Advertisement
Free access
Report

A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection

Science
5 Mar 2015
Vol 348, Issue 6230
pp. 136-139

Natural born killers for tumors

Cancer immunotherapies work by activating cytotoxic lymphocytes, usually CD8+ T cells, to kill tumors. But adding new approaches to the arsenal might boost these therapies. Deng et al. now report that natural killer (NK) cells, another type of lymphocyte, can also kill tumors (see the Perspective by Steinle and Cerwenka). Mouse tumors secrete a protein called MULT1 that binds to a protein called NKG2D on the surface of NK cells. This activates NK cells and signals them to kill the tumor cells. Treating tumorbearing mice with soluble MULT1 caused their NK cells to reject the tumors.
Science, this issue p. 136; see also p. 45

Abstract

Immune cells, including natural killer (NK) cells, recognize transformed cells and eliminate them in a process termed immunosurveillance. It is thought that tumor cells evade immunosurveillance by shedding membrane ligands that bind to the NKG2D-activating receptor on NK cells and/or T cells, and desensitize these cells. In contrast, we show that in mice, a shed form of MULT1, a high-affinity NKG2D ligand, causes NK cell activation and tumor rejection. Recombinant soluble MULT1 stimulated tumor rejection in mice. Soluble MULT1 functions, at least in part, by competitively reversing a global desensitization of NK cells imposed by engagement of membrane NKG2D ligands on tumor-associated cells, such as myeloid cells. The results overturn conventional wisdom that soluble ligands are always inhibitory and suggest a new approach for cancer immunotherapy.
Natural killer (NK) cells and some T cells use activating receptors such as NKG2D to recognize and eliminate infected and transformed cells that up-regulate ligands for these receptors (1). There are six to eight different NKG2D ligands, which are poorly expressed by normal cells but up-regulated in cancer cells (2). Many tumor cells release soluble NKG2D ligands through proteolytic shedding, alternative splicing, or exosome secretion (2, 3). Numerous reports conclude that excreted NKG2D ligands modulate NKG2D from the cell surface and desensitize antitumor effector cells (4, 5), although a functional impact of soluble NKG2D ligands is not always observed (69). To study shed NKG2D ligands in a controlled setting, we focused on the mouse ligand MULT1, which is commonly up-regulated in primary tumors (10) and is a transmembrane protein, as are the human ligands major histocompatibility complex (MHC) class I chain–related protein A (MICA), MICB, ULBP4, and ULBP5 (11). Analysis of fibroblasts transduced with either N- or C-terminally tagged MULT1 revealed an N-terminal species (23 kD after deglycosylation) shed into the culture supernatant (fig. S1A) and a 24 kD membrane “stub” in the cell lysates, in addition to full-length (around 42 kD) MULT1 (fig. S1B). Inhibiting matrix metalloproteinases blocked MULT1 shedding (fig. S1C).
Hemagglutinin (HA)–MULT1–transduced fibroblasts produced nearly eightfold more shed MULT1 than did untransduced fibroblasts (fig. S2). WEHI-7.1 and C1498 but not human 293T cell lines excreted MULT1 produced endogenously. We detected serum MULT1 (mean concentration ~250 ng/ml) in most tumor-bearing Eμ-myc transgenic mice, which frequently develop MULT1+ tumors (10), but not in most nontransgenic littermates (Fig. 1A). Very high concentrations of soluble MULT1 were also detected in sera of Apoe−/− mice fed a high-fat diet (Fig. 1A). Given that atherosclerosis and liver inflammation in such mice are largely dependent on NKG2D function (12), it seemed unlikely that soluble MULT1 inhibits NKG2D function. Thus, MULT1 is released from cell lines that naturally or ectopically express MULT1 and accumulates in sera of animals with spontaneous tumors and NKG2D-dependent inflammatory disease.
Fig. 1 NK cells promote the rejection of tumors that shed MULT1.
