Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Functions of NKG2D in CD8+ T cells: an opportunity for immunotherapy

Abstract

Natural killer group 2 member D (NKG2D) is a type II transmembrane receptor. NKG2D is present on NK cells in both mice and humans, whereas it is constitutively expressed on CD8+ T cells in humans but only expressed upon T-cell activation in mice. NKG2D is a promiscuous receptor that recognizes stress-induced surface ligands. In NK cells, NKG2D signaling is sufficient to unleash the killing response; in CD8+ T cells, this requires concurrent activation of the T-cell receptor (TCR). In this case, the function of NKG2D is to authenticate the recognition of a stressed target and enhance TCR signaling. CD28 has been established as an archetype provider of costimulation during T-cell priming. It has become apparent, however, that signals from other costimulatory receptors, such as NKG2D, are required for optimal T-cell function outside the priming phase. This review will focus on the similarities and differences between NKG2D and CD28; less well-described characteristics of NKG2D, such as the potential role of NKG2D in CD8+ T-cell memory formation, cancer immunity and autoimmunity; and the opportunities for targeting NKG2D in immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3

Similar content being viewed by others

References

  1. Houchins JP, Yabe T, McSherry C, Bach FH. DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human natural killer cells. J Exp Med 1991; 173: 1017–1020.

    CAS  PubMed  Google Scholar 

  2. Brown MG, Fulmek S, Matsumoto K, Cho R, Lyons PA, Levy ER et al. A 2-Mb YAC contig and physical map of the natural killer gene complex on mouse chromosome 6. Genomics 1997; 42: 16–25.

    CAS  PubMed  Google Scholar 

  3. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999; 285: 727–729.

    CAS  PubMed  Google Scholar 

  4. Jamieson AM, Diefenbach A, McMahon CW, Xiong N, Carlyle JR, Raulet DH. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. Immunity 2002; 17: 19–29.

    CAS  PubMed  Google Scholar 

  5. Diefenbach A, Jamieson AM, Liu SD, Shastri N, Raulet DH. Ligands for the murine NKG2D receptor: expression by tumor cells and activation of NK cells and macrophages. Nat Immunol 2000; 1: 119–126.

    CAS  PubMed  Google Scholar 

  6. Cerwenka A, Baron JL, Lanier LL. 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 2001; 98: 11521–11526.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. O'Callaghan CA, Cerwenka A, Willcox BE, Lanier LL, Bjorkman PJ. Molecular competition for NKG2D: H60 and RAE1 compete unequally for NKG2D with dominance of H60. Immunity 2001; 15: 201–211.

    CAS  PubMed  Google Scholar 

  8. Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W et al. ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 2001; 14: 123–133.

    CAS  PubMed  Google Scholar 

  9. Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH et al. Retinoic acid early inducible genes define a ligand family for the activating NKG2D receptor in mice. Immunity 2000; 12: 721–727.

    CAS  PubMed  Google Scholar 

  10. Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol 2002; 3: 1150–1155.

    CAS  PubMed  Google Scholar 

  11. Yim D, Jie HB, Sotiriadis J, Kim YS, Kim KS, Rothschild MF et al. Molecular cloning and characterization of pig immunoreceptor DAP10 and NKG2D. Immunogenetics 2001; 53: 243–249.

    CAS  PubMed  Google Scholar 

  12. Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nat Rev Immunol 2003; 3: 317–330.

    CAS  PubMed  Google Scholar 

  13. Park SG, Schulze-Luehrman J, Hayden MS, Hashimoto N, Ogawa W, Kasuga M et al. The kinase PDK1 integrates T cell antigen receptor and CD28 coreceptor signaling to induce NF-kappaB and activate T cells. Nat Immunol 2009; 10: 158–166.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ward SG. CD28: a signalling perspective. Biochem J 1996; 318 (Part 2): 361–377.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Bocko D, Kosmaczewska A, Ciszak L, Teodorowska R, Frydecka I. CD28 costimulatory molecule—expression, structure and function. Arch Immunol Ther Exp (Warsz) 2002; 50: 169–177.

