Review Special Issues

Ubiquitin and ubiquitin-like modifiers modulate NK cell-mediated recognition and killing of damaged cells

  • Received: 28 September 2017 Accepted: 21 November 2017 Published: 23 November 2017
  • Efficient elimination of transformed and virus-infected cells by natural killer (NK) cells mainly depends on the recognition of “induced self” ligands by activating receptors, including NKG2D and DNAM1. The surface expression of these ligands in stressed or diseased cells results from the integration of transcriptional, post-transcriptional and post-translational mechanisms. Among post-translational mechanisms, recent findings indicate that ubiquitin and ubiquitin-like modifications, namely ubiquitination and SUMOylation, contribute to a very rapid negative regulation of NKG2D and DNAM1 ligand surface expression promoting either ligand degradation or ligand intracellular retention. On the other hand, accumulating evidences demonstrate that NKG2D receptor expression is down-regulated by ubiquitin-dependent endocytosis upon ligand stimulation. In this scenario, the overall consequence of the post-translational modifications of activating NK cell receptors and of their ligands on target cells is to impair effector cell-mediated recognition of damaged cells. Our review summarizes recent findings on the role of post-translational modifications in the modulation of target cell susceptibility to NK cell-mediated killing.

    Citation: Rosa Molfetta, Beatrice Zitti, Angela Santoni, Rossella Paolini. Ubiquitin and ubiquitin-like modifiers modulate NK cell-mediated recognition and killing of damaged cells[J]. AIMS Allergy and Immunology, 2017, 1(4): 164-180. doi: 10.3934/Allergy.2017.4.164

    Related Papers:

  • Efficient elimination of transformed and virus-infected cells by natural killer (NK) cells mainly depends on the recognition of “induced self” ligands by activating receptors, including NKG2D and DNAM1. The surface expression of these ligands in stressed or diseased cells results from the integration of transcriptional, post-transcriptional and post-translational mechanisms. Among post-translational mechanisms, recent findings indicate that ubiquitin and ubiquitin-like modifications, namely ubiquitination and SUMOylation, contribute to a very rapid negative regulation of NKG2D and DNAM1 ligand surface expression promoting either ligand degradation or ligand intracellular retention. On the other hand, accumulating evidences demonstrate that NKG2D receptor expression is down-regulated by ubiquitin-dependent endocytosis upon ligand stimulation. In this scenario, the overall consequence of the post-translational modifications of activating NK cell receptors and of their ligands on target cells is to impair effector cell-mediated recognition of damaged cells. Our review summarizes recent findings on the role of post-translational modifications in the modulation of target cell susceptibility to NK cell-mediated killing.


    加载中
    [1] Caligiuri MA (2008) Human natural killer cells. Blood 112: 461–469. doi: 10.1182/blood-2007-09-077438
    [2] Morvan MG, Lanier LL (2016) NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 16: 7–19.
    [3] Long EO, Kim HS, Liu D, et al. (2013) Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol 31: 227–258. doi: 10.1146/annurev-immunol-020711-075005
    [4] Raulet DH, Gasser S, Gowen BG, et al. (2013) Regulation of ligands for the NKG2D activating receptor. Annu Rev Immunol 31: 413–441. doi: 10.1146/annurev-immunol-032712-095951
    [5] Cerboni C, Fionda C, Soriani A, et al. (2014) The DNA damage response: a common pathway in the regulation of NKG2D and DNAM-1 ligand expression in normal, infected, and cancer cells. Front Immunol 4: 508.
