Immune Netw. 2012 Dec;12(6):240-246. English.
Published online Dec 31, 2012.
Copyright © 2012 The Korean Association of Immunologists
Review

Signaling for Synergistic Activation of Natural Killer Cells

Hyung-Joon Kwon,1 and Hun Sik Kim1,2,3,4
    • 1Department of Medicine, Graduate School, University of Ulsan College of Medicine, Seoul 138-736, Korea.
    • 2Department of Microbiology, University of Ulsan College of Medicine, Seoul 138-736, Korea.
    • 3Cellular Dysfunction Research Center, University of Ulsan College of Medicine, Seoul 138-736, Korea.
    • 4Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul 138-736, Korea.
Received November 01, 2012; Revised November 12, 2012; Accepted November 15, 2012.

This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Natural killer (NK) cells play a pivotal role in early surveillance against virus infection and cellular transformation, and are also implicated in the control of inflammatory response through their effector functions of direct lysis of target cells and cytokine secretion. NK cell activation toward target cell is determined by the net balance of signals transmitted from diverse activating and inhibitory receptors. A distinct feature of NK cell activation is that stimulation of resting NK cells with single activating receptor on its own cannot mount natural cytotoxicity. Instead, specific pairs of co-activation receptors are required to unleash NK cell activation via synergy-dependent mechanism. Because each co-activation receptor uses distinct signaling modules, NK cell synergy relies on the integration of such disparate signals. This explains why the study of the mechanism underlying NK cell synergy is important and necessary. Recent studies revealed that NK cell synergy depends on the integration of complementary signals converged at a critical checkpoint element but not on simple amplification of the individual signaling to overcome intrinsic activation threshold. This review focuses on the signaling events during NK cells activation and recent advances in the study of NK cell synergy.

Keywords
Natural killer cell; Activating receptor; Signaling; Synergy

INTRODUCTION

Natural killer (NK) cells are derived from bone marrow-derived lymphocytes and belong to the innate immune system. In contrast to T and B cells, activation of NK cells is controlled by germ line-encoded receptors that do not undergo somatic DNA recombination and mutation. NK cells can provide early immune defense against virus-infected and transformed cells. Upon activation, NK cells eliminate target cells by polarized exocytosis of cytotoxic granules containing perforin, granzymes as well as Fas ligand (1). They also control immune responses by secretion of cytokines and chemokines including tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). NK cells via these effector functions play important roles in innate resistance to viral pathogens, tumor surveillance, shaping adaptive immunity, graft-versus-leukemia activity, antibody-independent natural cytotoxicity and antibodydependent cellular cytotoxicity (ADCC) against IgG-coated target cells.

The important role of NK cells in human health has been underscored by a wide spectrum of disease associations with NK cell dysfunction. Patients with leukocyte adhesion disorder (LAD), a syndrome due to the deficiency of β2 integrin subunit, display attenuated ADCC and natural cytotoxicity by NK cells (2, 3) and suffer severe recurrent infections of bacteria, infections of herpes simplex virus (HSV), and impaired immunity (4-6). NK cells from patients with hypohidrotic ectodermal dysplasia with immune deficiency (HED-ID) have defective natural cytotoxicity but not ADCC (6). In NK cells from patients with X-linked lymphoproliferative disease (XLP), activating receptors such as the signaling lymphocyte activation molecule (SLAM) family member 2B4 (CD244) and NTB-A induce inhibitory rather than activating signals as essential signaling adaptor SAP is deficient. Thus, 2B4 or NTB-A-mediated autologous killing of Epstein-Barr virus-infected B cells is abolished in XLP patients (7, 8).

Recently, NK cells have gained increasing attention and are now being considered as promising therapeutic tools for cancer immunotherapy due to their intrinsic ability to rapidly recognize and kill cancer cells, while sparing normal healthy cells. Allogeneic NK cells exhibit potent anti-tumor activities that are beneficial in the setting of hematopoietic stem cell transplantation into patients with acute myeloid leukemia (9). Moreover, KIR-HLA-incompatible NK cells were shown to lyse melanoma and renal cell carcinoma cells in vitro (10). Moreover, administration of rituximab (anti-CD20) to patients with large-cell non-Hodgkin's lymphoma induced NK cell cytotoxicity in vivo via ADCC (11).