    CAS  Google Scholar 

  16. Boomer JS, Green JM. An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect Biol 2010; 2: a002436.

    PubMed  PubMed Central  Google Scholar 

  17. Lee KP, Taylor C, Petryniak B, Turka LA, June CH, Thompson CB. The genomic organization of the CD28 gene. Implications for the regulation of CD28 mRNA expression and heterogeneity. J Immunol 1990; 145: 344–352.

    CAS  PubMed  Google Scholar 

  18. Carreno BM, Collins M. The B7 family of ligands and its receptors: new pathways for costimulation and inhibition of immune responses. Annu Rev Immunol 2002; 20: 29–53.

    CAS  PubMed  Google Scholar 

  19. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA 2003; 100: 4712–4717.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Walunas TL, Bakker CY, Bluestone JA. CTLA-4 ligation blocks CD28-dependent T cell activation. J Exp Med 1996; 183: 2541–2550.

    CAS  PubMed  Google Scholar 

  21. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL et al. An activating immunoreceptor complex formed by NKG2D and DAP10. Science 1999; 285: 730–732.

    CAS  PubMed  Google Scholar 

  22. Upshaw JL, Arneson LN, Schoon RA, Dick CJ, Billadeau DD, Leibson PJ. NKG2D-mediated signaling requires a DAP10-bound Grb2-Vav1 intermediate and phosphatidylinositol-3-kinase in human natural killer cells. Nat Immunol 2006; 7: 524–532.

    CAS  PubMed  Google Scholar 

  23. Garrity D, Call ME, Feng J, Wucherpfennig KW. The activating NKG2D receptor assembles in the membrane with two signaling dimers into a hexameric structure. Proc Natl Acad Sci USA 2005; 102: 7641–7646.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 2003; 3: 781–790.

    CAS  PubMed  Google Scholar 

  25. Upshaw JL, Leibson PJ. NKG2D-mediated activation of cytotoxic lymphocytes: unique signaling pathways and distinct functional outcomes. Semin Immunol 2006; 18: 167–175.

    CAS  PubMed  Google Scholar 

  26. Upshaw JL, Schoon RA, Dick CJ, Billadeau DD, Leibson PJ. The isoforms of phospholipase C-gamma are differentially used by distinct human NK activating receptors. J Immunol 2005; 175: 213–218.

    CAS  PubMed  Google Scholar 

  27. Irvin BJ, Williams BL, Nilson AE, Maynor HO, Abraham RT. Pleiotropic contributions of phospholipase C-gamma1 (PLC-gamma1) to T-cell antigen receptor-mediated signaling: reconstitution studies of a PLC-gamma1-deficient Jurkat T-cell line. Mol Cell Biol 2000; 20: 9149–9161.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Fu G, Chen Y, Schuman J, Wang D, Wen R. Phospholipase Cgamma2 plays a role in TCR signal transduction and T cell selection. J Immunol 2012; 189: 2326–2332.

    CAS  PubMed  Google Scholar 

  29. Ting AT, Karnitz LM, Schoon RA, Abraham RT, Leibson PJ. Fc gamma receptor activation induces the tyrosine phosphorylation of both phospholipase C (PLC)-gamma 1 and PLC-gamma 2 in natural killer cells. J Exp Med 1992; 176: 1751–1755.

    CAS  PubMed  Google Scholar 

  30. Billadeau DD, Upshaw JL, Schoon RA, Dick CJ, Leibson PJ. NKG2D-DAP10 triggers human NK cell-mediated killing via a Syk-independent regulatory pathway. Nat Immunol 2003; 4: 557–564.

    CAS  PubMed  Google Scholar 

  31. Tsuchida M, Manthei ER, Knechtle SJ, Hamawy MM. CD28 ligation induces rapid tyrosine phosphorylation of the linker molecule LAT in the absence of Syk and ZAP-70 tyrosine phosphorylation. Eur J Immunol 1999; 29: 2354–2359.

    CAS  PubMed  Google Scholar 

  32. Michel F, Attal-Bonnefoy G, Mangino G, Mise-Omata S, Acuto O. CD28 as a molecular amplifier extending TCR ligation and signaling capabilities. Immunity 2001; 15: 935–945.