    [6] Chan CJ, Smyth MJ, Martinet L (2014) Molecular mechanisms of natural killer cell activation in response to cellular stress. Cell Death Differ 21: 5–14. doi: 10.1038/cdd.2013.26
    [7] Ullrich E, Koch J, Cerwenka A, et al. (2013) New prospects on the NKG2D/NKG2DL system for oncology. Oncoimmunology 2: e26097. doi: 10.4161/onci.26097
    [8] Marcus A, Gowen BG, Thompson TW, et al. (2014) Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol 122: 91–128. doi: 10.1016/B978-0-12-800267-4.00003-1
    [9] Lanier LL (2015) NKG2D receptor and its ligands in host defense. Cancer Immunol Res 3: 575–582. doi: 10.1158/2326-6066.CIR-15-0098
    [10] Guerra N, Joncker NT, Choy A, et al. (2008) NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 28: 571–580. doi: 10.1016/j.immuni.2008.02.016
    [11] Zafirova B, Mandarić S, Antulov R, et al. (2009) Altered NK cell development and enhanced NK cell-mediated resistance to mouse cytomegalovirus in NKG2D-deficient mice. Immunity 31: 270–282. doi: 10.1016/j.immuni.2009.06.017
    [12] Bauer S, Groh V, Wu J, et al. (1999) Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285: 727–729. doi: 10.1126/science.285.5428.727
    [13] Wu J, Song Y, Bakker AB, et al. (1999) An activating immunoreceptor complex formed by NKG2D and DAP10. Science 285: 730–732. doi: 10.1126/science.285.5428.730
    [14] Upshaw JL, Leibson PJ (2006) NKG2D-mediated activation of cytotoxic lymphocytes: unique signaling pathways and distinct functional outcomes. Semin Immunol 18: 167–175. doi: 10.1016/j.smim.2006.03.001
    [15] Diefenbach A, Tomasello E, Lucas M, et al. (2002) Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol 3: 1142–1149. doi: 10.1038/ni858
    [16] Gilfillan S, Ho EL, Cella M, et al. (2002) NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol 3: 1150–1155. doi: 10.1038/ni857
    [17] Eagle RA, Trowsdale J (2007) Promiscuity and the single receptor: NKG2D. Nat Rev Immunol 7: 737–744. doi: 10.1038/nri2144
    [18] de Andrade LF, Smyth MJ, Martinet L (2013) DNAM-1 control of natural killer cells functions through nectin and nectin-like proteins. Immunol Cell Biol 92: 237–244.
    [19] Reymond N, Imbert AM, Devilard E, et al. (2004) DNAM-1 and PVR regulate monocyte migration through endothelial junctions. J Exp Med 199: 1331–1341. doi: 10.1084/jem.20032206
    [20] Iguchi-Manaka A, Kai H, Yamashita Y, et al. (2008) Accelerated tumor growth in mice deficient in DNAM-1 receptor. J Exp Med 205: 2959–2964. doi: 10.1084/jem.20081611
    [21] Gilfillan S, Chan CJ, Cella M, et al. (2008) DNAM-1 promotes activation of cytotoxic lymphocytes by nonprofessional antigen-presenting cells and tumors. J Exp Med 205: 2965–2973. doi: 10.1084/jem.20081752
    [22] Welch MJ, Teijaro JR, Lewicki HA, et al. (2012) CD8 T cell defect of TNF-α and IL-2 in DNAM-1 deficient mice delays clearance in vivo of a persistent virus infection. Virology 429: 163–170. doi: 10.1016/j.virol.2012.04.006
    [23] Shibuya A, Lanier LL, Phillips JH (1998) Protein kinase C is involved in the regulation of both signaling and adhesion mediated by DNAX accessory molecule-1 receptor. J Immunol 161: 1671–1676.