(A) Enzyme-linked immunosorbent assay detection of soluble MULT1 in sera from tumor-bearing Eμ-Myc mice, nontransgenic littermates, and diseased Apoe−/− mice fed a Western diet (n = 6 to 8 mice). Each point represents a different mouse. (B) Comparison of growth of 2 × 104 subcutaneously transferred B16 melanoma tumor cells transduced with secMULT1, full-length MULT1, or empty vector, in wild-type (WT) B6 mice (n = 4 mice). Rejection was usually partial but was complete in some animals in some experiments. (C) Subcutaneous growth of B16-secMULT1 tumors in B6 mice (2 × 104 cells were inoculated) treated with control imuunoglobulin G (IgG), NK1.1 antibody, or CD8 antibody (n = 13 mice). (D) After inoculation of 2 × 104 B16 cells transduced with pFG12-secMULT1, mice were treated or not with doxycycline starting from the time of tumor implantation (n = 6 mice). (E) Mice (n = 6) received 2 × 104 B16 cells alone, or 2 × 104 B16 cells mixed with 2 × 103 B16-secMULT1 cells. Shown are representative examples of ≥3 [(B) and (E)] or 2 (D) experiments performed, whereas (C) includes combined data from 3 experiments. Tumor volumes ± SE are shown. (A) was analyzed with a Mann-Whitney test, and (B) to (E) were analyzed by two-way analysis of variance (ANOVA) with Bonferroni multiple comparison tests. *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
Purified shed HA-MULT1 bound to NKG2D with high affinity [average dissociation constant (Kd) of 13 ± 3.8 nM] (fig. S3), similar to the affinity reported for recombinant MULT1 (13). In parallel, we engineered fibroblasts to secrete an ectodomain fragment of HA-MULT1 (which we call “secMULT”). SecMULT1 also bound to NKG2D with high affinity (19 ± 4.3 nM) (fig. S3).
To test the function of soluble MULT1, we engineered two NKG2D ligand-negative B6 strain tumor cell lines (the B16-BL6 melanoma and RMA lymphoma) to secrete secMULT1. Surprisingly, both cell lines were rejected by syngeneic B6 mice compared with cells transduced with empty vector (Fig. 1B and fig. S4A), despite the absence of cell surface MULT1 (fig. S4B). Tumor cells transduced with full-length MULT1 [mutated in the cytoplasmic tail to optimize cell surface expression (fig. S4B) (14)] were also rejected (Fig. 1B). B16-secMULT1 cells were still rejected in B6 hosts that had been depleted of CD8+ cells but grew progressively in B6 and Rag1−/− hosts that had been depleted of NK1.1+ cells (Fig. 1C and fig. S5). Hence, NK cells but not CD8+ cells participate in the rejection of B16-secMULT1. B16 cells with inducible secMULT1 (fig. S6) were also partially rejected (Fig. 1D). In this case, the secMULT1 lacked an epitope tag, showing that rejection occurs without a tag. A mixture of B16 (90%) and B16-secMULT1 (10%) cells was also rejected, demonstrating that sec-MULT1 acts extrinsically (Fig. 1E). These data ruled out the possibility that rejection was solely due to intrinsic stress responses in sec-MULT1–expressing tumor cells. Instead, the data suggested that secMULT1 mobilizes or activates antitumor effector cells.
To address whether tumor cells secreting secMULT1 activate NK cells, we adapted a short-term in vivo NK induction protocol (15, 16) by injecting irradiated tumor cells intraperitoneally in normal mice. Injection of B16-secMULT1 or B16 cells induced similar modest increases in the percentages of NK cells in the peritoneal washes 3 days later (fig. S7), but B16-secMULT1 cells induced more potent ex vivo killing activity against NK-sensitive YAC-1 tumor cells (Fig. 2A). Similar results were obtained with RMA-secMULT1 cells (fig. S8A). Furthermore, higher percentages of peritoneal NK cells from mice injected with B16-secMULT1 cells produced interferon-γ (IFN-γ) after stimulation ex vivo with YAC-1 tumor cells (Fig. 2B) or immobilized antibodies against NK-activating receptors (Fig. 2C and fig. S8B). To allow the recovery of intratumoral NK cells at early times after subcutaneous transfer, we implanted 3 × 105 to 5 × 105 tumor cells mixed with matrigel. Seven days later, NK cells extracted from B16-secMULT1 tumors exhibited stronger IFN-γ responses after stimulation ex vivo (Fig. 2D). Therefore, soluble MULT1 stimulated NK cell functional capacities in both subcutaneous and peritoneal tumors.
Fig. 2 Soluble MULT1 amplifies NK cell responses and causes tumor rejection.