    CAS  PubMed  Google Scholar 

  33. Sutherland CL, Chalupny NJ, Schooley K, VandenBos T, Kubin M, Cosman D. UL16-binding proteins, novel MHC class I-related proteins, bind to NKG2D and activate multiple signaling pathways in primary NK cells. J Immunol 2002; 168: 671–679.

    CAS  PubMed  Google Scholar 

  34. Markiewicz MA, Carayannopoulos LN, Naidenko OV, Matsui K, Burack WR, Wise EL et al. Costimulation through NKG2D enhances murine CD8+ CTL function: similarities and differences between NKG2D and CD28 costimulation. J Immunol 2005; 175: 2825–2833.

    CAS  PubMed  Google Scholar 

  35. Maasho K, Opoku-Anane J, Marusina AI, Coligan JE, Borrego F. NKG2D is a costimulatory receptor for human naive CD8+ T cells. J Immunol 2005; 174: 4480–4484.

    CAS  PubMed  Google Scholar 

  36. Rajasekaran K, Xiong V, Fong L, Gorski J, Malarkannan S. Functional dichotomy between NKG2D and CD28-mediated co-stimulation in human CD8+ T cells. PLoS ONE 2010; 5.

    PubMed  PubMed Central  Google Scholar 

  37. Ehrlich LI, Ogasawara K, Hamerman JA, Takaki R, Zingoni A, Allison JP et al. Engagement of NKG2D by cognate ligand or antibody alone is insufficient to mediate costimulation of human and mouse CD8+ T cells. J Immunol 2005; 174: 1922–1931.

    CAS  PubMed  Google Scholar 

  38. Barber A, Sentman CL. NKG2D receptor regulates human effector T-cell cytokine production. Blood 2011; 117: 6571–6581.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Serrano-Pertierra E, Cernuda-Morollon E, Lopez-Larrea C. NKG2D- and CD28-mediated costimulation regulate CD8+ T cell chemotaxis through different mechanisms: the role of Cdc42/N-WASp. J Leukocyte Biol 2014; 95: 487–495.

    PubMed  Google Scholar 

  40. Hu J, Batth IS, Xia X, Li S. Regulation of NKG2D(+CD8(+ T-cell-mediated antitumor immune surveillance: identification of a novel CD28 activation-mediated, STAT3 phosphorylation-dependent mechanism. Oncoimmunology 2016; 5: e1252012.

    PubMed  PubMed Central  Google Scholar 

  41. Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol 2012; 12: 749–761.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, Schoenberger SP. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003; 421: 852–856.

    CAS  PubMed  Google Scholar 

  43. Shedlock DJ, Shen H. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science 2003; 300: 337–339.

    CAS  PubMed  Google Scholar 

  44. Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 2003; 300: 339–342.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol 2003; 3: 269–279.

    CAS  PubMed  Google Scholar 

  46. Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL, Embers M et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J Exp Med 2000; 191: 771–780.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Schluns KS, Williams K, Ma A, Zheng XX, Lefrancois L. Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. J Immunol 2002; 168: 4827–4831.

    CAS  PubMed  Google Scholar 

  48. Andre MC, Sigurdardottir D, Kuttruff S, Pommerl B, Handgretinger R, Rammensee HG et al. Impaired tumor rejection by memory CD8 T cells in mice with NKG2D dysfunction. Int J Cancer 2012; 131: 1601–1610.

    CAS  PubMed  Google Scholar 

  49. McQueen B, Trace K, Whitman E, Bedsworth T, Barber A. Natural killer group 2D and CD28 receptors differentially activate mammalian/mechanistic target of rapamycin to alter murine effector CD8+ T-cell differentiation. Immunology 2016; 147: 305–320.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Soderquest K, Walzer T, Zafirova B, Klavinskis LS, Polic B, Vivier E et al. Cutting edge: CD8+ T cell priming in the absence of NK cells leads to enhanced memory responses. J Immunol 2011; 186: 3304–3308.

    CAS  PubMed  Google Scholar 

  51. Wensveen FM, Lenartic M, Jelencic V, Lemmermann NA, ten Brinke A, Jonjic S et al. NKG2D induces Mcl-1 expression and mediates survival of CD8 memory T cell precursors via phosphatidylinositol 3-kinase. J Immunol 2013; 191: 1307–1315.