    [24] Shibuya K, Lanier LL, Phillips JH, et al. (1999) Physical and functional association of LFA-1 with DNAM-1 adhesion molecule. Immunity 11: 615–623. doi: 10.1016/S1074-7613(00)80136-3
    [25] Bottino C, Castriconi R, Pende D, et al. (2003) Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med 198: 557–567. doi: 10.1084/jem.20030788
    [26] Tahara-Hanaoka S, Shibuya K, Onoda Y, et al. (2004) Functional characterization of DNAM-1 (CD226) interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112). Int Immunol 16: 533–538. doi: 10.1093/intimm/dxh059
    [27] Fuchs A, Colonna M (2006) The role of NK cell recognition of nectin and nectin-like proteins in tumor immunosurveillance. Semin Cancer Biol 16: 359–366. doi: 10.1016/j.semcancer.2006.07.002
    [28] Stanietsky N, Rovis TL, Glasner A, et al. (2013) Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur J Immunol 43: 2138–2150. doi: 10.1002/eji.201243072
    [29] Pende D, Cantoni C, Rivera P, et al. (2001) Role of NKG2D in tumor cell lysis mediated by human NK cells: cooperation with natural cytotoxicity receptors and capability of recognizing tumors of nonepithelial origin. Eur J Immunol 31: 1076–1086. doi: 10.1002/1521-4141(200104)31:4<1076::AID-IMMU1076>3.0.CO;2-Y
    [30] Jinushi M, Takehara T, Tatsumi T, et al. (2003) Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int J Cancer 104: 354–361. doi: 10.1002/ijc.10966
    [31] Salih HR, Antropius H, Gieseke F, et al. (2003) Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. Blood 102: 1389–1396. doi: 10.1182/blood-2003-01-0019
    [32] Friese MA, Platten M, Lutz SZ, et al. (2003) MICA/NKG2D-mediated immunogene therapy of experimental gliomas. Cancer Res 63: 8996–9006.
    [33] Groh V, Rhinehart R, Secrist H, et al. (1999) Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc Natl Acad Sci USA 96: 6879–6884. doi: 10.1073/pnas.96.12.6879
    [34] Pende D, Spaggiari GM, Marcenaro S, et al. (2005) Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112). Blood 105: 2066–2073. doi: 10.1182/blood-2004-09-3548
    [35] Carlsten M, Björkström NK, Norell H, et al. (2007) DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res 67: 1317–1325. doi: 10.1158/0008-5472.CAN-06-2264
    [36] El-Sherbiny YM, Meade JL, Holmes TD, et al. (2007) The requirement for DNAM-1, NKG2D, and NKp46 in the natural killer cell-mediated killing of myeloma cells. Cancer Res 67: 8444–8449. doi: 10.1158/0008-5472.CAN-06-4230
    [37] Lakshmikanth T, Burke S, Ali TH, et al. (2009) NCRs and DNAM-1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo. J Clin Invest 119: 1251–1263. doi: 10.1172/JCI36022
    [38] Torelli GF, Peragine N, Raponi S, et al. (2014) Recognition of adult and pediatric acute lymphoblastic leukemia blasts by natural killer cells. Haematologica 99: 1248–1254. doi: 10.3324/haematol.2013.101931
    [39] Chan CJ, Andrews DM, McLaughlin NM, et al. (2010) DNAM-1/CD155 interactions promote cytokine and NK cell-mediated suppression of poorly immunogenic melanoma metastases. J Immunol 184: 902–911. doi: 10.4049/jimmunol.0903225
    [40] Paolini R, Bernardini G, Molfetta R, et al. (2015) NK cells and interferons. Cytokine Growth F R 2: 113–120.
    [41] Groh V, Rhinehart R, Randolph-Habecker J, et al. (2001) Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol 2: 255–260. doi: 10.1038/85321
    [42] Cosman D, Müllberg J, Sutherland CL, et al. (2001) ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14: 123–133. doi: 10.1016/S1074-7613(01)00095-4
    [43] Welte SA, Sinzger C, Lutz SZ, et al. (2003) Selective intracellular retention of virally induced NKG2D ligands by the human cytomegalovirus UL16 glycoprotein. Eur J Immunol 33: 194–203. doi: 10.1002/immu.200390022
    [44] Rölle A, Mousavi-Jazi M, Eriksson M, et al. (2003) Effects of human cytomegalovirus infection on ligands for the activating NKG2D receptor of NK cells: up-regulation of UL16-binding protein (ULBP)1 and ULBP2 is counteracted by the viral UL16 protein. J Immunol 171: 902–908. doi: 10.4049/jimmunol.171.2.902
    [45] Tomasec P, Wang EC, Davison AJ, et al. (2005) Downregulation of natural killer cell-activating ligand CD155 by human cytomegalovirus UL141. Nat Immunol 6: 181–188.