(A to C) B6 mice were injected intraperitoneally with 5 × 106 irradiated B16 or B16-secMULT1 cells, or phosphate-buffered saline (PBS). Peritoneal wash cells (pooled from five mice) were recovered 3 days later and tested for (A) killing of YAC-1 target cells, (B) intracellular IFN-γ after stimulation with YAC-1 cells, or (C) immobilized NKp46 antibody; control responses to PBS are depicted by white segments of the bars. (D) B6 mice were injected subcutaneously with 3 × 105 to 5 × 105 B16 or B16-secMULT1 cells in 100 μl matrigel. The tumors were dissociated 7 days later, and gated NK cells from individual mice (n = 5 mice) were tested for responses to immobilized NKp46 or NKRP1C Abs. (E and F) Subcutaneous tumors were established with 3 × 105 to 5 × 105 B16 cells in matrigel. The tumor cells in one group were mixed with 1 μg of recombinant MULT1 (rMULT1). After 4 days, an additional 1 μg of rMULT1 (or PBS for control mice) was injected into each matrigel/tumor for that group. On day 7, tumors were extracted, (E) weighed, dissociated, and (E) the tumor cells were counted. The immune cells within the tumors were stimulated with immobilized NKp46 and NKRP1C Abs, and (F) the IFN-γ responses of gated NK cells were determined. Shown are representative examples of 2 (A) or ≥3 [(B) to (F)] experiments performed. (A) to (D) and (F) were analyzed with two-way ANOVA with Bonferroni multiple comparison tests, (E) was analyzed with Mann-Whitney test. *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
Recombinant MULT1 (rMULT1) is similar in size to shed MULT1. When injected with B16 tumor cells (fig. S9A), rMULT1 resulted in partial tumor rejection (Fig. 2E and fig. S9B), and NK cells extracted from the tumors exhibited increased functional activity after stimulation ex vivo (Fig. 2F). The rMULT1 sample was devoid of endotoxin or other PAMPs that activate macrophages (fig. S9C). These data established that soluble MULT1 causes tumor rejection, likely by activating NK cells.
secMULT1 and shed MULT1 are monomeric (fig. S10, A to C) and should not cross-link NKG2D, which is typically necessary for immune receptor activation. Indeed, monomeric rMULT1 failed to stimulate IFN-γ production when incubated with peritoneal NK cells for 4 hours (fig. S11A). Soluble MULT1 may form a multivalent array in vivo, but preliminary staining analyses failed to detect such arrays. These data argue that soluble MULT1 stimulates NK cells by other mechanisms.
Target cells bearing membrane NKG2D ligands, including MULT1, caused down-regulation of cell surface NKG2D, presumably by aggregating the receptor and triggering receptor endocytosis (Fig. 3A) (8, 17). Tumor cells secreting secMULT1, in contrast, caused NKG2D up-regulation on NK cells in vivo (Fig. 3, A and B), without affecting an irrelevant receptor, DNAM-1 (Fig. 3B). NKG2D up-regulation occurred without increases in NKG2D mRNA (fig. S12) or intracellular protein. These findings suggested the following hypothesis (Fig. 3C): that untransformed host cells express membrane NKG2D ligands that persistently engage NK cells, cause NKG2D down-regulation, and globally desensitize the NK cells as the tumors progress; and that soluble MULT1 enhances responsiveness and cell-surface NKG2D expression by blocking these interactions.
Fig. 3 Mechanisms of immune activation by soluble MULT1.
(A and B) Membrane NKG2D staining after exposure of NK cells to secMULT1 in intraperitoneal (A) or subcutaneous (B) tumors. (C) Model of secMULT1 action. Persistent NKG2D engagement by endogenous NKG2D ligand–expressing cells associated with the tumor desensitizes NK cells. Soluble MULT1 competitively blocks the NKG2D receptor, preventing NK cell desensitization and therefore augmenting tumor rejection mediated through distinct NK-activating receptors. (D) Expression of NKG2D ligand RAE-1 by gated CD11b+F480+ peritoneal myeloid cells in mice injected intraperitoneally 3 days before with PBS or 5 × 106 irradiated B16 or B16-secMULT1 tumor cells. Cells were stained with biotin-pan-RAE-1 Ab (blue). The staining was specific because it could be blocked by including an excess of unconjungated pan-RAE-1 antibody in the reaction (red). Gray shows