Despite significant progress made on the role of NK cells as a pivotal sentinel in virus and tumor surveillance, the molecular mechanisms that regulate NK cell responses remain unclear, which restricts the use of NK cells as a therapeutic measure. NK cells possess a combinatorial array of activating and inhibitory receptors instead of a dominant single antigen receptor to sense their target cells. Whether NK cells are activated toward target cells or maintain tolerance to self is determined by the signal balance between activating and inhibitory receptors (12-14). Thus, understanding how signals from discrete activating receptors cooperate to control NK cell activation and how signals from inhibitory receptor antagonize such activation may provide strategies for harnessing NK cells in various clinical settings.

ACTIVATING RECEPTORS AND INTRACELLULAR SIGNALING

A large array of NK cell activating receptors that are structurally distinct has been characterized (15, 16). In contrast to inhibitory receptors that display variegated expression on NK cells, activating receptors are uniformly expressed on most NK cells. Furthermore, activating receptors trigger diverse and discrete signaling cascades, whereas inhibitory receptors induce a common signaling mechanism where their cytoplasmic immunoreceptor tyrosine-based inhibition motif (ITIM) recruits the phosphatase Src homology 2 domain-containing phosphatase 1 (SHP-1) to dephosphorylate signaling molecules in the activation of NK cells (17, 18).

Among activating receptors containing immunoreceptor tyrosine-based activation motif (ITAM) or associated with ITAM-carrying adapters are the low-affinity Fc receptor FcγRIIIa (CD16), natural cytotoxicity receptors (NCRs) such as NKp30 (CD337), NKp44 (CD336), and NKp46 (CD335), activating KIRs including KIR2DS and KIR3DS, and CD94/NKG2C (CD94/159c) heterodimer (16). Upon engagement, these ITAM-associated receptors are tyrosine-phosphorylated by Src family kinases, and the phosphorylated ITAM-bearing subunits recruit Syk and ZAP-70 tyrosine kinases. Thereafter, these kinases phosphorylate linker for activation of T cells (LAT) and non-T-cell activation linker (NTAL) leading to activation of PI3K, PLC-γ1, 2 and Vav 2, 3 (19-23).

Non-ITAM receptors include C-type lectin-like family member NKG2D (CD314), CD2, 2B4, CRACC (CD319), NTB-A, and the immunoglobulin-like type I transmembrane glycoprotein DNAM-1 (CD226). Upon stimulation, NKG2D associates with the adaptor protein DNAX-activating protein of 10 kD (DAP10) through its transmembrane region (24). The cytoplasmic tail of DAP10 carries a tyrosine-based signaling motif (YINM). This YINM, after phosphorylation by Src family kinases, recruits either a p85 subunit of PI3K or the small adaptor protein Grb2. Both interactions can activate Vav1 and PLC-γ2 and thus are essential for the activation of Ca2+ mobilization and cytotoxicity toward target cells (25). Unlike ITAM-associated receptors, NKG2D signaling is independent of Syk, ZAP70, LAT and NTAL (26-28). SLAM family receptors including 2B4, NTB-A, and CRACC are also phosphorylated by Src family kinases at immunoreceptor tyrosine-based switch motifs (ITSM) in their cytoplasmic domain (29). This ITSM, after phosphorylation, recruits small adaptor proteins such as SAP, EAT-2, and ERT (30-33). SAP then binds Src family kinase Fyn and this SAP-Fyn interaction is indispensable for 2B4-mediated NK cell activation (34, 35). Vav1, PLC-γ2, and c-Cbl are phosphorylated by 2B4 stimulation and implicated in such activation (31, 36). DNAM-1 signaling in NK cells remains largely unclear. Cytoplasmic tail of DNAM-1 can be phosphorylated by Src-family kinases and protein kinase C, and recruit actin-binding proteins (37, 38).

Although various signaling molecules implicated in NK cell activation have been uncovered, it has been difficult to identify critical molecules essential for NK cell activation because NK cells use diverse kinases, adaptors, and signaling modules depending on the engagement of different activating receptors. However, intersection of signals from discrete activating receptors by a single class of ITIM-associated inhibitory receptors implies that there exist critical control elements in the signaling pathway of NK cell activation. Such examples include PLC-γ and Vav proteins that play a non-redundant role in NK cell activation by multiple activating receptors, and their deficiency is manifested by functional defects of NK cells (20, 39, 40).

