park
,
Y. S.
Cho
,
T. S.
Koo
,
H.
Lee
,
N. S.
Kang
.
2015
.
Discovery of Tyk2 inhibitors via the virtual site-directed fragment-based drug design.
Bioorg. Med. Chem. Lett.
25
:
3947
3952
.
27
Leitner
,
N. R.
,
A.
Witalisz-Siepracka
,
B.
Strobl
,
M.
Müller
.
2017
.
Tyrosine kinase 2 - surveillant of tumours and bona fide oncogene.
Cytokine
89
:
209
218
.
28
Liang
,
J.
,
A.
Van Abbema
,
M.
Balazs
,
K.
Barrett
,
L.
Berezhkovsky
,
W. S.
Blair
,
C.
Chang
,
D.
Delarosa
,
J.
DeVoss
,
J.
Driscoll
, et al
.
2017
.
Identification of an imidazopyridine scaffold to generate potent and selective TYK2 inhibitors that demonstrate activity in an in vivo psoriasis model.
Bioorg. Med. Chem. Lett.
27
:
4370
4376
.
29
Liang
,
Y.
,
Y.
Zhu
,
Y.
Xia
,
H.
Peng
,
X. K.
Yang
,
Y. Y.
Liu
,
W. D.
Xu
,
H. F.
Pan
,
D. Q.
Ye
.
2014
.
Therapeutic potential of tyrosine kinase 2 in autoimmunity.
Expert Opin. Ther. Targets
18
:
571
580
.
30
Menet
,
C. J.
2014
.
Toward selective TYK2 inhibitors as therapeutic agents for the treatment of inflammatory diseases.
Pharm. Pat. Anal.
3
:
449
466
.
31
Tron
,
A. E.
,
E. K.
Keeton
,
M.
Ye
,
M.
Casas-Selves
,
H.
Chen
,
K. S.
Dillman
,
R. E.
Gale
,
C.
Stengel
,
M.
Zinda
,
D. C.
Linch
, et al
.
2016
.
Next-generation sequencing identifies a novel ELAVL1-TYK2 fusion gene in MOLM-16, an AML cell line highly sensitive to the PIM kinase inhibitor AZD1208.
Leuk. Lymphoma
57
:
2927
2929
.
32
Waanders
,
E.
,
B.
Scheijen
,
M. C.
Jongmans
,
H.
Venselaar
,
S. V.
van Reijmersdal
,
A. H.
van Dijk
,
A.
Pastorczak
,
R. D.
Weren
,
C. E.
van der Schoot
,
M.
van de Vorst
, et al
.
2017
.
Germline activating TYK2 mutations in pediatric patients with two primary acute lymphoblastic leukemia occurrences.
Leukemia
31
:
821
828
.
33
Karaghiosoff
,
M.
,
H.
Neubauer
,
C.
Lassnig
,
P.
Kovarik
,
H.
Schindler
,
H.
Pircher
,
B.
McCoy
,
C.
Bogdan
,
T.
Decker
,
G.
Brem
, et al
.
2000
.
Partial impairment of cytokine responses in Tyk2-deficient mice.
Immunity
13
:
549
560
.
34
Vielnascher
,
R. M.
,
E.
Hainzl
,
N. R.
Leitner
,
M.
Rammerstorfer
,
D.
Popp
,
A.
Witalisz
,
R.
Rom
,
M.
Karaghiosoff
,
T.
Kolbe
,
S.
Müller
, et al
.
2014
.
Conditional ablation of TYK2 in immunity to viral infection and tumor surveillance.
Transgenic Res.
23
:
519
529
.
35
Eckelhart
,
E.
,
W.
Warsch
,
E.
Zebedin
,
O.
Simma
,
D.
Stoiber
,
T.
Kolbe
,
T.
Rülicke
,
M.
Mueller
,
E.
Casanova
,
V.
Sexl
.
2011
.
A novel Ncr1-Cre mouse reveals the essential role of STAT5 for NK-cell survival and development.
Blood
117
:
1565
1573
.
36
Zijlstra
,
M.
,
M.
Bix
,
N. E.
Simister
,
J. M.
Loring
,
D. H.
Raulet
,
R.
Jaenisch
.
1990
.
Beta 2-microglobulin deficient mice lack CD4-8+ cytolytic T cells.
Nature
344
:
742
746
.
37
Kärre
,
K.
,
H. G.
Ljunggren
,
G.
Piontek
,
R.
Kiessling
.
1986
.
Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy.
Nature
319
:
675
678
.
38
Cerwenka
,
A.
,
J. L.
Baron
,
L. L.
Lanier
.