isotype control staining. (E) Expression of RAE-1 by gated CD11b+F480+ intratumoral myeloid cells in mice injected subcutaneously with 2 × 104 B16 or B16-secMULT1 tumor cells 20 days before. (F) rMULT1 and NKG2D antibody [MI6 clone, in F(ab’)2 form] block RAE-1 binding to NKG2D on NK cells. The mean fluorescence intensity of RAE1ε-Fc staining of NK cells was used to calculate percent inhibition. (A) is combined data from 14 experiments. (B) and (D) to (F) show representative examples of ≥3 experiments performed. (A) was analyzed with one-way ANOVA Kruskal-Wallis test, (B) was analyzed with two-way ANOVA with Bonferroni multiple comparison tests; ns indicates P > 0.05, *P < 0.05, and ***P < 0.001.
Consistent with the hypothesis, host CD11b+F4/80+ myeloid cells associated with either peritoneal or subcutaneous B16 or B16-secMULT1 tumors displayed the NKG2D ligand RAE-1 (but not MULT1) on the cell surface (Fig. 3, D and E). Monocytes in patients with several types of cancer also expressed NKG2D ligands (18). Tumors may cause an increase in RAE-1 expression by myeloid cells in the peritoneum (Fig. 3D) and possibly other sites (19). Hence, myeloid cells, and possibly other nontumor cells, express NKG2D ligands in vivo. We further demonstrated that rMULT1 competitively blocks binding of RAE-1ε-Fc fusion protein to NKG2D on NK cells (Fig. 3F), confirming a distinct prediction of the hypothesis.
To test whether RAE-1 expressed on endogenous cells caused NK cell inactivation, we used the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 method to disrupt both B6 strain RAE-1 genes, Raet1d and Raet1e. Peritoneal NK cells from Raet1d−/−Raet1e−/− mice exhibited significant increases in membrane NKG2D expression as well as functional responses (Fig. 4, A to C). Greater differences occurred when the mice were injected with irradiated B16 tumor cells, suggesting that greater NK desensitization and NKG2D down-regulation occurs in the presence of tumors. The increase did not fully account for the effects of secMULT1 (Fig. 4, A and B), suggesting that secMULT1 may also operate by other mechanisms. NK cells in Raet1-deficient mice exhibited greater responses to both NKG2D-dependent stimuli (for example, to YAC-1 cells) (Fig. 4B) and NKG2D-independent stimuli (for example, to antibody to NKp46) (Fig. 4C), which is consistent with published reports that persistent stimulation by cells expressing NKG2D ligands results in a global desensitization of NK cells (17, 20). In mice injected with B16-secMULT1 tumor cells, smaller differences were observed, as predicted if secMULT1 blocks NKG2D interactions with RAE-1-expressing cells. Indeed, addition of rMULT1 to cultures of peritoneal wash cells, which contain NK cells and RAE-1+ myeloid cells, resulted in increased responses of NK cells to stimulation, when tested 8 to 20 hours later (fig. S11, B and C). These data indicated that interactions of NK cells with RAE-1 molecules on nontumor cells cause NKG2D down-regulation and functional desensitization, and that this may be accentuated in tumor-bearing mice.
Fig. 4 Augmented NK cell responses in RAE-1–deficient and NKG2D-deficient mice.
(A to C) Peritoneal NK cells from Raet1d−/−Raet1e−/− mice (KO) exhibited (A) increased amounts of cell-surface NKG2D and (B) increased functional responses ex vivo to YAC-1 tumor cells or (C) NKp46 antibody stimulation; control responses to PBS are depicted by white segments of the bars. The effects were larger when the mice were injected 3 days earlier with irradiated B16 tumor cells but smaller when they were injected with B16-secMULT1 tumor cells. (D) NKG2D-deficient (Klrk1−/−) NK cells exhibited increased functional activity. Splenic NK cells from Rag−/− and Rag−/− Klrk1−/− mice were stimulated ex vivo with immobilized NKp46 or NKRP1C Abs, and the IFN-γ responses of gated NK cells were determined. (E) B6 mice were injected intraperitoneally with 50 μg MI6 (anti-NKG2D) F(ab')2 or F(ab')2 of rat IgG on days 0, 3, and 6. On Day 8, peritoneal NK cells were stimulated ex vivo with immobilized NKp46 Abs, and the IFN-γ responses of gated NK cells were determined. (F and G) Increased tumor rejection responses in NKG2D-deficient mice. Growth of (F) B16-secMULT1 or (G) B16 tumor cells in Rag2−/− or Rag2−/−Klrk1−/− mice (n = 5 mice). (F) and (G) are from separate experiments. Separate, direct comparisons showed retarded growth of B16-secMULT1 versus B16 tumors in Rag2−/− mice. All experiments show representative examples of ≥3 experiments performed. Tumor volumes ± SE are shown. (A) to (G) were analyzed with two-way ANOVA with Bonferroni multiple comparison tests. *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001.