Src family kinases that include Lck, Fyn, Src, Lyn, Yes, and Fgr are important for NK cell activation as most of the activating receptors on NK cells are dependent on their activities for activation. However, no single Src family kinase is indispensable because their function is highly redundant for NK cell activation (41, 42). PLC-γ activation that induces Ca2+ mobilization from endoplasmic reticulum is critical for exocytosis of lytic granules and cytokine production. Although PLC-γ1 and PLC-γ2 are redundant in ITAM-dependent signaling, NKG2D/DAP10 and 2B4 signaling are highly dependent on PLC-γ2 (36, 40).

Vav has three isoforms, Vav1, Vav2, and Vav3, and NK cells express all of these isoforms (20). Vav acts as a guanine nucleotide exchange factor (GEF) that activates small GTPase proteins of the Rho family and as an adaptor protein through C-terminal SH2 and SH3 domains (43-45). It has an important role in the actin cytoskeleton dynamics for the co-activation receptor clustering and the lytic granule polarization, as well as in the lymphocyte receptor signaling including PLC-γ activation (45, 46). Vav1 is essential for DAP10 signaling, and Vav2 and Vav3 are required for ITAM-associated receptors (20, 47, 48). Thus, the identification of critical signaling molecules that function as essential checkpoints for NK cell activation provides clues for understanding the mechanism of cooperative interplay by multiple co-activation receptors.

SYNERGISTIC ACTIVATION OF NK CELLS

Activation of NK cells is tightly controlled by the requirement for the engagement of multiple co-activation receptors on NK cells upon encounter with target cells (16, 49). The combined signals from specific pairs of NK cell receptors, such as NKG2D and 2B4, can operate in synergy to induce potent cytotoxicity toward sensitive target cells (16, 50), while other combinations can result in different outcomes: no enhancement over each activating signal or simple additive effect. The mechanisms how distinct signals from different co-activation receptors are integrated to achieve proper functional activity are still unclear and difficult to study because each co-activation receptors use distinct signaling modules. Among the combinations of activating receptors, NKG2D and 2B4 as well as 2B4 and DNAM-1 are able to provide synergistic activation in resting NK cells, whereas NKG2D and DNAM-1 do not synergize. Even though NK cells are activated by synergistic signals from multiple co-activation receptors, inhibitory signals from a single class of ITIM-containing receptors, such as KIRs and CD94-NKG2A, are dominant over synergistic activating signals. This suggests that there are central common checkpoints where signals from multiple co-activation receptors are converged and integrated but inhibitory signals intersect.

A study with NKG2D and 2B4 synergy advocates such an example of checkpoint and an elaborate mechanism for regulating synergistic activation (36). The central role of Vav1 in the signaling pathway for NK cell activation is emphasized by its identification as an essential target of SHP-1 recruited to MHC class I-specific inhibitory receptors (51, 52). The Vav1 is phosphorylated by the engagement of NKG2D, 2B4 and DNAM-1 on NK cells and functions as an essential component in signaling from such activating receptors. Synergistic activation of NK cells via NKG2D and 2B4 combination results in synergistic phosphorylation of PLC-γ2, Ca2+ mobilization and cytotoxic degranulation, which are completely dependent on Vav1. Signals from single receptor activation can also induce Vav1 phosphorylation, but not the subsequent events such as the phosphorylation of PLC-γ2, Ca2+ mobilization, degranulation and cytokine secretion. Such dichotomy of NK cell activation can be explained by the finding that single receptor signal is unable to overcome activation threshold shaped by the E3 ubiquitin ligase c-Cbl mediating the inhibition of Vav1-dependent activation. Such threshold can be overcome by the convergence of signals from NKG2D and 2B4 to achieve optimal activation of Vav1 (Fig. 1). This mechanism supports the presence of a central checkpoint for NK cell activation where Vav1-mediated activation and c-Cbl-mediated inhibition compete with each other.

Figure 1
Proposed model for synergistic activation of NK cells. Complemetary phosphorylation of SLP-76 by synergizing receptors, NKG2D and 2B4, is required for optimal activation of Vav1 and thereby to overcome inhibition by c-Cbl.