    [46] Cerboni C, Neri F, Casartelli N, et al. (2007) Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J Gen Virol 88: 242–250. doi: 10.1099/vir.0.82125-0
    [47] Ward J, Bonaparte M, Sacks J, et al. (2007) HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood 110: 1207–1214. doi: 10.1182/blood-2006-06-028175
    [48] Matusali G, Potestà M, Santoni A, et al. (2012) The human immunodeficiency virus type 1 Nef and Vpu proteins downregulate the natural killer cell-activating ligand PVR. J Virol 86: 4496–4504. doi: 10.1128/JVI.05788-11
    [49] Pappworth IY, Wang EC, Rowe M (2007) The switch from latent to productive infection in epstein-barr virus-infected B cells is associated with sensitization to NK cell killing. J Virol 81: 474–482. doi: 10.1128/JVI.01777-06
    [50] Sirén J, Sareneva T, Pirhonen J, et al. (2004) Cytokine and contact-dependent activation of natural killer cells by influenza A or Sendai virus-infected macrophages. J Gen Virol 85: 2357–2364. doi: 10.1099/vir.0.80105-0
    [51] Cerboni C, Zingoni A, Cippitelli M, et al. (2007) Antigen-activated human T lymphocytes express cell-surface NKG2D ligands via an ATM/ATR-dependent mechanism and become susceptible to autologous NK-cell lysis. Blood 110: 606–615. doi: 10.1182/blood-2006-10-052720
    [52] Ardolino M, Zingoni A, Cerboni C, et al. (2011) DNAM-1 ligand expression on Ag-stimulated T lymphocytes is mediated by ROS-dependent activation of DNA-damage response: relevance for NK-T cell interaction. Blood 117: 4778–4786. doi: 10.1182/blood-2010-08-300954
    [53] Ward J, Davis Z, DeHart J, et al. (2009) HIV-1 Vpr triggers natural killer cell-mediated lysis of infected cells through activation of the ATR-mediated DNA damage response. PLoS Pathog 5: e1000613. doi: 10.1371/journal.ppat.1000613
    [54] Richard J, Sindhu S, Pham TN, et al. (2010) HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood 115: 1354–1363. doi: 10.1182/blood-2009-08-237370
    [55] Vassena L, Giuliani E, Matusali G, et al. (2013) The human immunodeficiency virus type 1 Vpr protein upregulates PVR via activation of the ATR-mediated DNA damage response pathway. J Gen Virol 94: 2664–2669. doi: 10.1099/vir.0.055541-0
    [56] Gasser S, Orsulic S, Brown EJ, et al. (2005) The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436: 1186–1190. doi: 10.1038/nature03884
    [57] Soriani A, Zingoni A, Cerboni C, et al. (2009) ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood 113: 3503–3511. doi: 10.1182/blood-2008-08-173914
    [58] Croxford JL, Tang ML, Pan MF, et al. (2013) ATM-dependent spontaneous regression of early Eμ-myc-induced murine B-cell leukemia depends on natural killer and T cells. Blood 121: 2512–2521. doi: 10.1182/blood-2012-08-449025
    [59] Fionda C, Abruzzese MP, Zingoni AS, et al. (2015) Nitric oxide donors increase PVR/CD155 DNAM-1 ligand expression in multiple myeloma cells: role of DNA damage response activation. BMC Cancer 15: 17. doi: 10.1186/s12885-015-1023-5
    [60] Champsaur M, Lanier LL (2010) Effect of NKG2D ligand expression on host immune responses. Immunol Rev 235: 267–285. doi: 10.1111/j.0105-2896.2010.00893.x