Our model further predicts that NKG2D receptor deficiency, or blocking with antibody, should have a similar effect as soluble MULT1. NK cells from NKG2D-deficient (Klrk1−/−) mice exhibited a modest increase in functional activity (fig. S13A), confirming recent findings (21, 22). A larger effect was evident in NKG2D-deficient NK cells on a Rag2−/− background (Fig. 4D). Similarly, intraperitoneal injections of B6 mice with F(ab’)2 fragments of blocking NKG2D antibody resulted in enhanced functional responses ex vivo (Fig. 4E). Most remarkably, NKG2D-deficient Rag2−/− mice exhibited a strongly enhanced rejection response against B16 and B16-secMULT1 tumors, compared with the responses of Rag2−/− mice (Fig. 4, F and G). Furthermore, incorporation of F(ab’)2 fragments of blocking NKG2D antibody into B16 tumors that were established in subcutaneous matrigel plugs resulted in partial tumor rejection and augmented responsiveness of NK cells within the residual tumors (fig. S13, B and C). Thus, NKG2D deficiency, or blockade, results in enhanced NK cell responsiveness and tumor rejection. These data strongly support the proposed model (Fig. 3C).
The finding that NK cells persistently stimulated through NKG2D or other receptors are broadly desensitized is consistent with published data (20, 23) and may reflect a defect in mitogen-activated protein kinase (MAPK)/extracellular signal–regulated kinase (ERK) signaling (23). Blocking or disabling NKG2D restores killing of B16 cells because NK cells use receptors distinct from NKG2D to target B16 cells. A more complex outcome should pertain with tumor cells that express membrane NKG2D ligands because the NK cells, although more active, will be partly blocked in tumor cell recognition. Tumor cells often express multiple NKG2D ligands, suggesting that therapeutic efficacy may be maximized by blocking only the specific NKG2D ligands expressed by host cells, rather than by blocking NKG2D altogether.
Our results are surprising because they show that soluble NKG2D ligands in vivo stimulate tumor rejection and increase membrane NKG2D, whereas the literature suggests that they should suppress tumor rejection and decrease membrane NKG2D. However, NKG2D down-regulation is frequently not observed in patients with soluble MICA/MICB (69). Moreover, MULT1 and MICA/MICB ligands differ in a key respect: affinity. Soluble MULT1 is a high-affinity (Kd ~ 10 nM) monomeric ligand. Soluble MICA and MICB are low-affinity ligands (Kd ~ 1 μM), which are present in patient sera at concentrations below 1 nM (4, 24), meaning that NKG2D occupancy is predicted to be extremely low. This consideration suggests that systemic effects of soluble MICA and MICB may be indirect, or that the active form of soluble MICA or MICB is actually a multimeric exosome form (25, 26), which can bind and cross-link the receptors despite a low affinity and low concentration. Binding and cross-linking are conditions known to cause modulation of other immune receptors from the cell surface (27). B16 cells secreting the low-affinity MICA ligand, when injected intraperitoneally, failed to induce significant NKG2D up-regulation or increased NK functional activity (fig. S14), which is in line with our expectations. The results identify an unexpected mechanism of immune activation and support efforts to evaluate the potential of soluble NKG2D ligands or antibodies that block NKG2D or its ligands for immunotherapy of cancer. Studies suggest that engagement of other NK-activating receptors, such as NKp46, may also lead to NK cell desensitization, suggesting that multiple targets exist for amplifying NK function (28).

Acknowledgments

We thank H. Nolla and A. Valeros for help with cell sorting; C. Kang for targeting eggs for creating the Raet1 knockout mice; Q. Yan for assistance with Biacore assays; and T. Trevino, L. Bai, S. Li for technical assistance. We thank Raulet Lab members and R. Vance for useful discussion and comments on the manuscript. The data presented in this manuscript are tabulated in the main paper and in the supplementary materials. D.H.R. is an inventor on a patent (U.S. 6821522) filed by University of California, Berkeley describing the use of soluble ligands for immunotherapy of cancer. W.D. was supported by a Cancer Research Institute postdoctoral fellowship. B.G.G. was supported by a National Science Foundation Graduate Research Fellowship and the Hirth Chair Graduate Fellowship of University of California, Berkeley. L.W. and A.I. were supported by Leukemia and Lymphoma Society Fellowships, and S.L. was supported by a NIH–Initiative for Maximizing Student Development grant. This work was supported by NIH grant R01 CA093678 to D.H.R.

Supplementary Material

Summary

Materials and Methods
Supplementary Text
Fig. S1 to S14
Table S1
References (2932)

Resources

File (deng-sm.pdf)

References and Notes

1
Raulet D. H., Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 3, 781–790 (2003).
2
Raulet D. H., Gasser S., Gowen B. G., Deng W., Jung H., Regulation of ligands for the NKG2D activating receptor. Annu. Rev. Immunol. 31, 413–441 (2013).
3
Chitadze G., Bhat J., Lettau M., Janssen O., Kabelitz D., Generation of soluble NKG2D ligands: Proteolytic cleavage, exosome secretion and functional implications. Scand. J. Immunol. 78, 120–129 (2013).
4
Groh V., Wu J., Yee C., Spies T., Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419, 734–738 (2002).
5
Song H., Kim J., Cosman D., Choi I., Soluble ULBP suppresses natural killer cell activity via down-regulating NKG2D expression. Cell. Immunol. 239, 22–30 (2006).
6
Salih H. R., Goehlsdorf D., Steinle A., Release of MICB molecules by tumor cells: Mechanism and soluble MICB in sera of cancer patients. Hum. Immunol. 67, 188–195 (2006).
7
Waldhauer I., Steinle A., Proteolytic release of soluble UL16-binding protein 2 from tumor cells. Cancer Res. 66, 2520–2526 (2006).
8
Wiemann K., Mittrücker H. W., Feger U., Welte S. A., Yokoyama W. M., Spies T., Rammensee H. G., Steinle A., Systemic NKG2D down-regulation impairs NK and CD8 T cell responses in vivo. J. Immunol. 175, 720–729 (2005).
9
von Lilienfeld-Toal M., Frank S., Leyendecker C., Feyler S., Jarmin S., Morgan R., Glasmacher A., Märten A., Schmidt-Wolf I. G., Brossart P., Cook G., Reduced immune effector cell NKG2D expression and increased levels of soluble NKG2D ligands in multiple myeloma may not be causally linked. Cancer Immunol. Immunother. 59, 829–839 (2010).
10
Guerra N., Tan Y. X., Joncker N. T., Choy A., Gallardo F., Xiong N., Knoblaugh S., Cado D., Greenberg N. M., Raulet D. H., NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 28, 571–580 (2008).
11
Eagle R. A., Flack G., Warford A., Martínez-Borra J., Jafferji I., Traherne J. A., Ohashi M., Boyle L. H., Barrow A. D., Caillat-Zucman S., Young N. T., Trowsdale J., Cellular expression, trafficking, and function of two isoforms of human ULBP5/RAET1G. PLOS ONE 4, e4503 (2009).
12
Xia M., Guerra N., Sukhova G. K., Yang K., Miller C. K., Shi G. P., Raulet D. H., Xiong N., Immune activation resulting from NKG2D/ligand interaction promotes atherosclerosis. Circulation 124, 2933–2943 (2011).
13
Carayannopoulos L. N., Naidenko O. V., Fremont D. H., Yokoyama W. M., Cutting edge: murine UL16-binding protein-like transcript 1: A newly described transcript encoding a high-affinity ligand for murine NKG2D. J. Immunol. 169, 4079–4083 (2002).
14
Nice T. J., Coscoy L., Raulet D. H., Posttranslational regulation of the NKG2D ligand Mult1 in response to cell stress. J. Exp. Med. 206, 287–298 (2009).
15
Glas R., Franksson L., Une C., Eloranta M. L., Ohlén C., Orn A., Kärre K., Recruitment and activation of natural killer (NK) cells in vivo determined by the target cell phenotype. An adaptive component of NK cell-mediated responses. J. Exp. Med. 191, 129–138 (2000).