    CAS  PubMed  Google Scholar 

  52. Zloza A, Kohlhapp FJ, Lyons GE, Schenkel JM, Moore TV, Lacek AT et al. NKG2D signaling on CD8(+ T cells represses T-bet and rescues CD4-unhelped CD8(+ T cell memory recall but not effector responses. Nat Med 2012; 18: 422–428.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005; 436: 1186–1190.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Eissmann P, Evans JH, Mehrabi M, Rose EL, Nedvetzki S, Davis DM. Multiple mechanisms downstream of TLR-4 stimulation allow expression of NKG2D ligands to facilitate macrophage/NK cell crosstalk. J Immunol 2010; 184: 6901–6909.

    CAS  PubMed  Google Scholar 

  55. Schwinn N, Vokhminova D, Sucker A, Textor S, Striegel S, Moll I et al. Interferon-gamma down-regulates NKG2D ligand expression and impairs the NKG2D-mediated cytolysis of MHC class I-deficient melanoma by natural killer cells. Int J Cancer 2009; 124: 1594–1604.

    CAS  PubMed  Google Scholar 

  56. Bui JD, Carayannopoulos LN, Lanier LL, Yokoyama WM, Schreiber RD. IFN-dependent down-regulation of the NKG2D ligand H60 on tumors. J Immunol 2006; 176: 905–913.

    CAS  PubMed  Google Scholar 

  57. Eisele G, Wischhusen J, Mittelbronn M, Meyermann R, Waldhauer I, Steinle A et al. TGF-beta and metalloproteinases differentially suppress NKG2D ligand surface expression on malignant glioma cells. Brain 2006; 129: 2416–2425.

    PubMed  Google Scholar 

  58. Butler JE, Moore MB, Presnell SR, Chan HW, Chalupny NJ, Lutz CT. Proteasome regulation of ULBP1 transcription. J Immunol 2009; 182: 6600–6609.

    CAS  PubMed  Google Scholar 

  59. Venkataraman GM, Suciu D, Groh V, Boss JM, Spies T. Promoter region architecture and transcriptional regulation of the genes for the MHC class I-related chain A and B ligands of NKG2D. J Immunol 2007; 178: 961–969.

    CAS  PubMed  Google Scholar 

  60. Andersson AK, Sumariwalla PF, McCann FE, Amjadi P, Chang C, McNamee K et al. Blockade of NKG2D ameliorates disease in mice with collagen-induced arthritis: a potential pathogenic role in chronic inflammatory arthritis. Arthritis Rheum 2011; 63: 2617–2629.

    CAS  PubMed  Google Scholar 

  61. Meresse B, Chen Z, Ciszewski C, Tretiakova M, Bhagat G, Krausz TN et al. Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity 2004; 21: 357–366.

    CAS  PubMed  Google Scholar 

  62. Zloza A, Lyons GE, Chlewicki LK, Kohlhapp FJ, O'Sullivan JA, Lacek AT et al. Engagement of NK receptor NKG2D, but not 2B4, results in self-reactive CD8+ T cells and autoimmune vitiligo. Autoimmunity 2011; 44: 599–606.

    CAS  PubMed  Google Scholar 

  63. Le Poole IC, Wankowicz-Kalinska A, van den Wijngaard RM, Nickoloff BJ, Das PK. Autoimmune aspects of depigmentation in vitiligo. J Invest Dermatol Symp Proc 2004; 9: 68–72.

    Google Scholar 

  64. Dai Z, Xing L, Cerise J, Wang EH, Jabbari A, de Jong A et al. CXCR3 blockade inhibits T cell migration into the skin and prevents development of alopecia areata. J Immunol 2016; 197: 1089–1099.

    CAS  PubMed  Google Scholar 

  65. Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y et al. Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature 2010; 466: 113–117.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Xing L, Dai Z, Jabbari A, Cerise JE, Higgins CA, Gong W et al. Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat Med 2014; 20: 1043–1049.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Ogasawara K, Hamerman JA, Ehrlich LR, Bour-Jordan H, Santamaria P, Bluestone JA et al. NKG2D blockade prevents autoimmune diabetes in NOD mice. Immunity 2004; 20: 757–767.

    CAS  PubMed  Google Scholar 

  68. Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR, Spies T. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol 2001; 2: 255–260.

    CAS  PubMed  Google Scholar 

  69. Tang F, Chen Z, Ciszewski C, Setty M, Solus J, Tretiakova M et al. Cytosolic PLA2 is required for CTL-mediated immunopathology of celiac disease via NKG2D and IL-15. J Exp Med 2009; 206: 707–719.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Guerra N, Tan YX, Joncker NT, Choy A, Gallardo F, Xiong N et al. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 2008; 28: 571–580.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Smyth MJ, Swann J, Cretney E, Zerafa N, Yokoyama WM, Hayakawa Y. NKG2D function protects the host from tumor initiation. J Exp Med 2005; 202: 583–588.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu G, Lu S, Wang X, Page ST, Higano CS, Plymate SR et al. Perturbation of NK cell peripheral homeostasis accelerates prostate carcinoma metastasis. J Clin Invest 2013; 123: 4410–4422.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Barber A, Zhang T, Sentman CL. Immunotherapy with chimeric NKG2D receptors leads to long-term tumor-free survival and development of host antitumor immunity in murine ovarian cancer. J Immunol 2008; 180: 72–78.

    CAS  PubMed  Google Scholar 

  74. Zhang T, Barber A, Sentman CL. Chimeric NKG2D modified T cells inhibit systemic T-cell lymphoma growth in a manner involving multiple cytokines and cytotoxic pathways. Cancer Res 2007; 67: 11029–11036.

    CAS  PubMed  Google Scholar 

  75. Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther 2011; 18: 509–516.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. VanSeggelen H, Hammill JA, Dvorkin-Gheva A, Tantalo DG, Kwiecien JM, Denisova GF et al. T cells engineered with chimeric antigen receptors targeting NKG2D ligands display lethal toxicity in mice. Mol Ther 2015; 23: 1600–1610.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Lynn RC, Powell DJ Jr.. Strain-dependent lethal toxicity in NKG2D ligand-targeted CAR T-cell therapy. Mol Ther 2015; 23: 1559–1561.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N. Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res 2005; 65: 11136–11145.

    CAS  PubMed  Google Scholar 

  79. Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M et al. Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 2005; 65: 6321–6329.

    CAS  PubMed  Google Scholar 

  80. Berghuis D, Schilham MW, Vos HI, Santos SJ, Kloess S, Buddingh EP et al. Histone deacetylase inhibitors enhance expression of NKG2D ligands in Ewing sarcoma and sensitize for natural killer cell-mediated cytolysis. Clin Sarcoma Res 2012; 2: 8.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Kennedy PT, Gehring AJ, Nowbath A, Selden C, Quaglia A, Dhillon A et al. The expression and function of NKG2D molecule on intrahepatic CD8+ T cells in chronic viral hepatitis. J Viral Hepatol 2008; 15: 901–909.

    CAS  Google Scholar 

  82. Walsh KB, Lanier LL, Lane TE. NKG2D receptor signaling enhances cytolytic activity by virus-specific CD8+ T cells: evidence for a protective role in virus-induced encephalitis. J Virol 2008; 82: 3031–3044.

    CAS  PubMed  Google Scholar 

  83. Deb C, Howe CL. NKG2D contributes to efficient clearance of picornavirus from the acutely infected murine brain. J Neurovirol 2008; 14: 261–266.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Kavazovic I, Lenartic M, Jelencic V, Jurkovic S, Lemmermann NAW, Jonjic S et al. NKG2D stimulation of CD8(+ T cells during priming promotes their capacity to produce cytokines in response to viral infection in mice. Eur J Immunol 2017; 47: 1123–1135.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jose A Guevara-Patino.

Ethics declarations

Conflict of interest

The authors declare no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Prajapati, K., Perez, C., Rojas, L.B. et al. Functions of NKG2D in CD8+ T cells: an opportunity for immunotherapy. Cell Mol Immunol 15, 470–479 (2018). https://doi.org/10.1038/cmi.2017.161

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/cmi.2017.161

This article is cited by

Search

Quick links