Consecutive study of synergy between NKG2D and 2B4 revealed another interesting mechanism for synergy (53). The phosphorylation of Vav1 by NKG2D and 2B4 synergy is equivalent to the sum of the extent of phosphorylation induced by each receptor alone. In contrast, phosphorylation of Vav1 was not augmented by inactive receptor combination although each receptor induced Vav1 phosphorylation independently. Thus, synergy involves the integration of different activation signals for optimal activation of Vav1, but such requires combination of specific receptor pairs, suggesting the presence of an upstream regulatory element to control Vav1 activation. The adaptor proteins SH2 domain-containing leukocyte phosphoprotein of 76 kD (SLP-76) and LAT are known to contribute to Vav1 signaling and are essential components of forming signaling complexes in T cells (54, 55). Furthermore, the activation of PLC-γ, which is required for synergistic activation of NK cells (36), is dependent on both SLP-76 and LAT in T cells (56-58). In NK cells, the synergistic signaling pathway induced by co-engagement of NKG2D and 2B4 is largely dependent on SLP-76, but not LAT. Signal from NKG2D induces preferential phosphorylation at tyrosine (Tyr)128 of SLP-76, whereas signal from 2B4 induces phosphorylation of SLP-76 at Tyr113 (Fig. 1). Both tyrosine phosphorylations at Tyr113 and Tyr128 of SLP-76 are required for the synergistic activation of NK cells (53). The SLP-76 phosphorylation in NK cells is differentially regulated compared to TCR signaling in T cells because the single TCR engagement is sufficient for phosphorylation of both tyrosine residues in SLP-76 (54). Moreover, complementary phosphorylation of SLP-76 at N-terminal tyrosine residues is a property of synergizing receptors that induce natural cytotoxicity because the engagement of the Fcγ receptor CD16 also induces SLP-76 phosphorylation at both tyrosines. Selective phosphorylation of SLP-76 at Tyr113 or Tyr128 each enabled the binding of SLP-76 to Vav1 to promote Vav1 phosphorylation. Thus, SLP-76 may serve to integrate signals from synergizing co-activation receptors by recruiting Vav1 to its phosphorylated tyrosine residues. This suggests that permissive activation of NK cells is kept in check by the requirement for the complementary phosphorylation of SLP-76 during synergy among receptors that stimulate natural cytotoxicity.

CONCLUDING REMARKS

NK cell activation upon encounter with target cells is a result from dynamic process of signaling cascades triggered by combination of multiple receptors on NK cells. Thus, diverse mechanisms may control each step of NK cell activation depending on the signaling pathways originated from not only a single receptor but also a cooperation of co-activation receptors. In this review, the requirement for complementary phosphorylation of SLP-76 by synergizing receptors for optimal activation of Vav1 and thereby to overcome inhibition by c-Cbl is introduced as recently discovered mechanism regulating synergistic activation of NK cells. A challenge for future study is to find different checkpoints and signaling mechanisms that control synergistic activation of NK cells. This information will provide innovative and applicable strategies for exploiting NK cells in clinical immunotherapy.

Notes

The authors have no financial conflict of interest.

Abbreviations

NK natural killer
TNF-α tumor necrosis factor-α
IFN-γ interferon-γ
ADCC antibody-dependent cellular cytotoxicity
LAD leukocyte adhesion disorder
HSV herpes simplex virus
HED-ID hypohidrotic ectodermal dysplasia with immune deficiency
XLP X-linked lymphoproliferative disease
SLAM signaling lymphocyte activation molecule
ITIM immunoreceptor tyrosine-based inhibition motif
ITAM immunoreceptor tyrosine-based activation motif
SHP-1 Src homology 2 domain-containing phosphatase 1
NCRs natural cytotoxicity receptors
LAT linker for activation of T cells
NTAL non-T-cell activation linker
YINM tyrosine-based signaling motif
ITSM immunoreceptor tyrosine-based switch motif
GEF guanine nucleotide exchange factor
SLP-76 SH2 domain-containing leukocyte phosphoprotein of 76 kD

ACKNOWLEDGEMENTS

This study was supported by a National Research Foundation of Korea Grant from the Korean Government (NRF-2012-0003453) and by the National R&D Program for Cancer Control (1220030).