16
Diefenbach A., Jensen E. R., Jamieson A. M., Raulet D. H., Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 413, 165–171 (2001).
17
Oppenheim D. E., Roberts S. J., Clarke S. L., Filler R., Lewis J. M., Tigelaar R. E., Girardi M., Hayday A. C., Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat. Immunol. 6, 928–937 (2005).
18
Crane C. A., Austgen K., Haberthur K., Hofmann C., Moyes K. W., Avanesyan L., Fong L., Campbell M. J., Cooper S., Oakes S. A., Parsa A. T., Lanier L. L., Immune evasion mediated by tumor-derived lactate dehydrogenase induction of NKG2D ligands on myeloid cells in glioblastoma patients. Proc. Natl. Acad. Sci. U.S.A. 111, 12823–12828 (2014).
19
Nausch N., Galani I. E., Schlecker E., Cerwenka A., Mononuclear myeloid-derived “suppressor” cells express RAE-1 and activate natural killer cells. Blood 112, 4080–4089 (2008).
20
Coudert J. D., Scarpellino L., Gros F., Vivier E., Held W., Sustained NKG2D engagement induces cross-tolerance of multiple distinct NK cell activation pathways. Blood 111, 3571–3578 (2008).
21
Zafirova B., Mandarić S., Antulov R., Krmpotić A., Jonsson H., Yokoyama W. M., Jonjić S., Polić B., Altered NK cell development and enhanced NK cell-mediated resistance to mouse cytomegalovirus in NKG2D-deficient mice. Immunity 31, 270–282 (2009).
22
Sheppard S., Triulzi C., Ardolino M., Serna D., Zhang L., Raulet D. H., Guerra N., Characterization of a novel NKG2D and NKp46 double-mutant mouse reveals subtle variations in the NK cell repertoire. Blood 121, 5025–5033 (2013).
23
Ardolino M., Azimi C. S., Iannello A., Trevino T. N., Horan L., Zhang L., Deng W., Ring A. M., Fischer S., Garcia K. C., Raulet D. H., Cytokine therapy reverses NK cell anergy in MHC-deficient tumors. J. Clin. Invest. 124, 4781–4794 (2014).
24
Märten A., von Lilienfeld-Toal M., Büchler M. W., Schmidt J., Soluble MIC is elevated in the serum of patients with pancreatic carcinoma diminishing gammadelta T cell cytotoxicity. Int. J. Cancer 119, 2359–2365 (2006).
25
Clayton A., Mitchell J. P., Court J., Linnane S., Mason M. D., Tabi Z., Human tumor-derived exosomes down-modulate NKG2D expression. J. Immunol. 180, 7249–7258 (2008).
26
Ashiru O., Boutet P., Fernández-Messina L., Agüera-González S., Skepper J. N., Valés-Gómez M., Reyburn H. T., Natural killer cell cytotoxicity is suppressed by exposure to the human NKG2D ligand MICA*008 that is shed by tumor cells in exosomes. Cancer Res. 70, 481–489 (2010).
27
Taylor R. B., Duffus W. P., Raff M. C., de Petris S., Redistribution and pinocytosis of lymphocyte surface immunoglobulin molecules induced by anti-immunoglobulin antibody. Nat. New Biol. 233, 225–229 (1971).
28
Narni-Mancinelli E., Jaeger B. N., Bernat C., Fenis A., Kung S., De Gassart A., Mahmood S., Gut M., Heath S. C., Estellé J., Bertosio E., Vely F., Gastinel L. N., Beutler B., Malissen B., Malissen M., Gut I. G., Vivier E., Ugolini S., Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science 335, 344–348 (2012).
29
Iannello A., Thompson T. W., Ardolino M., Lowe S. W., Raulet D. H., p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells. J. Exp. Med. 210, 2057–2069 (2013).
30
Wang H., Yang H., Shivalila C. S., Dawlaty M. M., Cheng A. W., Zhang F., Jaenisch R., One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).
31
Nice T. J., Deng W., Coscoy L., Raulet D. H., Stress-regulated targeting of the NKG2D ligand Mult1 by a membrane-associated RING-CH family E3 ligase. J. Immunol. 185, 5369–5376 (2010).
32
Jung H., Hsiung B., Pestal K., Procyk E., Raulet D. H., RAE-1 ligands for the NKG2D receptor are regulated by E2F transcription factors, which control cell cycle entry. J. Exp. Med. 209, 2409–2422 (2012).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 348 | Issue 6230
3 April 2015

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 17 July 2014
Accepted: 30 January 2015
Published in print: 3 April 2015

Permissions

Request permissions for this article.