    [61] Kamran N, Takai Y, Miyoshi J, et al. (2013) Toll-like receptor ligands induce expression of the costimulatory molecule CD155 on antigen-presenting cells. PLoS One 8: e54406. doi: 10.1371/journal.pone.0054406
    [62] Pignoloni B, Fionda C, Dell'Oste V, et al. (2016) Distinct roles for human cytomegalovirus immediate early proteins IE1 and IE2 in the transcriptional regulation of MICA and PVR/CD155 Expression. J Immunol 197: 4066–4078. doi: 10.4049/jimmunol.1502527
    [63] Solecki DJ, Gromeier M, Mueller S, et al. (2002) Expression of the human poliovirus receptor/CD155 gene is activated by sonic hedgehog. Biol Chem 277: 25697–25702. doi: 10.1074/jbc.M201378200
    [64] Hirota T, Irie K, Okamoto R, et al. (2005) Transcriptional activation of the mouse Necl-5/Tage4/PVR/CD155 gene by fibroblast growth factor or oncogenic Ras through the Raf-MEK-ERK-AP-1 pathway. Oncogene 24: 2229–2235. doi: 10.1038/sj.onc.1208409
    [65] Jung H, Hsiung B, Pestal K, et al. (2012) RAE-1 ligands for the NKG2D receptor are regulated by E2F transcription factors, which control cell cycle entry. J Exp Med 209: 2409–2422. doi: 10.1084/jem.20120565
    [66] Soriani A, Iannitto ML, Ricci B, et al. (2014) Reactive oxygen species- and DNA damage response-dependent NK cell activating ligand upregulation occurs at transcriptional levels and requires the transcriptional factor E2F1. J Immunol 193: 950–960. doi: 10.4049/jimmunol.1400271
    [67] Stern-Ginossar N, Gur C, Biton M, et al. (2008) Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D. Nat Immunol 9: 1065–1073. doi: 10.1038/ni.1642
    [68] Yadav D, Ngolab J, Lim RS, et al. (2009) Cutting edge: down-regulation of MHC class I-related chain A on tumor cells by IFN-gamma-induced microRNA. J Immunol 182: 39–43. doi: 10.4049/jimmunol.182.1.39
    [69] Nachmani D, Stern-Ginossar N, Sarid R, et al. (2009) Diverse herpesvirus microRNAs target the stress-induced immune ligand MICB to escape recognition by natural killer cells. Cell Host Microbe 5: 376–385. doi: 10.1016/j.chom.2009.03.003
    [70] Heinemann A, Zhao F, Pechlivanis S, et al. (2012) Tumor suppressive microRNAs miR-34a/c control cancer cell expression of ULBP2, a stress-induced ligand of the natural killer cell receptor NKG2D. Cancer Res 72: 460–471. doi: 10.1158/0008-5472.CAN-11-1977
    [71] Boutet P, Agüera-González S, Atkinson S, et al. (2009) Cutting edge: the metalloproteinase ADAM17/TNF-alpha-converting enzyme regulates proteolytic shedding of the MHC class I-related chain B protein. J Immunol 182: 49–53. doi: 10.4049/jimmunol.182.1.49
    [72] Waldhauer I, Steinle A (2006) Proteolytic release of soluble UL16-binding protein 2 from tumor cells. Cancer Res 66: 2520–2526. doi: 10.1158/0008-5472.CAN-05-2520
    [73] Zingoni A, Cecere F, Vulpis E, et al. (2015) Genotoxic stress induces senescence-associated ADAM10-dependent release of NKG2D MIC ligands in multiple myeloma cells. J Immunol 195: 736–748. doi: 10.4049/jimmunol.1402643
    [74] Ashiru O, Boutet P, Fernández-Messina L, et al. (2010) 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. doi: 10.1158/0008-5472.CAN-09-1688
