Introduction

Natural killer (NK) cells are large globular lymphocytes that represent our innate immune response against virally-infected or transformed cells1,2. After NK cells recognize tumor cells, NK cell receptors are activated, which likely aids the formation of an immunological synapse, towards which cytolytic granules containing perforin and granzymes and the microtubule organizing center of NK cells are polarized3,4. After the cytolytic granules fuse with the plasma membrane through the degranulation process, the secreted perforin forms pores in the plasma membrane of the tumor cells. Serine protease granzyme B enters tumor cells through perforin and induces caspase-dependent and independent apoptotic cancer cell death5,6. Intracellular Ca2+ mobilization is required for target cell adhesion, granule polarization and the degranulation process of NK cells, which are necessary in order to display their natural cytotoxicity7. Prior study suggests that cytotoxic lymphocyte degranulation and target cell lysis are Ca2+-dependent through STIM1/ORAI1-mediated calcium influx8. Recently, it has also been reported that exocytotic granules are themselves acidic Ca2+ stores and a more target-specific Ca2+-mobilizing messenger, such as nicotinic acid adenine dinucleotide phosphate (NAADP), has been identified as being critical for the release of Ca2+ from exocytolytic granules via their cognate two-pore channels (TPCs), leading to cytolytic activity in cytotoxic T lymphocytes (CTLs)9. However, the precise mechanism by which Ca2+ signals interplay in cytolytic granule exocytosis and the killing of NK cells has remained unclear.

Transient receptor potential melastatin 2 (TRPM2) is a Ca2+-permeable nonselective cation channel localized at the lysosomal membrane as well as the plasma membrane10,11,12,13,14,15,16 and TRPM2-mediated Ca2+ signaling is involved in innate immunity17. TRPM2 channels are opened through the binding of intracellular ADP-ribose (ADPR) and can be synergistically activated by the presence of cyclic ADP-ribose (cADPR), NAADP, hydrogen peroxide (H2O2) and Ca2+ 18,19,20,21.

CD38 is a multifunctional enzyme that catalyzes the synthesis of Ca2+-mobilizing second messengers, cADPR and NAADP, from β-nicotinamide adenine dinucleotide (β-NAD+) and its phosphate form (β-NADP+), respectively22,23,24. NAADP and cADPR are further converted to ADP-ribose 2′-phosphate and ADPR, respectively22,23,25. CD38 has long been known to trigger cytotoxic responses and release granzymes in activated NK cells26, but the precise mechanisms by which CD38 mediates cytolytic activity have remained obscure. Interleukin 2 (IL-2)-activated NK cells are more lytic to target cells than resting NK cells, suggesting that IL-2 induces the de novo expression of proteins that act between CD38 and the lytic machinery in NK cells27.

In this study, we explored the possibility that ADPR may affect the antitumor effects of NK cells by modulating [Ca2+] via the TRPM2 channel. We have identified a novel mechanism for antitumor function of NK cells, in which ADPR produced by CD38 and TRPM2-dependent Ca2+ release from acidic Ca2+ stores result in cytolytic granule polarization and degranulation. These findings may help to better understand the regulation of NK cell cytotoxicity and offer a therapeutic strategy for enhancing the antitumor function of NK cells.

Results

NK cells from TRPM2-deficient mice have an intrinsic defect in antitumor activity

To evaluate the possibility that TRPM2-mediated Ca2+ signaling is required for the antitumor effector function of NK cells, we first examined the tumor-induced Ca2+ change in NK cells from TRPM2+/+ and TRPM2−/− mice. We noticed robust Ca2+ signals in both TRPM2+/+ and TRPM2−/− NK cells upon contact with B16F10 cells, a melanoma tumor cell line. However, TRPM2+/+ NK cells were distinct from TRPM2−/− NK cells in their ability to sustain the Ca2+ signals. TRPM2+/+ NK cells exhibited a rapid initial increase, after which the elevated levels remained for the duration of our measurement (500 s). In contrast, TRPM2−/− NK cells were not able to sustain the initial intracellular [Ca2+] ([Ca2+]i) rise (Fig. 1a; 31.5% of area under curve (AUC) of Ca2+ trace in TRPM2+/+ NK cells). As [Ca2+]i increase is important to the degranulation of NK cells7, we investigated whether TRPM2 was involved in degranulation activity. We compared the surface expression of CD107a in TRPM2+/+ and TRPM2−/− NK cells following stimulation with B16F10. B16F10-induced degranulation was absent in TRPM2−/− NK cells (Fig. 1b). Tumor cell-induced granzyme B secretion and cytotoxicity were also completely absent in TRPM2−/− NK cells (Fig. 1c and d). We further evaluated the ability of TRPM2 to protect against B16F10 tumor growth in vivo. Consistent with our finding that TRPM2−/− NK cells have decreased cytolytic activity (Fig. 1d), we observed a defect in the ability of TRPM2 deficient mice to control B16F10 tumor formation and their consequent survival rates (Fig. 1e and f). These results suggest that TRPM2 channel-mediated Ca2+ signals are critical for the antitumor effect of NK cells.

Figure 1
figure 1

Critical role of TRPM2 for cytotoxicity of NK cells against tumor cells via Ca2+-dependent degranulation.

(a) Tumor cell-induced Ca2+ signals in NK cells from TRPM2+/+ or TRPM2−/− mice. Arrow indicates the time of addition of B16F10 tumor cells. Data are mean ± SEM of three independent experiments. *P < 0.001 vs basal; #P < 0.05. (b) Impairment of degranulation in TRPM2−/− NK cells upon stimulation with B16F10 cells. TRPM2+/+ or TRPM2−/− NK cells were stimulated with target cells for 2 h at 37°C and then stained with FITC-conjugated anti-CD107a mAb and PE-conjugated anti-NK1.1 mAb. NK cells were gated on forward scatter/side scatter characteristics. (c) Reduced granzyme B release in tumor cell-stimulated TRPM2−/− NK cells. The amount of granzyme B released into the media was measured by ELISA after incubation with NK cells and B16F10 cells for 30 min. (d) Decrease in cytolytic activity against B16F10 cells in TRPM2−/− NK cells. TRPM2+/+ or TRPM2−/− NK cells were assessed for cytolytic activity against B16F10 target cells in a 4-h calcein-release assay. Data shown in b, c and d are representative of three independent experiments. *P < 0.001. (e) B16F10 cells (1 × 105) were injected into the flanks of TRPM2+/+ and TRPM2−/− mice (n = 10 per cohort) and tumor growth was monitored. *P < 0.001. (f) Kaplan-Meier plot of TRPM2+/+ and TRPM2−/− recipient mice after s.c. injection with B16F10 cells (1 × 105) (n = 10 per cohort). (g) Antitumor effect of TRPM2+/+ or TRPM2−/− NK cells in the lung metastasis model of B16F10 cells. TRPM2−/− mice (n = 4 per cohort) were injected i.v. with 1 × 105 B16F10 melanoma cells. IL-2 activated NK cells (2 × 106 cells) from TRPM2+/+ or TRPM2−/− mice were injected i.v. into B16F10-bearing TRPM2−/− mice 2 d later. Lungs were harvested 14 d after and fixed with 4% paraformaldehyde and metastatic nodules were counted. Representative images of lungs are shown. *P < 0.01.

To assess whether the defect in B16F10 tumor control was due to NK cell dysfunction, we adoptively transferred IL-2 activated NK cells from TRPM2+/+ and TRPM2−/− mice into B16F10 tumor-bearing TRPM2−/− mice. Following B16F10 tumor cell injection, a markedly higher number of metastatic cells were observed in TRPM2−/− NK cell-treated mice than in TRPM2+/+ NK cell-treated mice (Fig. 1g). These findings suggest that TRPM2-deficient NK cells have an intrinsic defect in antitumor activity.

CD38 is associated with the granule polarization and degranulation process of NK cells

The TRPM2 channel can be synergistically opened by various Ca2+-mobilizing second messengers, such as ADPR, cADPR, or NAADP, all of which are produced by CD3818,19,23,24. NK cell-mediated tumor clearance is composed of target cell adhesion, granule polarization and degranulation and CD38 is known to trigger cytotoxic responses as a receptor in NK cells3,26. Therefore, we further examined whether mouse Cd38+/+ NK cells and Cd38−/− NK cells differed in their ability to induce tumor cell adhesion when co-cultured with B16F10 cells. Time-dependent increases in the number of conjugates between NK and tumor cells were observed in both Cd38+/+ and Cd38−/− NK cells with no significant differences (Fig. 2a).

Figure 2
figure 2

Tumor cell-induced NK cell granule polarization and degranulation requires CD38-mediated Ca2+ signals.

(a) Cd38+/+ or Cd38−/− NK cells stimulated with B16F10 tumor cells. The percentage of conjugation formation at indicated time was analyzed by flow cytometry. Data are mean ± SEM of three independent experiments. (b) Cd38−/− NK cells were inhibited tumor-triggered translocation of perforin and granzyme B towards immunological synapses. Cd38+/+ or Cd38−/− NK cells were stimulated with B16F10 cells at 1:1 ratio at 37°C for 20 min and then stained with perforin or granzyme B. The dashed line indicates the immunological synapse. (Scale bar, 5 μm). All images were representative of at least three independent experiments. (c) Degranulation of Cd38+/+ or Cd38−/− NK cells upon stimulation with B16F10 cells for 2 h at 37°C. Shown is a representative of three independent experiments. (d and e) Ca2+ signals in Cd38+/+ or Cd38−/− NK cells upon stimulation with B16F10 cells. NK cells were loaded with Fluo-4 AM for 40 min at 37°C and Ca2+ levels in NK cells were measured following treatment with B16F10 cells labeled with cell tracker orange CMRA (red). Arrow indicates the time of addition of B16F10 cells. (Scale bars, 10 μm). Shown is a representative of three independent experiments.

We next investigated whether tumor cell-induced NK cell granule polarization was affected by CD38 by analyzing the translocation of cytolytic granules containing perforin and granzyme B. In the absence of target cells, perforin and granzyme B displayed dispersed granules in NK cells. Upon conjugation with B16F10, perforin and granzyme B were translocated towards the immunological synapse area between Cd38+/+ NK cells and B16F10 cells. However, such tumor cell-induced translocation of perforin and granzyme B was not observed in Cd38−/− NK cells (Fig. 2b). To address the question of whether the degranulation activity of NK cells was affected by CD38, we compared the surface expression of CD107a in Cd38+/+ and Cd38−/− NK cells following stimulation with B16F10. B16F10 induced strong degranulation in Cd38+/+ NK cells but not in Cd38−/− NK cells (Fig. 2c). These results demonstrate that CD38 is involved in the granule polarization and cytolytic degranulation process of NK cell during contact with tumor cells, but not in tumor cell adhesion.

We next investigated whether the tumor-induced [Ca2+]i increase was dependent on CD38. Similar to TRPM2−/− NK cells, Cd38−/− NK cells displayed the tumor-induced initial rise in [Ca2+]i, but failed to maintain the elevated levels (Fig. 2d and e; 36.1% of AUC of Ca2+ trace in Cd38+/+ NK cells). These results demonstrate that CD38-mediated Ca2+ signaling may be involved in the granule polarization and cytolytic degranulation process of NK cell during contact with tumor cells.