Acknowledgments

We thank H. Nolla and A. Valeros for help with cell sorting; C. Kang for targeting eggs for creating the Raet1 knockout mice; Q. Yan for assistance with Biacore assays; and T. Trevino, L. Bai, S. Li for technical assistance. We thank Raulet Lab members and R. Vance for useful discussion and comments on the manuscript. The data presented in this manuscript are tabulated in the main paper and in the supplementary materials. D.H.R. is an inventor on a patent (U.S. 6821522) filed by University of California, Berkeley describing the use of soluble ligands for immunotherapy of cancer. W.D. was supported by a Cancer Research Institute postdoctoral fellowship. B.G.G. was supported by a National Science Foundation Graduate Research Fellowship and the Hirth Chair Graduate Fellowship of University of California, Berkeley. L.W. and A.I. were supported by Leukemia and Lymphoma Society Fellowships, and S.L. was supported by a NIH–Initiative for Maximizing Student Development grant. This work was supported by NIH grant R01 CA093678 to D.H.R.

Authors

Affiliations

Weiwen Deng
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Benjamin G. Gowen
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Li Zhang
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Lin Wang
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Stephanie Lau
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Alexandre Iannello
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Jianfeng Xu
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.
Tihana L. Rovis
Center for Proteomics University of Rijeka Faculty of Medicine Brace Branchetta 20, 51000 Rijeka, Croatia.
Na Xiong
Department of Veterinary and Biomedical Sciences, Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.
David H. Raulet* [email protected]
Department of Molecular and Cell Biology, and Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA.

Notes

*
Corresponding author. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. DNA Repair Deficiency Regulates Immunity Response in Cancers: Molecular Mechanism and Approaches for Combining Immunotherapy, Cancers, 15, 5, (1619), (2023).https://doi.org/10.3390/cancers15051619
    Crossref
  2. Cytokinopathy with aberrant cytotoxic lymphocytes and profibrotic myeloid response in SARS-CoV-2 mRNA vaccine–associated myocarditis, Science Immunology, 8, 83, (2023)./doi/10.1126/sciimmunol.adh3455
    Abstract
  3. Nanomaterials: A powerful tool for tumor immunotherapy, Frontiers in Immunology, 13, (2022).https://doi.org/10.3389/fimmu.2022.979469
    Crossref
  4. Underlying mechanisms of evasion from NK cells as rationale for improvement of NK cell-based immunotherapies, Frontiers in Immunology, 13, (2022).https://doi.org/10.3389/fimmu.2022.910595
    Crossref
  5. Recruited and Tissue-Resident Natural Killer Cells in the Lung During Infection and Cancer, Frontiers in Immunology, 13, (2022).https://doi.org/10.3389/fimmu.2022.887503
    Crossref
  6. Natural Killer Cells in the Malignant Niche of Multiple Myeloma, Frontiers in Immunology, 12, (2022).https://doi.org/10.3389/fimmu.2021.816499
    Crossref
  7. NKG2D signaling regulates IL-17A-producing γδT cells in mice to promote cancer progression, Discovery Immunology, 1, 1, (2022).https://doi.org/10.1093/discim/kyac002
    Crossref
  8. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy, Nature Reviews Immunology, 23, 2, (90-105), (2022).https://doi.org/10.1038/s41577-022-00732-1
    Crossref
  9. p53 missense mutant G242A subverts natural killer cells in sheltering mouse breast cancer cells against immune rejection, Experimental Cell Research, 417, 1, (113210), (2022).https://doi.org/10.1016/j.yexcr.2022.113210
    Crossref
  10. Natural killer cell dysfunction in cancer and new strategies to utilize NK cell potential for cancer immunotherapy, Molecular Immunology, 144, (58-70), (2022).https://doi.org/10.1016/j.molimm.2022.02.015
    Crossref
  11. See more
Loading...

View Options

View options

PDF format

Download this article as a PDF file

Download PDF

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media