2001
.
Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing tumor in vivo.
Proc. Natl. Acad. Sci. USA
98
:
11521
11526
.
39
Elpek
,
K. G.
,
M. P.
Rubinstein
,
A.
Bellemare-Pelletier
,
A. W.
Goldrath
,
S. J.
Turley
.
2010
.
Mature natural killer cells with phenotypic and functional alterations accumulate upon sustained stimulation with IL-15/IL-15Ralpha complexes.
Proc. Natl. Acad. Sci. USA
107
:
21647
21652
.
40
Mizutani
,
T.
,
N.
Neugebauer
,
E. M.
Putz
,
N.
Moritz
,
O.
Simma
,
E.
Zebedin-Brandl
,
D.
Gotthardt
,
W.
Warsch
,
E.
Eckelhart
,
H. P.
Kantner
, et al
.
2012
.
Conditional IFNAR1 ablation reveals distinct requirements of Type I IFN signaling for NK cell maturation and tumor surveillance.
OncoImmunology
1
:
1027
1037
.
41
Prchal-Murphy
,
M.
,
C.
Semper
,
C.
Lassnig
,
B.
Wallner
,
C.
Gausterer
,
I.
Teppner-Klymiuk
,
J.
Kobolak
,
S.
Müller
,
T.
Kolbe
,
M.
Karaghiosoff
, et al
.
2012
.
TYK2 kinase activity is required for functional type I interferon responses in vivo.
PLoS One
7
:
e39141
.
42
Aken
,
B. L.
,
S.
Ayling
,
D.
Barrell
,
L.
Clarke
,
V.
Curwen
,
S.
Fairley
,
J.
Fernandez Banet
,
K.
Billis
,
C.
García Girón
,
T.
Hourlier
, et al
.
2016
.
The Ensembl gene annotation system.
Database (Oxford)
. DOI: 10.1093/database/baw093.
43
de Araújo-Souza
,
P. S.
,
S. C.
Hanschke
,
J. P.
Viola
.
2015
.
Epigenetic control of interferon-gamma expression in CD8 T cells.
J. Immunol. Res.
2015
:
849573
.
44
Fodil
,
N.
,
D.
Langlais
,
P.
Moussa
,
G. A.
Boivin
,
T.
Di Pietrantonio
,
I.
Radovanovic
,
A.
Dumaine
,
M.
Blanchette
,
E.
Schurr
,
P.
Gros
,
S. M.
Vidal
.
2014
.
Specific dysregulation of IFNγ production by natural killer cells confers susceptibility to viral infection.
PLoS Pathog.
10
:
e1004511
.
45
Huntington
,
N. D.
,
H.
Tabarias
,
K.
Fairfax
,
J.
Brady
,
Y.
Hayakawa
,
M. A.
Degli-Esposti
,
M. J.
Smyth
,
D. M.
Tarlinton
,
S. L.
Nutt
.
2007
.
NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation.
J. Immunol.
178
:
4764
4770
.
46
Sun
,
J. C.
,
L. L.
Lanier
.
2011
.
NK cell development, homeostasis and function: parallels with CD8+ T cells.
Nat. Rev. Immunol.
11
:
645
657
.
47
van Helden
,
M. J.
,
S.
Goossens
,
C.
Daussy
,
A. L.
Mathieu
,
F.
Faure
,
A.
Marçais
,
N.
Vandamme
,
N.
Farla
,
K.
Mayol
,
S.
Viel
, et al
.
2015
.
Terminal NK cell maturation is controlled by concerted actions of T-bet and Zeb2 and is essential for melanoma rejection.
J. Exp. Med.
212
:
2015
2025
.
48
Goh
,
W.
,
N. D.
Huntington
.
2017
.
Regulation of murine natural killer cell development.
Front. Immunol.
8
:
130
.
49
Dubois
,
S.
,
J.
Mariner
,
T. A.
Waldmann
,
Y.
Tagaya
.
2002
.
IL-15Ralpha recycles and presents IL-15 in trans to neighboring cells.
Immunity
17
:
537
547
.
50
Mortier
,
E.
,
R.
Advincula
,
L.
Kim
,
S.
Chmura
,
J.
Barrera
,
B.
Reizis
,
B. A.
Malynn
,
A.
Ma
.
2009
.
Macrophage- and dendritic-cell-derived interleukin-15 receptor alpha supports homeostasis of distinct CD8+ T cell subsets.
Immunity
31
:
811
822
.
51
Soderquest
,
K.
,
N.
Powell
,
C.
Luci
,
N.
van Rooijen
,
A.
Hidalgo
,
F.
Geissmann
,
T.
Walzer
,
G. M.
Lord
,
A.
Martín-Fontecha
.
2011
.
Monocytes control natural killer cell differentiation to effector phenotypes.
Blood
117
:
4511
4518
.
52
Luu
,
T. T.
,
S.
Ganesan
,
A. K.
Wagner
,
D.
Sarhan
,
S.
Meinke
,
N.
Garbi
,
G.
Hämmerling
,
E.
Alici
,
K.
Kärre
,
B. J.
Chambers
, et al
.
2016
.
Independent control of natural killer cell responsiveness and homeostasis at steady-state by CD11c+ dendritic cells.
Sci. Rep.
6
:
37996
.
53
Jaeger
,
B. N.
,
J.
Donadieu
,
C.
Cognet
,
C.
Bernat
,
D.
Ordoñez-Rueda
,
V.
Barlogis
,
N.
Mahlaoui
,
A.
Fenis
,
E.
Narni-Mancinelli
,
B.
Beaupain
, et al
.
2012
.
Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis.
J. Exp. Med.
209
:
565
580
.
54
Huntington
,
N. D.
2014
.
The unconventional expression of IL-15 and its role in NK cell homeostasis.
Immunol. Cell Biol.
92
:
210
213
.
55
Ma
,
A.
,
R.
Koka
,
P.
Burkett
.
2006
.
Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis.
Annu. Rev. Immunol.
24
:
657
679
.
56
Castillo
,
E. F.
,
S. W.
Stonier
,
L.
Frasca
,
K. S.
Schluns
.
2009
.
Dendritic cells support the in vivo development and maintenance of NK cells via IL-15 trans-presentation.
J. Immunol.
183
:
4948
4956
.
57
Aizu
,
K.
,
W.
Li
,
T.
Yajima
,
T.
Arai
,
K.
Shimoda
,
Y.
Nimura
,
Y.
Yoshikai
.
2006
.
An important role of Tyk2 in APC function of dendritic cells for priming CD8+ T cells producing IFN-gamma.
Eur. J. Immunol.
36
:
3060
3070
.
58
Kärre
,
K.
2002
.
NK cells, MHC class I molecules and the missing self.
Scand. J. Immunol.
55
:
221
228
.
59
Raulet
,
D. H.
2006
.
Missing self recognition and self tolerance of natural killer (NK) cells.
Semin. Immunol.
18
:
145
150
.
60
Lanier
,
L. L.
2005
.
NK cell recognition.
Annu. Rev. Immunol.
23
:
225
274
.
61
Luetke-Eversloh
,
M.
,
B. B.
Cicek
,
F.
Siracusa
,
J. T.
Thom
,
A.
Hamann
,
S.
Frischbutter
,
R.
Baumgrass
,
H. D.
Chang
,
A.
Thiel
,
J.
Dong
,
C.
Romagnani
.
2014
.
NK cells gain higher IFN-γ competence during terminal differentiation.
Eur. J. Immunol.
44
:
2074
2084
.
62
Lucas
,
M.
,
W.
Schachterle
,
K.
Oberle
,
P.
Aichele
,
A.
Diefenbach
.
2007
.
Dendritic cells prime natural killer cells by trans-presenting interleukin 15.
Immunity
26
:
503
517
.
63
Gough
,
D. J.
,
N. L.
Messina
,
C. J.
Clarke
,
R. W.
Johnstone
,
D. E.
Levy
.
2012
.
Constitutive type I interferon modulates homeostatic balance through tonic signaling.
Immunity
36
:
166
174
.
64
Robbins
,
S. H.
,
M. S.
Tessmer
,
L.
Van Kaer
,
L.
Brossay
.
2005
.
Direct effects of T-bet and MHC class I expression, but not STAT1, on peripheral NK cell maturation.
Eur. J. Immunol.
35
:
757
765
.
65
Vivier
,
E.
,
D. H.
Raulet
,
A.
Moretta
,
M. A.
Caligiuri
,
L.
Zitvogel
,
L. L.
Lanier
,
W. M.
Yokoyama
,
S.
Ugolini
.
2011
.
Innate or adaptive immunity? The example of natural killer cells.
Science
331
:
44
49
.
66
Lee
,
C. K.
,
D. T.
Rao
,
R.
Gertner
,
R.
Gimeno
,
A. B.
Frey
,
D. E.
Levy
.
2000
.
Distinct requirements for IFNs and STAT1 in NK cell function.
J. Immunol.
165
:
3571
3577
.
67
Meazza
,
R.
,
B.
Azzarone
,
A. M.
Orengo
,
S.
Ferrini
.
2011
.
Role of common-gamma chain cytokines in NK cell development and function: perspectives for immunotherapy.
J. Biomed. Biotechnol.
2011
:
861920
.
68
Brodin
,
P.
,
T.
Lakshmikanth
,
K.
Kärre
,
P.
Höglund
.
2012
.
Skewing of the NK cell repertoire by MHC class I via quantitatively controlled enrichment and contraction of specific Ly49 subsets.
J. Immunol.
188
:
2218
2226
.
69
Höglund
,
P.
,
P.
Brodin
.
2010
.
Current perspectives of natural killer cell education by MHC class I molecules.
Nat. Rev. Immunol.
10
:
724
734
.
70
Clark
,
S. E.
,
H. C.
Filak
,
B. S.
Guthrie
,
R. L.
Schmidt
,
A.
Jamieson
,
P.
Merkel
,
V.
Knight
,
C. M.
Cole
,
D. H.
Raulet
,
L. L.
Lenz
.
2016
.
Bacterial manipulation of NK cell regulatory activity increases susceptibility to Listeria monocytogenes infection.
PLoS Pathog.
12
:
e1005708
.
71
Teixeira
,
H. C.
,
S. H.
Kaufmann
.
1994
.
Role of NK1.1+ cells in experimental listeriosis. NK1+ cells are early IFN-gamma producers but impair resistance to Listeria monocytogenes infection.
J. Immunol.
152
:
1873
1882
.
72
Viegas
,
N.
,
L.
Andzinski
,
C. F.
Wu
,
R. M.
Komoll
,
N.
Gekara
,
K. E.
Dittmar
,
S.
Weiss
,
J.
Jablonska
.
2013
.
IFN-γ production by CD27+ NK cells exacerbates Listeria monocytogenes infection in mice by inhibiting granulocyte mobilization.
Eur. J. Immunol.
43
:
2626
2637
.
73
Hashiguchi
,
T.
,
A.
Oyamada
,
K.
Sakuraba
,
K.
Shimoda
,
K. I.
Nakayama
,
Y.
Iwamoto
,
Y.
Yoshikai
,
H.
Yamada
.
2014
.
Tyk2-dependent bystander activation of conventional and nonconventional Th1 cell subsets contributes to innate host defense against Listeria monocytogenes infection.
J. Immunol.
192
:
4739
4747
.
74
Humann
,
J.
,
L. L.
Lenz
.
2010
.
Activation of naive NK cells in response to Listeria monocytogenes requires IL-18 and contact with infected dendritic cells.
J. Immunol.
184
:
5172
5178
.
75
Ma
,
F.
,
S.
Xu
,
X.
Liu
,
Q.
Zhang
,
X.
Xu
,
M.
Liu
,
M.
Hua
,
N.
Li
,
H.
Yao
,
X.
Cao
.
2011
.
The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-γ.
Nat. Immunol.
12
:
861
869
.
76
Zhao
,
D.
,
Q.
Zhang
,
Y.
Liu
,
X.
Li
,
K.
Zhao
,
Y.
Ding
,
Z.
Li
,
Q.
Shen
,
C.
Wang
,
N.
Li
,
X.
Cao
.
2016
.
H3K4me3 demethylase Kdm5a is required for NK cell activation by associating with p50 to suppress SOCS1.
Cell Rep.
15
:
288
299
.
77
Berg
,
R. E.
,
E.
Crossley
,
S.
Murray
,
J.
Forman
.
2005
.
Relative contributions of NK and CD8 T cells to IFN-gamma mediated innate immune protection against Listeria monocytogenes.
J. Immunol.
175
:
1751
1757
.
78
Dunn
,
P. L.
,
R. J.
North
.
1991
.
Early gamma interferon production by natural killer cells is important in defense against murine listeriosis.
Infect. Immun.
59
:
2892
2900
.
79
Perona-Wright
,
G.
,
K.
Mohrs
,
F. M.
Szaba
,
L. W.
Kummer
,
R.
Madan
,
C. L.
Karp
,
L. L.
Johnson
,
S. T.
Smiley
,
M.
Mohrs
.
2009
.
Systemic but not local infections elicit immunosuppressive IL-10 production by natural killer cells.
Cell Host Microbe
6
:
503
512
.
80
Vivier
,
E.
,
S.
Ugolini
.
2009
.
Regulatory natural killer cells: new players in the IL-10 anti-inflammatory response.
Cell Host Microbe
6
:
493
495
.

The authors have no financial conflicts of interest.

This article is distributed under The American Association of Immunologists, Inc., Reuse Terms and Conditions for Author Choice articles.

Supplementary data