    [75] Fernández-Messina L, Ashiru O, Boutet P, et al. (2010) Differential mechanisms of shedding of the glycosylphosphatidylinositol (GPI)-anchored NKG2D ligands. J Biol Chem 285: 8543–8551. doi: 10.1074/jbc.M109.045906
    [76] Iguchi-Manaka A, Okumura G, Kojima H, et al. (2016) Increased soluble CD155 in the serum of cancer patients. PLoS One 11: e0152982. doi: 10.1371/journal.pone.0152982
    [77] Ashiru O, Bennett NJ, Boyle LH, et al. (2009) NKG2D ligand MICA is retained in the cis-Golgi apparatus by human cytomegalovirus protein UL142. J Virol 83: 12345–12354. doi: 10.1128/JVI.01175-09
    [78] Bennett NJ, Ashiru O, Morgan FJ, et al. (2010) Intracellular sequestration of the NKG2D ligand ULBP3 by human cytomegalovirus. J Immunol 185: 1093–10102. doi: 10.4049/jimmunol.1000789
    [79] Prod'Homme V, Sugrue DM, Stanton RJ, et al. (2010) Human cytomegalovirus UL141 promotes efficient downregulation of the natural killer cell activating ligand CD112. J Gen Virol 91: 2034–2039. doi: 10.1099/vir.0.021931-0
    [80] Schepis D, D'Amato M, Studahl M, et al. (2009) Herpes simplex virus infection downmodulates NKG2D ligand expression. Scand J Immunol 69: 429–436. doi: 10.1111/j.1365-3083.2009.02241.x
    [81] Gong J, Fang L, Liu R, et al. (2014) UPR decreases CD226 ligand CD155 expression and sensitivity to NK cell-mediated cytotoxicity in hepatoma cells. Eur J Immunol 44: 3758–3767. doi: 10.1002/eji.201444574
    [82] Bologna S, Ferrari S (2013) It takes two to tango: ubiquitin and SUMO in the DNA damage response. Front Genet 4: 106.
    [83] Wimmer P, Schreiner S (2015) Viral mimicry to usurp ubiquitin and SUMO host pathways. Viruses 7: 4854–4872. doi: 10.3390/v7092849
    [84] Ardorisio S, Fierabracci A, Muscari I, et al. (2017) SUMO proteins: guardians of immune system. J Autoimmun 84: 21–28. doi: 10.1016/j.jaut.2017.09.001
    [85] Thomas M, Boname JM, Field S, et al. (2008) Down-regulation of NKG2D and NKp80 ligands by Kaposi's sarcoma-associated herpesvirus K5 protects against NK cell cytotoxicity. Proc Natl Acad Sci USA 105: 1656–1661. doi: 10.1073/pnas.0707883105
    [86] Nice TJ, Coscoy L, Raulet DH (2009) Posttranslational regulation of the NKG2D ligand Mult1 in response to cell stress. J Exp Med 206: 287–298. doi: 10.1084/jem.20081335
    [87] Nice TJ, Deng W, Coscoy L, et al. (2010) Stress-regulated targeting of the NKG2D ligand Mult1 by a membrane-associated RING-CH family E3 ligase. J Immunol 185: 5369–7536. doi: 10.4049/jimmunol.1000247
    [88] Zitti B, Molfetta R, Fionda C, et al. (2017) Innate immune activating ligand SUMOylation affects tumor cell recognition by NK cells. Sci Rep 7: 10445. doi: 10.1038/s41598-017-10403-0
    [89] Komander D, Rape M (2012) The ubiquitin code. Annu Rev Biochem 81: 203–229. doi: 10.1146/annurev-biochem-060310-170328
    [90] Varshavsky A (2017) The ubiquitin system, autophagy, and regulated protein degradation. Annu Rev Biochem 86: 123–128. doi: 10.1146/annurev-biochem-061516-044859
    [91] Molfetta R, Gasparrini F, Santoni A, et al. (2010) Ubiquitination and endocytosis of the high affinity receptor for IgE. Mol Immunol 47: 2427–2434. doi: 10.1016/j.molimm.2010.06.003