Cytolytic granule polarization of NK cells requires ADPR-mediated Ca2+ signaling

We examined whether various Ca2+ signaling messengers produced by CD38 were actually involved in tumor cell-induced Ca2+ signals and degranulation in NK cells. To this end, we tested various inhibitors of the CD38 signaling pathway to observe their effects on tumor-induced degranulation and Ca2+ signals as shown in Fig. 2. Among the inhibitors that we tested (8-Br-ADPR, an antagonistic analog of ADPR; 8-Br-cADPR, a cADPR antagonist; Ned19, an NAADP antagonist; and Xestospongin C (XeC), an IP3 receptor antagonist), only 8-Br-ADPR blocked the late, sustained portion of tumor-induced Ca2+ signals (Fig. 3a) and degranulation (Fig. 3b). The effects of 8-Br-ADPR were confirmed by confocal microscopic findings that 8-Br-ADPR blocked the tumor cell-induced translocation of perforin and granzyme B towards the immunological synapse between B16F10 and NK cells (Fig. 3c).

Figure 3
figure 3

ADPR-mediated Ca2+ signals are required for cytolytic degranulation of NK cells.

(a) 8-Br-ADPR inhibits tumor cell-induced sustained Ca2+ signals in NK cells. NK cells were pretreated with 100 μM 8-Br-ADPR, 100 μM 8-Br-cADPR, 10 μM Ned19, or 2 μM XeC for 20 min. Arrow indicates the time of addition of B16F10 cells. (b) Inhibition of tumor cell-induced NK cell degranulation by 8-Br-ADPR. Data shown in a and b are representative of three independent experiments. *P < 0.001 vs basal; #P < 0.05. (c) 8-Br-ADPR inhibits tumor cell-induced translocation of perforin and granzyme B towards immunological synapses. NK cells were added to B16F10 cells at 1:1 ratio at 37°C for 20 min and stained with perforin or granzyme B. The dashed line indicates the immunological synapse. (Scale bars, 5 μm). All images were representative of at least three independent experiments. (d) NK cells were treated with PME (30 μg) for 20 min and homogenized and fractionated on a continuous 0.4 M to 1.6 M sucrose density gradient using ultracentrifugation. Fractions were analyzed by immunoblotting to identify cellular organelles and identified using antibodies against each marker: early endosome (EEA1), lysosomes (LAMP1), plasma membrane (Na+K+-ATPase). Distributions of granzyme B and perforin were assessed by immunoblotting. Shown is a representative of three independent experiments. The gels have been run under the same experimental conditions. The full-length blots and the cropped blots are presented in Supplementary Figure 2. (e and f) 8-Br-cADPR inhibits tumor-induced granzyme B secretion (e) and cytotoxicity activity (f) of NK cells. 100 μM 8-Br-ADPR, 100 μM 8-Br-cADPR, 10 μM Ned19, or 2 μM XeC was preincubated for 20 min. Data are mean ± SEM of three independent experiments. #P < 0.001.

To confirm the inhibitory effects of 8-Br-ADPR on the translocation of intracellular perforin and granzyme B to the plasma membrane in response to tumor cell contact, we examined the subcellular distributions of perforin and granzyme B by Western blot. Subcellular organelles of NK cells were separated by sucrose density gradient centrifugation. Perforin and granzyme B were enriched in higher density fractions, consisting of plasma membrane from NK cells treated with plasma membrane extracts (PME) from B16F10 cells, compared to those from NK cells that had not been treated (Fig. 3d). 8-Br-ADPR inhibited the tumor-induced translocation of cytoplasmic granular proteins to the plasma membrane. These results are consistent with those in Fig. 3c, showing that intracellular perforin and granzyme B were translocated to the plasma membrane in response to tumor contact. Furthermore, we found that the B16F10 cell-induced granzyme B release from NK cells, as well as the cytolytic activity of NK cells against B16F10 cells, were inhibited by 8-Br-ADPR but not by any other inhibitors we tested (Fig. 3e and f). Taken together, these results suggest that ADPR is a key player in mediating the degranulation and cytolytic activity of NK cells.

Intracellular production of ADPR is necessary for the cytolytic activity of NK cells against tumors and depends on CD38 signaling, which facilitates sustained Ca2+ signals

Because 8-Br-ADPR, an antagonistic analog of ADPR, inhibited the degranulation and cytolytic activity of NK cells, we investigated whether ADPR was endogenously produced in tumor cell-stimulated NK cells and if CD38 was the enzyme responsible for ADPR production. The level of ADPR in Cd38+/+ NK cells increased 2.4 fold when stimulated by PME, which was not observed in Cd38−/− NK cells (Fig. 4a). By contrast, the level of cADPR, another Ca2+ signaling second messenger produced by CD38, was not affected by stimulation with the PME of tumor cells (Fig. 4b). These results show that ADPR is specifically induced by CD38 in response to tumor cells.

Figure 4
figure 4

Tumor-induced ADPR production by intracellular CD38 is responsible for sustained Ca2+ signals in NK cells.

(a and b) Tumor cells induce ADPR production but cADPR in Cd38+/+ NK cells. Levels of ADPR and cADPR were determined after treatment of NK cells with 30 μg PME prepared from B16F10 tumor cells for 40 s. Data are mean ± SEM of three independent experiments. *P < 0.001; #P < 0.05. ns, not significant. (c–e) Cibacron blue 3GA (CB) inhibits of tumor-stimulated ADPR induction (c), sustained Ca2+ signal (d), cytotoxicity (e). CB (100 μM), ara-2′-F-NAD (200 nM), bafilomycin A1 (200 nM) or GPN (50 μM) was preincubated for 30 min. Data shown in c–e are representative of three independent experiments. *P < 0.001 vs basal; #P < 0.05.

CD38 is primarily located at two sites, the plasma membrane and intracellular organelles28,29,30,31, Therefore, we investigated the involvement of intracellular and extracellular CD38 in inducing ADPR by testing two CD38 inhibitors13,32, Cibacron blue 3GA (a cell-permeable CD38 inhibitor) and ara-2′-F-NAD (a cell-impermeable CD38 inhibitor). The former inhibited tumor cell-stimulated ADPR production, sustained Ca2+ increase and cytotoxicity, whereas the latter did not (Fig. 4c–e), indicating that CD38 in intracellular organelles is responsible for the ADPR production in response to tumor PME stimulation. Interestingly, lysosomal disrupting reagents such as bafilomycin A1 (Baf A1), an inhibitor of vacuolar-type H+-ATPase that prevents the re-acidification of acidic organelles, or glycyl-phenylalanine-2-naphthylamide (GPN), a reagent that osmotically ruptures lysosomes, also inhibited NK cells from inducing ADPR production in response to tumor PME stimulation (Fig. 4c).

Tumor-induced ADPR production was abolished in media devoid of extracellular Ca2+, suggesting that the initial transient Ca2+ influx was a central upstream pathway for the induction of ADPR. Cibacron blue 3GA, Baf A1, as well as extracellular Ca2+-free conditions, which prevent NK cells from inducing ADPR production and maintaining an elevated [Ca2+]i in response to tumor cells, diminished the cytolytic activity of NK cells against B16F10 cells (Fig. 4c–e). This suggests that tumor cell-induced production of ADPR, which is necessary for the cytolytic activity of NK cells, depends on the initial transient Ca2+ influx, followed by sustained Ca2+ concentration achieved by CD38 signaling.

Tumor-induced ADPR production depends on PKA activation

It has been demonstrated that ADPR-cyclases, including CD38, are activated by PKA or PKG29,33,34,35. To elucidate the upstream signaling pathways of ADPR production by CD38 in tumor-induced NK cell activation, we determined [ADPR]i in NK cells following treatments with various agonistic and antagonistic agents. Tumor cell-induced ADPR production was inhibited by Rp-8-Br-cAMPS (a membrane-permeable PKA inhibitor), but not Rp-8-pCPT-cGMPS (a membrane-permeable PKG inhibitor). Moreover, N6-Benzoyl (a membrane-permeable PKA activator) significantly increased [ADPR]i, but this increase was inhibited in Ca2+-free conditions (Fig. 5a). These findings suggest that the initial transient Ca2+ influx and cAMP/PKA are required for CD38 activation in tumor-induced ADPR production. We further examined whether cAMP was produced in tumor cell-stimulated NK cells. cAMP was produced after treating NK cells with PME, regardless of extracellular Ca2+ levels (Fig. 5b), suggesting that tumor-induced cAMP production is independent of initial transient Ca2+ influx. Furthermore, the tumor cell-induced sustained [Ca2+]i increase was inhibited by Rp-8-Br-cAMPS or SQ 22536 (an adenylyl cyclase inhibitor) (Fig. 5c). These findings suggest that both the initial Ca2+ influx and cAMP/PKA are required in order for CD38 activation to produce ADPR in tumor-induced NK cells.

Figure 5
figure 5

Tumor cells produce ADPR by activation of PKA but not PKG in NK cells.

(a) ADPR levels were determined after treatment of NK cells with 30 μg PME for 40 s. Rp-8-Br-cAMPS (100 μM) or Rp-8-pCPT-cGMPS (20 μM) was preincubated for 30 min. 100 μM N6-benzoyl-cAMP (PKA activator, 40 s) was used for ADPR measurement. Data are mean ± SEM of three independent experiments. *P < 0.001 vs basal; #P < 0.05. (b) cAMP levels were determined after treatment of NK cells with 30 μg PME. Data are mean ± SEM of three independent experiments. (c) Inhibition of tumor cell-induced sustained Ca2+ increase by a PKA inhibitor, Rp-8-Br-cAMPS and an adenylate cyclase inhibitor, SQ 22536. Rp-8-Br-cAMPS (100 μM) or SQ 22536 (250 μM) was preincubated for 30 min. (d) SOCE induced by bafilomycin A1. Bafilomycin A1 (1 μM)-induced SOCE was inhibited with 50 μM SK96365 but not by 20 μM ACA. SK96365 or ACA was pre-incubated for 30 min. Data shown in c and d are mean ± SEM of three independent experiments. n = 10.

To test the contribution of store-operated Ca2+ entry (SOCE) to the tumor cell–induced sustained Ca2+ signal, we used Baf A1 to deplete Ca2+ from acidic organelles, which are the Ca2+ stores responsible for tumor cell-mediated Ca2+ signals stimulated by ADPR (Fig. 4c and d). Depletion of Ca2+ stores by Baf A1 induced Ca2+ entry after Ca2+ was added to the bath perfusion solution, as described earlier36 (Fig. 5d). This SOCE was completely blocked by SK96365, a SOCE blocker, but not by ACA, a specific TRPM2 channel blocker. These findings indicate that the sustained Ca2+ signal is attributable to SOCE following Ca2+ mobilization by ADPR and that TRPM2 is not involved in ADPR-mediated SOCE.

TRPM2 is required for the migration of perforin-containing granules towards the immunological synapse as NK cells encounter tumor cells

Because ADPR is known to activate TRPM2 in order to elicit Ca2+ release from intracellular stores15,16, we focused on studying TRPM2 to explore its possible role in perforin exocytosis. To this end, we first examined whether CD38 co-localized with TRPM2 in NK cells. CD38 and TRPM2 were dispersed at different locations in non-stimulated NK cells. However, contact with B16F10 cells changed the locations of CD38 and TRPM2, concentrating them at the immunological synapse formed between NK cells and B16F10 cells (Fig. 6a). Furthermore, CD38 and TRPM2 proteins appeared to physically interact only after NK cells were stimulated by B16F10 cells, as judged by the co-immunoprecipitation of TRPM2 with CD38 (Fig. 6b). These results suggest that the formation of ADPR by CD38 in NK cells leads to activation of TRPM2 channels, due to the proximity of TRPM2 and CD38.

Figure 6
figure 6

Tumor cell increases the interaction of CD38 with TRPM2 in NK cells.

(a) Increased co-localization of CD38 with TRPM2 in NK cells upon tumor cell treatment. NK cells were incubated with B16F10 cells for 20 min and then stained with CD38 and TRPM2. (b) Tumor cell PME increases association of CD38 with TRPM2 in NK cells. NK cells were treated with PME for 15 s. Cells were extracted with a lysis buffer and then subjected to immunoprecipitation (IP) using an anti-CD38 mAb. Immunoprecipitated proteins were analyzed by western blotting (WB) with anti-CD38 pAb or anti-TRPM2 pAb. The gels have been run under the same experimental conditions. The full-length blots and the cropped blot are presented in Supplementary Figure 3. (c) Defect of tumor cell-stimulated translocation of perforin towards immunological synapses in TRPM2−/− NK cells. TRPM2+/+ or TRPM2−/− NK cells were added to B16F10 target cells at 1:1 ratio at 37°C for 20 min and then stained with perforin and TRPM2. DAPI was used as a nuclear stain (blue). The dashed line indicates the immunological synapse. (Scale bar, 5 μm). All images were representative of at least three independent experiments.

Similar to the findings observed with CD38 and TRPM2 in NK cells, perforin was scattered inside the NK cells mostly inside cytolytic granules and co-localizing with TRPM2 even before stimulation with tumor cells (Fig. 6c, upper panel). Like CD38, perforin co-migrated with TRPM2 to the immunological synapse upon stimulation with B16F10 cells (Fig. 6c, middle panel). Such tumor cell-induced perforin migration was not detected in TRPM2−/− NK cells (Fig. 6c, lower panel), suggesting that TRPM2 was essential for perforin migration towards the immunological synapse. The finding that perforin migration towards the synapse requires CD38 as well (Fig. 2b) suggests that both CD38 and TRPM2 are absolutely essential for the migration of perforin (or possibly cytolytic granules as well) toward the immunological synapse when NK cells encounter tumor cells.

Adoptive transfer of Cd38+/+ NK cells, but not Cd38−/− NK cells, controls B16F10 melanoma lung metastasis

Based on the above findings, we extended our study to the B16F10 experimental metastasis model in C57BL/6 mouse to test whether the adoptive transfer of NK cells to mice with B16F10 cell metastasis could reduce the number of tumor nodules and increase survival rates. Indeed, Cd38+/+ NK cells significantly reduced the number of metastasized B16F10 cells in the lung, which did not hold true for Cd38−/− NK cells, indicating that CD38 was essential for the antitumor activity of NK cells (Fig. 7a). In vitro cytotoxic assays demonstrated that the cytolytic activity of Cd38+/+ NK cells was superior to that of Cd38−/− NK cells against not only B16F10 but also YAC-1 and EL4 murine lymphoma cell lines (Fig. 7b), indicating that Cd38+/+ NK cells exhibited a broad spectrum of natural cytotoxicity. Both Cd38+/+ and Cd38−/− NK cells expressed equivalent amounts of perforin and granzyme B mRNA and protein levels (Fig. 7c and d). However, B16F10-induced secretion of granzyme B was lower in CD38−/− NK cells than CD38++ NK cells (Fig. 7e). These results suggest that the enhancing effect of CD38 on the cytolytic activity of NK cells results from the stimulated release of granzyme B to target cells.

Figure 7
figure 7

CD38 is critical for the antitumor effects of NK cells via cytolytic granule exocytosis.

(a) Antitumor effects of Cd38+/+ and Cd38−/− NK cells in a lung metastasis model of B16F10 cells. C57BL/6 mice (n = 4 per cohort) were injected i.v. with 1 × 105 B16F10 melanoma cells. After 2 d, IL-2 activated NK cells (2 × 106 cells) from Cd38+/+ or Cd38−/− mice were injected i.v. into B16F10-bearing wild-type mice. Lungs were harvested 14 d later and metastatic nodules were counted. nd, not detected. Data are mean ± SEM of three independent experiments. Representative images of lungs are shown. *P < 0.001. (b) Comparison of cytolytic activity of Cd38+/+ and Cd38−/− NK cells against various target cell lines. Cd38+/+ or Cd38−/− NK cells cultured for 10 d were assessed for cytolytic activity as indicated, over 4 h. Data are mean ± SEM of three independent experiments. *P < 0.001; #P < 0.05. (c and d) Cd38+/+ and Cd38−/− NK cells are expressed equivalent amounts of perforin and granzyme B at mRNA (c) and protein levels (d). The gels have been run under the same experimental conditions. The full-length blots and the cropped blots are presented in Supplementary Figure 4. Data are mean ± SEM of three independent experiments. ns, not significant. (e) Comparison of the amount of granzyme B released into the media by Cd38+/+ and Cd38−/− NK cells on stimulation with B16F10 cells for 30 min. Data are mean ± SEM of three independent experiments. *P < 0.001 vs basal; #P < 0.05.

Discussion

The cytolytic activity of NK cells requires an initial Ca2+ influx for conjugation with the tumor cells and intracellular Ca2+ mobilization is required for granule polarization and the degranulation process7. However, the activation processes that lead to the polarized degranulation of NK cells are not well understood. We have demonstrated here that cytolytic granule exocytosis and the antitumor functions of NK cells rely upon signaling pathways involving CD38, ADPR and TRPM2 and Ca2+ release from acidic stores. The inhibition of this signaling pathway significantly decreased the cytolytic granule exocytosis and antitumor activity in both in vitro and in vivo models.

The role of CD38 as an adhesion molecule in NK cell antitumor function has been demonstrated in earlier studies26. However, the proposed function of CD38 as an adhesion molecule is not likely, as evidenced by the lack of difference in the formation of conjugates between the tumor and Cd38+/+ and Cd38−/− NK cells (Fig. 2a). Our data reveal that the production of ADPR by CD38 in response to tumor cells was ablated in Cd38−/− NK cells (Fig. 4a), indicating that CD38 was responsible for the production of ADPR in response to tumor cells. Intriguingly, cADPR has been known as a Ca2+ signaling second messenger involved in IL8-induced chemotaxis in lymphokine activate killer cells35. Our data shown in Fig. 4b, which showed no significant increase of cADPR in tumor stimulated NK cells, rule out the possibility that ADPR is produced from cADPR, another one of CD38's products. This indicates that the interaction with tumor cells through cognate receptors on NK cells transmits signals for the activation of CD38's NAD glycohydrolase (NADase) to produce ADPR. Further studies will be required to identify the mechanisms regarding how specific enzymatic activities of CD38 are selectively activated under specific situations.

We found that the sustained Ca2+ signaling, mediated by ADPR and TRPM2, was correlated with polarized degranulation and cytolytic activity of NK cells against the melanoma tumor cell line B16F10. Furthermore, we have shown the importance of acidic environments for CD38 activity in NK cells, because the presence vacuolar H+ pump inhibitors resulted in the failure of ADPR production, Ca2+ mobilization and cytolytic activity induced by CD38. All the CD38 activities in NK cells we described depend exclusively on the tumors contacted or the PME prepared from tumor cells. Finally, our results unveiled the critical role of TRPM2 in NK cell cytotoxicity against tumor cells when NK cells were introduced to mice bearing B16F10 cells as passive immunotherapy.

TRPM2 is expressed in the plasma membranes as well as in intracellular compartments. The functions of intracellular TRPM2 have been studied only on insulin secretion in pancreatic β cells15 and the maturation and chemotaxis of dendritic cells16. We show that TRPM2 is an ion channel located in the cytolytic granule and co-migrates with CD38 to the immunological synapse upon tumor stimulation (Fig. 6). Like CD38, TRPM2 appears to be essential for sustained Ca2+ signals, directed degranulation and cytolytic activity, suggesting that CD38 is tightly coupled to TRPM2 ion channel in inducing sustained Ca2+ signals, which may be pre-requisite for the directed migration of cytolytic granules to the immunological synapses. Therefore, cytolytic granules contain all the necessary machinery for Ca2+ signaling, including Ca2+ stores and exocytosis capabilities. The lack of either CD38 or TRPM2 results in a significant increase of tumor formation and reduced survival rates. Thus, CD38 and TRPM2 cooperate to generate the sustained Ca2+ signal and directed migration of cytolytic granules to the immunological synapse in response to tumor stimulation.

Our data show that the majority of TRPM2 is not expressed in the plasma membrane, but is localized in intracellular granules (Fig. 6). Although our data (Fig. 5d) show that TRPM2 is not involved in SOCE, we cannot rule out a possibility that the granular TRPM2 can gain access to external Ca2+ only after degranulation and fusion of the granules with the plasma membrane. To clarify the cause-effect relationship between translocation of TRPM2 and Ca2+ influx, we compared the time course of the tumor cell-induced translocation of TRPM2 and Ca2+ influx at 5 min after stimulation with B16F10 cells and found that TRPM2 remained at intracellular granules (Supplementary Figure S1) when the tumor cell-induced Ca2+ influx occurred (Fig. 1a and 2e). These findings suggest that Ca2+ influx precedes degranulation and that the sustained Ca2+ signal is required for degranulation. Taken together, our data indicate that an ion channel other than TRPM2 is responsible for ADPR-mediated SOCE.

In CTLs, TCR activation recruits NAADP to activate TPC channels present on cytolytic granules9. These cytolytic granules store and release Ca2+, although SOCE through the Orai1-STIM1 complex is necessary for lytic granule exocytosis and tumor cell killing8. Our data show that NAADP is not involved in tumor-induced cytolytic degranulation in NK cells (Fig. 3). Thus, different cells use different Ca2+ signaling messengers for similar cellular events and such interesting mechanisms remain to be determined.

As described, polarized cytolytic degranulation towards the immunological synapse is likely guided by specific Ca2+ channels. Our data affirm a key role of the CD38/ADPR/TRPM2 pathway in generating polarized cytolytic degranulation towards the immunological synapses via Ca2+ signals from cytolytic granules that subsequently lead to target cell killing. We found that the sustained Ca2+ signaling was a pre-requisite for polarized degranulation and lysis of the target cell in vitro. TRPM2 serves to couple CD38/ADPR activation with sustained Ca2+ signals and contributes to potentiating NK cell cytotoxicity against tumor cells in the mouse model. Understanding how suitable and specific Ca2+ mobilization signals drive the polarized migration of cytolytic proteins towards the immunological synapse to kill target cells is important. This study highlights the selective role of CD38 in stimulating directional exocytosis, which is crucial for target cell killing and may help to design better immunotherapy through the use of NK cells.

In conclusion, we demonstrate that ADPR is produced by CD38, localized in cytolytic granules, upon tumor stimulation. ADPR modulates Ca2+ signaling by gating TRPM2 channels and causes cytolytic polarized degranulation from NK cells. These data constitute a novel mechanism of Ca2+ signaling for the antitumor functions of NK cells and may provide an antitumor therapeutic strategy utilizing increased NK cell cytotoxicity.

Methods

Reagents

Antibodies were obtained as follows: granzyme B pAb from Cell Signaling (Danvers, MA) and perforin pAb from Santa Cruz Biotechnology (Santa Cruz, CA); anti-perforin mAb, DyLight 488 anti-perforin mAb and DyLight 550 anti-TRPM2 pAb from Novus Biologicals (Littleton, CO). Human recombinant IL-2 was from Chiron BV (Amsterdam, Netherlands) and Xestospongin C was from Santa Cruz Biotechnology (Santa Cruz, CA). 8-Br-ADPR, ara-2′-F-NAD and N-(p-amylcinnamoyl) anthranilic acid (ACA) were from Biolog Life Science Institute (Bremen, Germany). All other reagents were obtained from Sigma-Aldrich (St. Louis, MO).

Mice

Cd38−/− mice (B6.129P2-Cd38tm/Lud) were purchased from Jackson Laboratory (Bar Harbor, ME). TRPM2−/− mice were kindly provided by Mori (Kyoto University, Japan). Mice were bred and housed in the facilities of Chonbuk National University Medical School under specific pathogen free conditions. All experimental animals were used under a protocol approved by the institutional animal care and user committee of the Chonbuk National University Medical School (CBU 2014-00031). Standard guidelines for laboratory animal care were performed in accordance with the Guide for the care and use of laboratory animals published by the National Institutes of Health37.

Cell culture

Mouse NK cells were isolated by negative immunomagnetic selection (Stem cell Technologies). The cells were cultured in a 5% CO2 incubator at 37°C in culture media containing 5000 IU/ml IL-2 for 10 d. The culture medium used was RPMI-1640 supplemented with 10% FBS, 0.25 μg/ml amphotericin B, 10 U/ml penicillin G, 100 μg/ml streptomycin, 1 mM L-glutamine, 1% nonessential amino acids (Invitrogen) and 50 μM 2-mercaptoethanol. Purity of NK cells was determined by FACS analysis. The murine melanoma B16F10 and lymphoma EL4 and YAC-1 cells were purchased from ATCC (Manassas, VA).

Animal model

To establish tumor in the flanks of C57BL/6 and TRPM2−/− mice, tumor cells were resuspended in 200 μl of PBS and injected subcutaneously. The tumor developed at the site of injection was measured and the mean tumor size was calculated [length (cm) × width (cm)]. To produce experimental lung metastasis, C57BL/6 mice received intravenous injections of B16F10 cells (1 × 105 cells/mouse). After 2 d, NK cells (2 × 106 cells/mouse) from Cd38+/+, Cd38−/−, or TRPM2−/− mice were injected intravenously into wild-type or TRPM2−/− mice. Mice were sacrificed 14 d later. Lungs were fixed with 4% paraformaldehyde and metastatic nodules on the surface of the lungs were counted.

Calcium imaging

NK cells were loaded with 5 μM Fluo-4 AM (Invitrogen) in HBSS at 37°C for 40 min and changes in [Ca2+]i were determined at 488 nm excitation/530 nm emission wavelengths using a confocal microscope (Nikon). For calculation of [Ca2+]i, the method of Tsien et al.38 was used with the following equation: [Ca2+]i = Kd(FFmin)/(FmaxF), where Kd is 335 nM for Fluo-4 and F is the observed fluorescence. Each tracing was calibrated for maximal intensity (Fmax) by addition of ionomycin (8 μM) and for minimal intensity (Fmin) by addition of EGTA 50 mM after each measurement.

Measurement of [ADPR]i

ADPR level was measured using LC-MS/MS as described previously39. Briefly, cells were treated with 5% trichloroacetic acid under sonication and precipitates were removed by centrifugation at 20,000 × g for 10 min. Supernatants were loaded onto a Waters ACQUITY UPLC system coupled to a Waters Xevo TQ-S mass spectrometer and separated using a BEH Amide column (Waters ACQUITY UPLC BEH Amide, 130 Å, 1.7 μm, 2.1 mm × 50 mm). The column was equilibrated with 100% buffer B (90% acetonitrile/10% 50 mM ammonium formate) and eluted with a 5-min gradient to 60% buffer A (10 mM ammonium formate in water) at a flow rate of 0.5 ml/min. The following parameters were used for ADPR MS analysis: cone gas, 150 l/h; nebulizer, 7 Bar; and desolvation temperature, 350°C. Ion transitions used for confirmation and quantification of ADPR were m/z 558.17→346.01.

Measurement of [cADPR]i

cADPR levels were measured with a previously described cycling method40. Increases in resorufin fluorescence were measured at 544 nm excitation/590 nm emission wavelengths using a SpectraMax Gemini fluorescence plate reader (Molecular Devices Corp.).

Measurement of [cAMP]i

cAMP levels were determined by a cAMP immunoassay kit according to the manufacturer's protocol (Enzo Life Science). NK cells were preincubated with 0.5 mM isobutylmethylxanthine, a phosphodiesterase inhibitor. After incubation with PME for the indicated times, the cells were lysed in 0.1 M HCl to stop the reaction. After centrifugation at 20,000 × g for 10 min at 4°C, supernatants were collected and acetylated and immunoassays were performed.

Quantitative RT-PCR

Total RNA was isolated from NK cells using an RNeasy Mini Kit (Qiagen, Valencia, CA). cDNA was synthesized by reverse transcription from 50 ng total RNA using a cDNA Reverse Transcriptase Kit (TaKaRa, Japan). The PCR reaction was carried out in 384-well plate using the ABI Prism 7900HT Sequence Detection System (Applied Biosystems). Real-time PCR primers for perforin, granzyme B and GAPDH were as follows: perforin (forward, 5′-AGCACAAGTTCGTGCCAGG-3′ and reverse, 5′-GCGTCTCTCATTAGGGAGTTTTT-3′); granzyme B (forward, 5′-CCACTCTCGACCCTACATGG-3′ and reverse, 5′-GGCCCCCAAAGTGACATTTATT-3′); GAPDH (forward, 5′-CATGGCCTTCCGTGTTCCTA-3′ and reverse, 5′-ATGCCTGCTTCACCACCTTCT-3′).

Formation of conjugation

NK cells were stained with PKH-26 (red) and B16F10 cells were stained with CellTrace CFSE (green) according to the manufacturer's instructions. Effector and target cells (E/T ratio: 1/1) were co-cultured at 37°C. At the time point, the samples were fixed with ice-cold 0.5% paraformaldehyde. The double-positive population was assessed using flow cytometry (FACSAria).

Degranulation assay

NK cell degranulation was assessed as previously described41. Briefly, 2 × 105 NK cells were added to 2 × 105 target cells in 200 μl of complete medium. Cells were mixed, centrifuged for 3 min at 20 × g and incubated for 2 h at 37°C. Thereafter, cells were centrifuged, stained with fluorochrome-conjugated mAbs against NK1.1 and CD107a (BD Biosciences) in PBS supplemented with 2% FBS and 2 mM EDTA for 45 min on ice, washed, re-suspended in PBS supplemented with 2% FBS and 2 mM EDTA and analyzed by flow cytometry. Data were analyzed with FlowJo version10 software.

Cytokine measurement

NK cells were added to 4 × 105 B16F10 cells in 200 μl complete medium. Cells were incubated for 30 min at 37°C in a 5% CO2 incubator. Thereafter, supernatants were collected and cytokines were assayed with ELIZA kits from eBioscience (granzyme B) according to the instructions of the manufacturer.

Cytotoxicity

NK cell cytotoxicity was assessed as previously described42. Target (T) cells labeled with 5 μM calcein AM (Invitrogen) for 1 h at 37°C and effector (E) cells were co-incubated with 200 μl complete medium in a 96 well plate for 4 h at 37°C. Spontaneous release of calcein was determined by incubating loaded target cells in medium alone and maximal release was determined by adding 0.1% Triton X-100 to lyse all target cells. After incubation, plates were centrifuged at 300 × g for 5 min and 100 μl of supernatant from each sample was transferred to a 96-well plate. Fluorescence was measured at 480 nm excitation/538 nm emission wavelengths. Cytotoxicity, measured as a percent specific release of calcein, was calculated using the following formula: percent specific release = (experimental release - spontaneous release)/(maximum release - spontaneous release) × 100.

Immunoprecipitation and immunoblotting

Immunoprecipitation and immunoblotting were carried out as described previously29.

Plasma membrane preparation and subcellular fractionation

For preparation of B16F10 cell plasma membranes, cells were homogenized with a Dounce homogenizer in 0.5 ml of homogenate buffer (0.25 M sucrose, 0.5 mM EDTA, 10 mM HEPES, pH 7.4 and 1 mM PMSF). The homogenate was centrifuged at 1000 × g for 10 min at 4°C, supernatants were centrifuged at 20,000 × g for 20 min at 4°C and then supernatants were ultracentrifuged at 100,000 × g for 2 h at 4°C. The pellet was washed with ice-cold PBS and then used as plasma membrane. NK cells were homogenized as above and the homogenate was subcellular fractionated as described previously29.

Confocal imaging

NK cells were mixed with B16F10 cells at 1:1 ratio at 37°C for 20 min, fixed with ice-cold methanol for 10 min and then washed with ice-cold PBS. After blocking with 3% BSA, 0.25% Triton X-100 and PBS at RT for 1 h, samples were incubated with DyLight 488 anti-perforin mAb (1:200) or rabbit anti-granzyme B pAb (1:200) followed by Alexa Flour 546-conjugated donkey anti-rabbit antibody (1:200, Invitrogen) in the presence of 1% BSA at RT for 1 h. Cells were visualized with a Zeiss LSM510 Axiovert 200 M laser-scanning confocal microscope. The images were obtained using 63× Zeiss Plan-Aprochromat objective and LSM510 (version 7.1). To check for co-localization of CD38 and TRPM2, NK cells were incubated with B16F10 cells for indicated time periods and fixed with ice-cold methanol for 10 min. After blocking with 3% BSA, 0.25% Triton X-100 and PBS at RT for 1 h, samples were incubated with goat anti-CD38 pAb (1:100, Santa Cruz Biotechnology) followed by Alexa Flour 488-conjugated donkey anti-goat antibody (1:200, Invitrogen) and DyLight 550 anti-TRPM2 (1:100, Abcam) pAb at RT for 1 h.

Statistical analysis

Data are represented as the mean ± SEM of at least three independent experiments. Statistical analysis was performed using Student's t-test or one-way ANOVA as appropriate. P < 0.05 was considered significant.