References

    1. Krzewski K, Coligan JE. Human NK cell lytic granules and regulation of their exocytosis. Front Immunol 2012;3:335.
    1. Kohl S, Loo LS, Schmalstieg FS, Anderson DC. The genetic deficiency of leukocyte surface glycoprotein Mac-1, LFA-1, p150,95 in humans is associated with defective antibody-dependent cellular cytotoxicity in vitro and defective protection against herpes simplex virus infection in vivo. J Immunol 1986;137:1688–1694.
    1. Anderson DC, Schmalsteig FC, Finegold MJ, Hughes BJ, Rothlein R, Miller LJ, Kohl S, Tosi MF, Jacobs RJ, Waldrop TC, et al. The severe and moderate phenotypes of heritable Mac-1, LFA-1 deficiency: their quantitative definition and relation to leukocyte dysfunction and clinical features. J Infect Dis 1985;152:668–689.
    1. Bunting M, Harris ES, McIntyre TM, Prescott SM, Zimmerman GA. Leukocyte adhesion deficiency syndromes: adhesion and tethering defects involving beta 2 integrins and selectin ligands. Curr Opin Hematol 2002;9:30–35.
    1. Inwald D, Davies EG, Klein N. Demystified...adhesion molecule deficiencies. Mol Pathol 2001;54:1–7.
    1. Orange JS. Human natural killer cell deficiencies and susceptibility to infection. Microbes Infect 2002;4:1545–1558.
    1. Parolini S, Bottino C, Falco M, Augugliaro R, Giliani S, Franceschini R, Ochs HD, Wolf H, Bonnefoy JY, Biassoni R, Moretta L, Notarangelo LD, Moretta A. X-linked lymphoproliferative disease. 2B4 molecules displaying inhibitory rather than activating function are responsible for the inability of natural killer cells to kill Epstein-Barr virus-infected cells. J Exp Med 2000;192:337–346.
    1. Bottino C, Falco M, Parolini S, Marcenaro E, Augugliaro R, Sivori S, Landi E, Biassoni R, Notarangelo LD, Moretta L, Moretta A. NTB-A [correction of GNTB-A], a novel SH2D1A-associated surface molecule contributing to the inability of natural killer cells to kill Epstein-Barr virus-infected B cells in X-linked lymphoproliferative disease. J Exp Med 2001;194:235–246.
    1. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, Posati S, Rogaia D, Frassoni F, Aversa F, Martelli MF, Velardi A. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002;295:2097–2100.
    1. Igarashi T, Wynberg J, Srinivasan R, Becknell B, McCoy JP Jr, Takahashi Y, Suffredini DA, Linehan WM, Caligiuri MA, Childs RW. Enhanced cytotoxicity of allogeneic NK cells with killer immunoglobulin-like receptor ligand incompatibility against melanoma and renal cell carcinoma cells. Blood 2004;104:170–177.
    1. Fischer L, Penack O, Gentilini C, Nogai A, Muessig A, Thiel E, Uharek L. The anti-lymphoma effect of antibody-mediated immunotherapy is based on an increased degranulation of peripheral blood natural killer (NK) cells. Exp Hematol 2006;34:753–759.
    1. Billadeau DD, Leibson PJ. ITAMs versus ITIMs: striking a balance during cell regulation. J Clin Invest 2002;109:161–168.
    1. Vivier E, Nunès JA, Vély F. Natural killer cell signaling pathways. Science 2004;306:1517–1519.
    1. Long EO. Negative signaling by inhibitory receptors: the NK cell paradigm. Immunol Rev 2008;224:70–84.
    1. Lanier LL. NK cell recognition. Annu Rev Immunol 2005;23:225–274.
    1. Bryceson YT, March ME, Ljunggren HG, Long EO. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol Rev 2006;214:73–91.
    1. Burshtyn DN, Scharenberg AM, Wagtmann N, Rajagopalan S, Berrada K, Yi T, Kinet JP, Long EO. Recruitment of tyrosine phosphatase HCP by the killer cell inhibitor receptor. Immunity 1996;4:77–85.
    1. Binstadt BA, Brumbaugh KM, Dick CJ, Scharenberg AM, Williams BL, Colonna M, Lanier LL, Kinet JP, Abraham RT, Leibson PJ. Sequential involvement of Lck and SHP-1 with MHC-recognizing receptors on NK cells inhibits FcR-initiated tyrosine kinase activation. Immunity 1996;5:629–638.
    1. 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.
    1. Cella M, Fujikawa K, Tassi I, Kim S, Latinis K, Nishi S, Yokoyama W, Colonna M, Swat W. Differential requirements for Vav proteins in DAP10- and ITAM-mediated NK cell cytotoxicity. J Exp Med 2004;200:817–823.
    1. Billadeau DD, Brumbaugh KM, Dick CJ, Schoon RA, Bustelo XR, Leibson PJ. The Vav-Rac1 pathway in cytotoxic lymphocytes regulates the generation of cell-mediated killing. J Exp Med 1998;188:549–559.
    1. Tassi I, Presti R, Kim S, Yokoyama WM, Gilfillan S, Colonna M. Phospholipase C-gamma 2 is a critical signaling mediator for murine NK cell activating receptors. J Immunol 2005;175:749–754.
    1. 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.
    1. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, Phillips JH. An activating immunoreceptor complex formed by NKG2D and DAP10. Science 1999;285:730–732.
    1. 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.
    1. 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.
    1. Zompi S, Hamerman JA, Ogasawara K, Schweighoffer E, Tybulewicz VL, Di Santo JP, Lanier LL, Colucci F. NKG2D triggers cytotoxicity in mouse NK cells lacking DAP12 or Syk family kinases. Nat Immunol 2003;4:565–572.
    1. Chiesa S, Mingueneau M, Fuseri N, Malissen B, Raulet DH, Malissen M, Vivier E, Tomasello E. Multiplicity and plasticity of natural killer cell signaling pathways. Blood 2006;107:2364–2372.
    1. Claus M, Meinke S, Bhat R, Watzl C. Regulation of NK cell activity by 2B4, NTB-A and CRACC. Front Biosci 2008;13:956–965.
    1. Sayos J, Wu C, Morra M, Wang N, Zhang X, Allen D, van Schaik S, Notarangelo L, Geha R, Roncarolo MG, Oettgen H, De Vries JE, Aversa G, Terhorst C. The X-linked lymphoproliferative-disease gene product SAP regulates signals induced through the co-receptor SLAM. Nature 1998;395:462–469.
    1. Eissmann P, Beauchamp L, Wooters J, Tilton JC, Long EO, Watzl C. Molecular basis for positive and negative signaling by the natural killer cell receptor 2B4 (CD244). Blood 2005;105:4722–4729.
    1. Eissmann P, Watzl C. Molecular analysis of NTB-A signaling: a role for EAT-2 in NTB-A-mediated activation of human NK cells. J Immunol 2006;177:3170–3177.
    1. Roncagalli R, Taylor JE, Zhang S, Shi X, Chen R, Cruz-Munoz ME, Yin L, Latour S, Veillette A. Negative regulation of natural killer cell function by EAT-2, a SAP-related adaptor. Nat Immunol 2005;6:1002–1010.
    1. Chan B, Lanyi A, Song HK, Griesbach J, Simarro-Grande M, Poy F, Howie D, Sumegi J, Terhorst C, Eck MJ. SAP couples Fyn to SLAM immune receptors. Nat Cell Biol 2003;5:155–160.
    1. Latour S, Roncagalli R, Chen R, Bakinowski M, Shi X, Schwartzberg PL, Davidson D, Veillette A. Binding of SAP SH2 domain to FynT SH3 domain reveals a novel mechanism of receptor signalling in immune regulation. Nat Cell Biol 2003;5:149–154.
    1. Kim HS, Das A, Gross CC, Bryceson YT, Long EO. Synergistic signals for natural cytotoxicity are required to overcome inhibition by c-Cbl ubiquitin ligase. Immunity 2010;32:175–186.
    1. Shibuya K, Shirakawa J, Kameyama T, Honda S, Tahara-Hanaoka S, Miyamoto A, Onodera M, Sumida T, Nakauchi H, Miyoshi H, Shibuya A. CD226 (DNAM-1) is involved in lymphocyte function-associated antigen 1 costimulatory signal for naive T cell differentiation and proliferation. J Exp Med 2003;198:1829–1839.
    1. Ralston KJ, Hird SL, Zhang X, Scott JL, Jin B, Thorne RF, Berndt MC, Boyd AW, Burns GF. The LFA-1-associated molecule PTA-1 (CD226) on T cells forms a dynamic molecular complex with protein 4.1G and human discs large. J Biol Chem 2004;279:33816–33828.
    1. Caraux A, Kim N, Bell SE, Zompi S, Ranson T, Lesjean-Pottier S, Garcia-Ojeda ME, Turner M, Colucci F. Phospholipase C-gamma2 is essential for NK cell cytotoxicity and innate immunity to malignant and virally infected cells. Blood 2006;107:994–1002.
    1. Ombrello MJ, Remmers EF, Sun G, Freeman AF, Datta S, Torabi-Parizi P, Subramanian N, Bunney TD, Baxendale RW, Martins MS, Romberg N, Komarow H, Aksentijevich I, Kim HS, Ho J, Cruse G, Jung MY, Gilfillan AM, Metcalfe DD, Nelson C, O'Brien M, Wisch L, Stone K, Douek DC, Gandhi C, Wanderer AA, Lee H, Nelson SF, Shianna KV, Cirulli ET, Goldstein DB, Long EO, Moir S, Meffre E, Holland SM, Kastner DL, Katan M, Hoffman HM, Milner JD. Cold urticaria, immunodeficiency, and autoimmunity related to PLCG2 deletions. N Engl J Med 2012;366:330–338.
    1. van Oers NS, Lowin-Kropf B, Finlay D, Connolly K, Weiss A. alpha beta T cell development is abolished in mice lacking both Lck and Fyn protein tyrosine kinases. Immunity 1996;5:429–436.
    1. Mason LH, Willette-Brown J, Taylor LS, McVicar DW. Regulation of Ly49D/DAP12 signal transduction by Src-family kinases and CD45. J Immunol 2006;176:6615–6623.
    1. Turner M, Billadeau DD. VAV proteins as signal integrators for multi-subunit immune-recognition receptors. Nat Rev Immunol 2002;2:476–486.
    1. Sylvain NR, Nguyen K, Bunnell SC. Vav1-mediated scaffolding interactions stabilize SLP-76 microclusters and contribute to antigen-dependent T cell responses. Sci Signal 2011;4:ra14.
    1. Saveliev A, Vanes L, Ksionda O, Rapley J, Smerdon SJ, Rittinger K, Tybulewicz VL. Function of the nucleotide exchange activity of vav1 in T cell development and activation. Sci Signal 2009;2:ra83.
    1. Tybulewicz VL. Vav-family proteins in T-cell signalling. Curr Opin Immunol 2005;17:267–274.
    1. Chan G, Hanke T, Fischer KD. Vav-1 regulates NK T cell development and NK cell cytotoxicity. Eur J Immunol 2001;31:2403–2410.
    1. Colucci F, Rosmaraki E, Bregenholt S, Samson SI, Di Bartolo V, Turner M, Vanes L, Tybulewicz V, Di Santo JP. Functional dichotomy in natural killer cell signaling: Vav1-dependent and -independent mechanisms. J Exp Med 2001;193:1413–1424.
    1. Bryceson YT, Ljunggren HG, Long EO. Minimal requirement for induction of natural cytotoxicity and intersection of activation signals by inhibitory receptors. Blood 2009;114:2657–2666.
    1. Lanier LL, Corliss B, Phillips JH. Arousal and inhibition of human NK cells. Immunol Rev 1997;155:145–154.
    1. Stebbins CC, Watzl C, Billadeau DD, Leibson PJ, Burshtyn DN, Long EO. Vav1 dephosphorylation by the tyrosine phosphatase SHP-1 as a mechanism for inhibition of cellular cytotoxicity. Mol Cell Biol 2003;23:6291–6299.
    1. Peterson ME, Long EO. Inhibitory receptor signaling via tyrosine phosphorylation of the adaptor Crk. Immunity 2008;29:578–588.
    1. Kim HS, Long EO. Complementary phosphorylation sites in the adaptor protein SLP-76 promote synergistic activation of natural killer cells. Sci Signal 2012;5:ra49.
    1. Koretzky GA, Abtahian F, Silverman MA. SLP76 and SLP65: complex regulation of signalling in lymphocytes and beyond. Nat Rev Immunol 2006;6:67–78.
    1. Balagopalan L, Coussens NP, Sherman E, Samelson LE, Sommers CL. The LAT story: a tale of cooperativity, coordination, and choreography. Cold Spring Harb Perspect Biol 2010;2:a005512.
    1. Beach D, Gonen R, Bogin Y, Reischl IG, Yablonski D. Dual role of SLP-76 in mediating T cell receptor-induced activation of phospholipase C-gamma1. J Biol Chem 2007;282:2937–2946.
    1. Zhang W, Sloan-Lancaster J, Kitchen J, Trible RP, Samelson LE. LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular activation. Cell 1998;92:83–92.
    1. Finco TS, Kadlecek T, Zhang W, Samelson LE, Weiss A. LAT is required for TCR-mediated activation of PLCgamma1 and the Ras pathway. Immunity 1998;9:617–626.

Metrics
Share
Figures

1 / 1

PERMALINK