    [92] Wilkinson KA, Henley JM (2010) Mechanisms, regulation and consequences of protein SUMOylation. Biochem J 428: 133–145. doi: 10.1042/BJ20100158
    [93] Flotho A, Melchior F (2013) Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem 82: 357–385. doi: 10.1146/annurev-biochem-061909-093311
    [94] Fuertes MB, Girart MV, Molinero LL, et al. (2008) Intracellular retention of the NKG2D ligand MHC class I chain-related gene A in human melanomas confers immune privilege and prevents NK cell-mediated cytotoxicity. J Immunol 180: 4606–4614. doi: 10.4049/jimmunol.180.7.4606
    [95] Agüera-González S, Boutet P, Reyburn HT, et al. (2009) Brief residence at the plasma membrane of the MHC class I-related chain B is due to clathrin-mediated cholesterol-dependent endocytosis and shedding. J Immunol 182: 4800–4908. doi: 10.4049/jimmunol.0800713
    [96] Fernández-Messina L, Reyburn HT, Valés-Gómez M (2016) A short half-life of ULBP1 at the cell surface due to internalization and proteosomal degradation. Immunol Cell Biol 94: 479–485. doi: 10.1038/icb.2016.2
    [97] Castriconi R, Cantoni C, Della CM, et al. (2003) Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci USA 100: 4120–4125. doi: 10.1073/pnas.0730640100
    [98] Lee JC, Lee KM, Kim DW, et al. (2004) Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol 172: 7335–7340. doi: 10.4049/jimmunol.172.12.7335
    [99] Dasgupta S, Bhattacharya-Chatterjee M, Jr OB, et al. (2005) Inhibition of NK cell activity through TGF-beta 1 by down-regulation of NKG2D in a murine model of head and neck cancer. J Immunol 175: 5541–5550. doi: 10.4049/jimmunol.175.8.5541
    [100] Burgess SJ, Marusina AI, Pathmanathan I, et al. (2006) IL-21 down-regulates NKG2D/DAP10 expression on human NK and CD8+ T cells. J Immunol 176: 1490–1497. doi: 10.4049/jimmunol.176.3.1490
    [101] Zhang C, Zhang J, Niu J, et al. (2008) Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum Immunol 69: 490–500. doi: 10.1016/j.humimm.2008.06.004
    [102] Crane CA, Han SJ, Barry JJ, et al. (2010) TGF-beta downregulates the activating receptor NKG2D on NK cells and CD8+ T cells in glioma patients. Neuro Oncol 12: 7–13. doi: 10.1093/neuonc/nop009
    [103] Muntasell A, Magri G, Pende D, et al. (2010) Inhibition of NKG2D expression in NK cells by cytokines secreted in response to human cytomegalovirus infection. Blood 115: 5170–5179. doi: 10.1182/blood-2009-11-256479
    [104] Park YP, Choi SC, Kiesler P, et al. (2011) Complex regulation of human NKG2D-DAP10 cell surface expression: opposing roles of the γc cytokines and TGF-β1. Blood 118: 3019–3027. doi: 10.1182/blood-2011-04-346825
    [105] Groh V, Wu J, Yee C, et al. (2002) Tumor-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419: 734–738. doi: 10.1038/nature01112
    [106] Doubrovina ES, Doubrovin MM, Vider E, et al. (2003) Evasion from NK cell immunity by MHC class I chain-related molecules expressing colon adenocarcinoma. J Immunol 171: 6891–6899. doi: 10.4049/jimmunol.171.12.6891
    [107] Ogasawara K, Hamerman JA, Hsin H, et al. (2003) Impairment of NK cell function by NKG2D modulation in NOD mice. Immunity 18: 41–51. doi: 10.1016/S1074-7613(02)00505-8
    [108] Oppenheim DE, Roberts SJ, Clarke SL, et al. (2005) Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat Immunol 6: 928–937. doi: 10.1038/ni1239
    [109] Wiemann K, Mittrücker HW, Feger U, et al. (2005) Systemic NKG2D down-regulation impairs NK and CD8 T cell responses in vivo. J Immunol 175: 720–729. doi: 10.4049/jimmunol.175.2.720
    [110] Cerboni C, Ardolino M, Santoni A, et al. (2009) Detuning CD8+ T lymphocytes by down-regulation of the activating receptor NKG2D: role of NKG2D ligands released by activated T cells. Blood 113: 2955–2964. doi: 10.1182/blood-2008-06-165944
    [111] Molfetta R, Quatrini L, Capuano C, et al. (2014) c-Cbl regulates MICA- but not ULBP2-induced NKG2D down-modulation in human NK cells. Eur J Immunol 44: 2761–2770. doi: 10.1002/eji.201444512
    [112] Molfetta R, Quatrini L, Zitti B, et al. (2016) Regulation of NKG2D expression and signaling by endocytosis. Trends Immunol 37: 790–802. doi: 10.1016/j.it.2016.08.015
    [113] Roda-Navarro P, Reyburn HT (2009) The traffic of the NKG2D/Dap10 receptor complex during natural killer (NK) cell activation. J Biol Chem 284: 16463–16472. doi: 10.1074/jbc.M808561200
    [114] Quatrini L, Molfetta R, Zitti B, et al. (2015) Ubiquitin-dependent endocytosis of NKG2D-DAP10 receptor complexes activates signaling and functions in human NK cells. Sci Signal 8: ra108. doi: 10.1126/scisignal.aab2724
    [115] Horng T, Bezbradica JS, Medzhitov R (2007) NKG2D signaling is coupled to the interleukin 15 receptor signaling pathway. Nat Immunol 8: 1345–1352. doi: 10.1038/ni1524
    [116] Paolini R, Serra A, Molfetta R, et al. (1999) Tyrosine kinase-dependent ubiquitination of CD16 zeta subunit in human NK cells following receptor engagement. Eur J Immunol 29: 3179–3187. doi: 10.1002/(SICI)1521-4141(199910)29:10<3179::AID-IMMU3179>3.0.CO;2-9
    [117] Paolini R, Molfetta R, Piccoli M, et al. (2001) Ubiquitination and degradation of Syk and ZAP-70 protein tyrosine kinases in human NK cells upon CD16 engagement. Proc Natl Acad Sci USA 98: 9611–9616. doi: 10.1073/pnas.161298098
    [118] Capuano C, Romanelli M, Pighi C, et al. (2015) Anti-CD20 therapy acts via FcγRIIIA to diminish responsiveness of human natural killer cells. Cancer Res 75: 4097–4108. doi: 10.1158/0008-5472.CAN-15-0781
    [119] Capuano C, Pighi C, Molfetta R, et al. (2017) Obinutuzumab-mediated high-affinity ligation of FcγRIIIA/CD16 primes NK cells for IFNγ production. Oncoimmunology 6: e1290037. doi: 10.1080/2162402X.2017.1290037
    [120] Wilson EB, El-Jawhari JJ, Neilson AL, et al. (2011) Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS One 6: e22842. doi: 10.1371/journal.pone.0022842
    [121] Sanchez-Correa B, Gayoso I, Bergua JM, et al. (2012) Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol Cell Biol 90: 109–115. doi: 10.1038/icb.2011.15
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5363) PDF downloads(1181) Cited by(0)

Article outline

Figures and Tables

Figures(3)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog