NKG2D in CD8 PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Cellular and Molecular Immunology (2018) 14, 1–10

& 2018 CSI and USTC All rights reserved 2042-0226/18 $32.00
www.nature.com/cmi

REVIEW

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


immunotherapy
Kushal Prajapati, Cynthia Perez, Lourdes Beatriz Plaza Rojas, Brianna Burke and
Jose A Guevara-Patino
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.
Cellular and Molecular Immunology advance online publication, 5 February 2018; doi:10.1038/cmi.2017.161

Keywords: NKG2D; CD28; CD8+ T Cell; Cancer; Immunity; Auto-immunity; Memory; Immunotherapy

INTRODUCTION binding proteins 1–6 in humans.8 The expression of NKG2D


Natural killer group 2 member D (NKG2D) is a type II ligands on the cell surface is strictly regulated in the body.
transmembrane receptor encoded by the killer cell lectin-like Under healthy conditions, few or no NKG2D ligands are
receptor subfamily K member 1 (Klrk1) gene.1,2 NKG2D is expressed.6,7 However, expression of these ligands is
present on all murine and human NK cells. It is constitutively induced by transcriptional upregulation under stress-related
expressed on human CD8+ T cells, whereas its expression on conditions, such as infection and transformation.6,7 Notably,
CD8+ T cells occurs after activation in mice.3 In addition to other forms of cellular insult (e.g., exposure to DNA-damaging
NK and CD8+ T cells, NKG2D can also be expressed by natural agents, TLR signaling, cell proliferation or exposure to certain
killer T (NKT) cells, γδ T cells, activated murine macrophages cytokines) can also result in surface expression of these
and a small subgroup of CD4+ T cells.4 The functions of ligands.5,8,9
NKG2D, however, differ between NK cells and CD8+ T cells. NKG2D has a short intracellular domain that lacks any
In NK cells, NKG2D signaling alone is sufficient to mediate signaling motifs. In CD8+ T cells, NKG2D signals through the
direct killing of target cells,4 whereas in CD8+ T cells, adaptor protein DNAX-activating protein 10 (DAP10), whereas
simultaneous activation of the T-cell receptor (TCR) is in NK cells, it can pair with either DAP10 or its homolog
required for NKG2D to be functional. In these cells, NKG2D DAP12.3,10,11 DAP10 contains a YINM motif, which recruits
acts as a costimulatory receptor, and its signaling leads to p85 to induce PI3K signaling and Grb2 to activate Vav-SOS
enhanced TCR activation and T-cell function.3–5 signaling.12,13 Interestingly, the same YxNM signaling motif
NKG2D is a promiscuous receptor, binding to a variety of exists within the CD28 costimulatory receptor,14–16 suggesting
stress ligands. These ligands include members of the Rae-1ε that these motifs are the result of coevolutionary adaptation, as
and H60a-c families6,7 in mice and major histocompatibility both NKG2D and CD28 function as costimulatory receptors in
complex (MHC) class-I-related chain (MIC) A/B and UL16 CD8+ T cells. However, it is important to note that CD28 is

Loyola University Chicago, Oncology Institute, Maywood, IL 60153, USA


Correspondence: Dr JA Guevara, Loyola University Chicago, Oncology Institute, 2160 South First Avenue, Maywood, IL 60153, USA.
E-mail: jaguevara@luc.edu
Received: 30 September 2017; Revised: 30 November 2017; Accepted: 30 November 2017
Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

primarily required during the priming of naïve T cells, while motif, among other motifs (Figure 2).14–16 NKG2D, lacking
NKG2D is used by effector and memory CD8+ T cells. any signaling motif, relies on the YINM motif-containing
In addition to its costimulatory activity, NKG2D appears to adaptor protein DAP10 to initiate its signaling pathways.21,22
be involved in non-canonical functions, such as memory The NKG2D-DAP10 complex is expressed as a hexamer
formation, although the few studies that exist concerning this consisting of a homodimer of two NKG2D molecules, each
subject are conflicting. For instance, memory CD8+ T cells can bound to two DAP10 molecules (Figure 2).23 Thus, each CD28
be activated by cytokines only under specific conditions, which and NKG2D-DAP10 complex consists of two and four YxNM
transform these cells into NK-like killer cells. In such cases, signaling motifs, respectively. In addition to these structural
they eliminate target cells through an NKG2D-mediated and stoichiometric differences, CD28 expresses additional non-
mechanism without TCR engagement. Importantly, this YxNM signaling domains. These include the membrane
TCR-independent killing has been linked to the development proximal proline-rich PRRP domain, which can recruit
of several autoimmune disorders, highlighting the importance inducible-2 (IL-2) T-cell kinase, and the distal PYAP domain,
of NKG2D under pathophysiological conditions. This review which is capable of binding to Lck, Filamin A and Grb2.16
will focus on the less well-described characteristics of NKG2D. For both CD28 and NKG2D-DAP10 receptors, activation
The similarities and differences between NKG2D and CD28 leads to phosphorylation of tyrosine within the YxNM motif by
will be discussed, as well as the potential role of NKG2D in Src family kinase, which is followed by the recruitment of Grb2
CD8+ T-cell memory formation. Last, the involvement of and p85 subunits to asparagine and methionine residues,
NKG2D signaling under various pathological conditions, such respectively. This in turn results in the activation of a wide
as autoimmune diseases and cancer, will also be discussed array of well-characterized Grb2 → Vav and p85 → PDK1 → Akt
(Figure 1). signaling pathways, ultimately resulting in AP-1, NFAT and
NF-κB nuclear translocation and subsequent increases in cell
NKG2D AND CD28 COSTIMULATORY FUNCTIONS IN survival, proliferation and expression of effector molecules and
CD8+ T CELLS cytokines, as well as the release of cytolytic granules.14–16,24,25
In CD8+ T cells, engagement of the NK receptor, NKG2D, In NK cells, it has been shown that NKG2D-DAP10
results in enhanced TCR activation and function,1,3–5,9 which specifically stimulates phospholipase C-γ2 (PLC-γ2) down-
facilitates the recognition and destruction of target cells.3 The stream of the Grb2-Vav1 pathway.26 For CD28, no such
NKG2D receptor has been defined as a costimulatory molecule preference for PLC-γ1 has been described. It is thought that
in CD8+ T cells, and thus, it is often compared with CD28, an PLC-γ1 and PLC-γ2 have redundant functions,27,28 and thus
extensively studied costimulatory receptor in CD4+ and CD8+ the functional relevance of this preferential signaling of
T cells. Despite sharing many similarities, both the NKG2D NKG2D is not clear. It is important to note that although
and CD28 receptors possess distinct expression patterns, both of these isoforms are equally expressed in NK cells, PLC-
structures, signaling pathways and functional outcomes. γ1 is preferentially expressed over PLC-γ2 in T cells.29 Based on
In humans, both CD28 and NKG2D are expressed in CD8+ previous studies that have established PLC-γ as a key signaling
T cells; however, the proportions of these cells expressing these molecule for costimulation signaling, it is plausible that
receptors differ. NKG2D is expressed by all CD8+ T cells,3 NKG2D’s costimulatory potential is reduced compared with
whereas CD28 is expressed by ~ 50% of mature human CD8+ CD28 in CD8+ T cells. The same study demonstrated that
T cells, in addition to the vast majority (~95%) of human overexpression of PLC-γ2, but not PLC-γ1, led to increased
CD4+ T cells.17 The NKG2D receptor has gained increasing NKG2D-mediated cytotoxicity in the majority of human CTL
research interest based on the widespread expression of its clones.26 However, the differences between NKG2D and CD28
ligands compared to those of CD28 (CD80/CD86), which are signaling in the context of differential PLC-γ isoform expres-
expressed only in antigen-presenting cells (APCs). NKG2D sion were not addressed in this work.
ligands can be expressed in any cell type upon cell stress, In 2003, it was reported that NKG2D-DAP10 does not
including viral infection and DNA damage. This is also true in activate LAT, an important adaptor molecule that recruits key
cancer cells, underscoring the relevance of this receptor under proteins in T-cell signaling, in NK cells.30 Notably, CD28 has
pathophysiological conditions. While NKG2D ligands do not been shown to induce phosphorylation of LAT independent of
have any known association with inhibitory receptors, the Syk or ZAP70 kinases.31 One study reported that although
CD28 ligands B7.1 and B7.2 are shared by the inhibitory CD28 is necessary to phosphorylate LAT, its signaling motif-
receptor cytotoxic T-lymphocyte-associated protein-4 (CTLA- containing cytoplasmic tail is not involved in the initiation or
-4), which blocks CD28-dependent T-cell responses.18–20 persistence of TCR-induced LAT phosphorylation.32 NKG2D’s
Both NKG2D and CD28 receptors use a similar YxNM effects on LAT in the context of TCR stimulation remain to be
motif to transduce signaling pathways. However, they differ determined. As mentioned above, CD28 has two additional
remarkably in their structural properties and assembly, which signaling domains that are absent in DAP10, namely, PRRP
determine the unique functional outcomes of their signals. and PYAP. Itk binds to the PRRP domain, the importance of
CD28 is expressed as a homodimer on the cell surface with an which is not yet clear in CD28 signaling. PYAP recruits Lck,
extracellular ligand-binding domain, a transmembrane domain Filamin A and Grb2, and activates signaling pathways that lead
and an intracellular domain containing the YMNM signaling to cytoskeletal rearrangement and greater cytokine mRNA

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

unable to costimulate human and murine CD8+ T cells by


engaging the NKG2D receptor (unlike CD28), presumably due
to different culture conditions and/or cytokine milieu. Owing
to the unique characteristics of the signaling pathways dis-
cussed above, it is not altogether surprising that NKG2D and
CD28 would have different functional effects on T cells. In
experiments performed on DUC18 transgenic CTLs, only
CD28 could promote survival upon antigen encounter, whereas
only NKG2D could form immunological synapses in the
absence of antigen.34 Interestingly, in another study, Barber
and Sentman38 demonstrated that in either non-transduced or
NKG2D-transduced human CD8+ T cells, NKG2D engage-
ment specifically activated β-catenin, increased the expression
of β-catenin-induced genes and led to reduced production of
the anti-inflammatory cytokines IL-9, IL-10, IL-13 and vascular
endothelial growth factor-α in a peroxisome proliferator-
activated receptor-γ-dependent manner. CD28 stimulation
either failed to exert these effects or caused effects opposite
to that of NKG2D in this study. It was also found that both
CD28 and NKG2D stimulation decreased the migration of
human CD8+ T cells; however, only NKG2D activated Cdc42, a
Rho GTPase involved in T-cell motility, and only NKG2D’s
Figure 1 NKG2D’s role in physiology, diseases and its potential use
effects on T-cell migration were dependent on N-WASP.39
in medical interventions: In CD8+ T cells, NKG2D signaling is
thought to occur exclusively via DAP10 and requires simultaneous These data suggest that CD28 and NKG2D may also use
TCR engagement, and thus acting as an enhancer rather than an different signaling pathways to produce similar functional
inducer of activation. DAP10 uses a YINM motif to recruit PI3K outcomes in T cells. There is also evidence that both receptors
and Grb2, and activate the associated pathways. Major outcomes of can synergize with each other or modulate the other’s
the signaling is enhanced TCR signaling, cytotoxicity and enhanced function,36,38 suggesting possible crosstalk between the two
T-cell survival. However, alterations in the NKG2D signaling and receptors at certain signaling notches. Hu et al.40 studied the
expression levels can lead to autoimmune diseases that are either
effects of CD28 signaling on the expression of NKG2D and
TCR dependent: vitiligo, type I diabetes, and RA or TCR-
independent: celiac disease. Based on the contribution of NKG2D found that CD28 costimulation resulted in sustained NKG2D
signaling to CD8+ T-cell function, approaches are being expression in murine and human CD8+ T cells via Lck/JAK/
investigated for the treatment of cancer, autoimmunity and some STAT3 signaling.
infections diseases. DAP10, DNAX-activating protein 10; NKG2D, Collectively, both NKG2D and CD28, despite sharing a
natural killer group 2 member D; RA, rheumatoid arthritis; TCR, similar YxNM motif and associated upstream signaling events,
T-cell receptor. lead to unique signaling cascades, most likely due to different
structures, recruitment of adaptor proteins, downstream effec-
tors and possible crosstalk with other immune receptors. This
stability.16 Although it apparently lacks this signaling potential,
results in NKG2D- or CD28-mediated costimulation of CD8+
NKG2D-DAP10 has been linked to other non-YINM signaling T cells with unique features/functional outcomes. In addition
pathways. In experiments performed with human NK cells, to the above factors, differential expression levels of these
Sutherland et al.33 demonstrated that NKG2D stimulation leads receptors on T cells as well as those of the activating ligands in
to specific phosphorylation of JAK2 and STAT5. However, the different tissues and cytokine environments may dictate the
molecular basis and functional relevance of this pathway is not relevance of NKG2D and CD28 at the physiological level.
yet completely understood.
Both CD28 and NKG2D stimulation have been shown to NKG2D AND CD8+ T-CELL MEMORY FORMATION
have canonical costimulatory effects on T cells, including During the normal course of an immune response, the
increases in cell survival, proliferation, expression of effector formation of memory CD8+ T cells is one of the fundamental
molecules and cytokines, and release of cytolytic goals of the adaptive immune system. T-cell differentiation into
granules.14–16,24,25 Many studies have reported that NKG2D effector or memory cells is a tightly regulated process that is
is capable of costimulating human and mouse T cells to an still not fully understood. Current views suggest that T-cell
extent similar to that of CD28 at the level of proliferation, memory formation involves a passive transition from effector
cytolytic function and interferon-γ (IFN-γ) production.4,34–36 It to memory T cells and that memory is the result of initial
is important to note that factors such as cell activation status signaling cues promoting long-term survival.41 Among the
and cytokine treatment can alter the responsiveness of CD8+ known cues, CD4+ T cells and cytokines, such as IL-15, have
T cells to NKG2D. For instance, Lanier and co-workers37 were been shown to play an essential role in not only activating

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

Figure 2 Differences in the structure and signaling motifs of CD28


and NKG2D receptors: NKG2D-DAP10 complex is a hexameric
structure with two NKG2D molecules linked to total of four DAP10
adaptors, whereas CD28 is expressed as a homodimer on the cell
membrane. Both receptors contain YxNM signaling motif capable of
Figure 3 NKG2D rescues the function of CD4+ T-cell-unhelped
recruiting Grb2 and p85 subunit of PI3K. In addition, CD28
CD8+ T cells. (a–c) depict three possible scenarios of CD8+ T cells
possesses PRRP, and PYAP domains capable of recruiting Itk, Lck,
priming by an APC, which lead to three different outcomes for the
Fil A and Grb2. DAP10, DNAX-activating protein 10; ltk, IL-2-
CD8+ T cells: classical CD8+ T-cell priming by p:MHC-I (peptide-
inducible T-cell kinase; NKG2D, natural killer group 2 member D;
MHC-I complex) on APCs in the presence of CD4+ T-cell help,
PI3K, phosphatidylinositol-3 kinase.
which results in the optimal proliferation, CTL capacity and
formation of memory (a). These properties are defective when CD8+
T cell are activated in the absence of CD4+ T-cell help (b), which
CD8+ T cells but also programing them for memory
can be rescued by engaging NKG2D on CD8+ T cells during their
formation.42–47 However, recent work has increasingly estab- priming (c). APC, antigen-presenting cell; CTL, cytotoxic
lished that other factors, including NKG2D signaling, are T-lymphocyte; NKG2D, natural killer group 2 member D; MHC,
responsible for CD8+ T-cell memory formation.48–52 Mamma- major histocompatibility complex.
lian target of rapamycin complex 1 (mTORC1) has been
shown to play a pivotal role in deciding the fate of primed
CD8+ T cells. Low mTORC1 levels favor differentiation into NKG2D expression on CD8+ T cells and was independent of
memory precursor cells, while high levels of mTORC1 activity NK cells. This was shown in experiments using CD8+ T cells
leads to the development of terminally differentiated CD8+ T from NKG2D-deficient mice and NK cell-depleted animals.
cells.49 Interestingly, McQueen et al.49 demonstrated that both Further indicating the biological importance of these findings,
CD28 and NKG2D signaling activated the mTORC1 pathway we also determined that NKG2D engagement by antibody
but to different extents. CD28 signaling induced strong crosslinking was able to rescue the function of extremely
mTORC1 signaling, which led to the transcription of marker suppressed CD8+ T cells from HIV+ patients with low CD4+
genes of terminally differentiated effector CD8+ T cells, that is, T-cell counts. At the molecular level, we found that this rescue
T-bet, BLIMP1 or IRF-4. In contrast, activation of NKG2D was associated with the repression of T-bet, a transcription
weakly induced the mTORC1 signaling pathway,49 resulting in factor involved in the terminal differentiation of effector cells.52
the transcription of memory-associated genes, such as CD62L, Paradoxically, while NKG2D signaling was able to rescue CD8+
Eomes and CD127. Therefore, mTORC1 activation by CD28 T-cell memory formation, the initial effector response
and/or NKG2D mediates CD8+ T-cell differentiation into remained compromised.52 These studies indicate that NKG2D
terminally differentiated cells or memory cells. signaling on CD8+ T cells extends beyond known canonical
CD8+ T-cell priming in the absence of CD4+ T-cell help functions (e.g., facilitating target recognition and favored
results in the formation of so-called ‘helpless’ or ‘unhelped’ killing), by providing a prosurvival signal. Moreover, these
CD8+ T cells.42–44 Notably, these helpless CD8+ T cells have data support the hypothesis that the effector and memory
reduced effector functions and little to no memory functions of T cells can be disconnected, and CD8+ T-cell
formation.42–44 Published work from our lab has demonstrated memory formation can occur in the absence of an efficient
that immunological help for CD8+ T cells in memory forma- effector response.
tion can also be provided via NKG2D signaling.52 We demon- To date, few studies have investigated the role of NKG2D
strated, using a murine vaccination model of helpless CD8+ signaling in CD8+ T-cell memory formation. A study by
T cells overexpressing NKG2D ligands during immunization of Wensveen et al.51 showed that NKG2D signaling enhanced
mice depleted of CD4+ T cells, that these mice completely IL-15-mediated PI3K activity in activated CD8+ T cells, thus
restored the quantity and quality of the CD8+ T-cell memory assisting in memory commitment. In a series of adoptive
response (Figure 3). Importantly, this rescue was dependent on transfer experiments, the authors found that NKG2D

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

deficiency in OT-I cells resulted in impaired immunity against The expression of NKG2D ligands is strictly regulated in
challenge with LCMV expressing OVA. Moreover, in vitro- normal cells and little or no NKG2DL is expressed in healthy
activated T cells upregulated the antiapoptotic Mcl-1 protein, adult tissues.6 NKG2D ligands can be upregulated following
which was found to be downregulated in NKG2D-KO CD8+ T activation of the ATM/ATR (ataxia telangiectasia mutated/
cells,51 suggesting that NKG2D induces memory formation by ATM- and Rad3-related) DNA damage repair pathway. Studies
increasing levels of survival proteins, such as Mcl-1. In contrast have shown that DNA-damaging agents (fluorouracil or
to these findings, a study by Andre et al.48 suggested that cisplatin) or local ionizing radiation can induce NKG2D ligand
NKG2D had no major role in the generation and expansion of surface expression in an ATM/ATR-dependent manner.53 In
memory CD8+ T cells, but instead substantially enhanced the addition, activation of other pathways, for example, by TLR4
cytolytic effector responses of reactivated memory T cells, and TLR7/8 agonists, also induces NKG2D ligand expression.54
thereby contributing to efficacious tumor rejection. In this Moreover, cytokines such as IFN-γ and tumor growth factor-β
study, the authors used a transgenic mouse model in which the can affect the expression of NKG2D ligands. IFN-γ down-
human NKG2D ligand MHC class-I chain-related proteins A regulates the expression of NKG2D ligands in murine and
(MICA) was ubiquitously and constitutively expressed, result- human melanoma cells,55,56 while transforming growth factor-
ing in severe dysfunction of NKG2D signaling due to negative β has also been shown to decrease the transcription of several
feedback loops. These mice were used to assess the rejection of NKG2D ligands in glioma cells.57 Interestingly, studies have
OVA-expressing tumors (EG7) by tumor-specific memory demonstrated that inhibition of the proteasomal pathway can
CD8+ T cells. The authors observed that the generation and also induce the expression of NKG2D ligands.58 Previous
magnitude of memory CD8+ T-cell responses were indepen- research has shown that heat shock protein (HSP70) and
dent of NKG2D function, whereas in the effector phase, several NKG2D ligands share the same promoter sequence,
efficient tumor rejection by CD8+ T cells was dependent on which serves as a docking site for HSP 1 (HSF1).59 In response
to cellular stress, HSF1 is released in the cytoplasm. Following
NKG2D. However, there may be a possible relationship
nuclear translocation, HSF1 binds to the promoter regions of
between strong antigens and the need for NKG2D signaling.
both HSP70i and NKG2D ligands, initiating their
In this case, OVA antigen could be considered a strong TCR
transcription.59 Expression of NKG2D ligands acts as a clear
agonist, and OVA-reactive CD8+ T cells may therefore be
indicator of cell damage, which can be interpreted as suicide by
NKG2D-independent. Moreover, in these studies, NKG2D
proxy (in NKG2D-expressing cells). However, the spatiotem-
signaling was lacking throughout the life of the CD8+ T cells
poral control of HSP70i (prosurvival) and NKG2D ligand
(e.g., CD8+ NKG2D-KO T cells or dysfunctional NKG2D),
(suicide by proxy) expression in stressed cells remains to be
which may have unexpected effects on their peripheral
elucidated, representing an important area of translational
behavior.
research.
These studies indicate that CD8+ T-cell memory formation
is a dynamic process in which multiple factors have distinct NKG2D AND AUTOIMMUNITY
and redundant contributions. Based on these data, the role of The prevention of autoimmunity is regulated by both central
NKG2D signaling in memory formation may be to transcrip- and peripheral mechanisms. The central mechanisms involve
tionally certify CD8+ T cells that have actually recognized and thymic education, in which early self-reactive T cells that
killed stressed cells. While this may not apply to CD8+ T cells strongly react with peptide:MHC complexes undergo negative
with high-affinity TCRs, this may play an important role in selection in the thymus, during which cell death is induced.
CD8+ T cells that have weaker TCRs or are responding in a Nevertheless, many T cells expressing low-affinity TCRs against
subinflammatory environment. Moreover, these studies raise self-antigen are able to populate the periphery. In most cases,
important questions, such as whether the reduced quality of a these self-reactive T cells remain inactive. However, under
less-potent memory response is due only to the lack of NKG2D certain pathological conditions, they can become activated and
signaling or whether it is a reflection of a poor effector phase. mediate host tissue destruction, causing autoimmunity. There
Future studies will be required to fully delineate the role of is emerging evidence that NKG2D signaling is involved in
NKG2D in CD8+ T-cell memory formation. certain forms of T-cell-mediated autoimmunity.52,60,61
Although interactions between autoreactive TCRs and pep-
NKG2D LIGANDS AND THEIR REGULATION tide:MHC complexes are normally of low affinity, the outcome
In humans, NKG2D ligands include two families: MICA and of these interactions can be influenced by factors outside the
MICB and HCMV UL16-binding proteins (ULBP1–6). While TCR:peptide:MHC complex. NKG2D is an activating receptor
these ligands are HLA class I homologs, they do not associate that enhances TCR signaling,3 potentially reducing the thresh-
with β-2m and play no known role in antigen presentation. In old for T-cell activation via the TCR. In the great majority of
mice, there are three families of ligands for NKG2D: the Rae-1 cases, autoimmunity does not occur during CD8+ T-cell
family, which includes five members (Rae-1α, Rae-1β, Rae-1γ, responses against intracellular infections. However, in certain
Rae-1δ and Rae-1ε); the H60 family, which contains three instances, TCR-independent cytotoxic functions of NKG2D
members (H60a, H60b, H60c); and the UL16-binding protein- have been observed with higher expression levels on CD8+
like transcript 1 family, consisting only of MULT1. T cells when NKG2D is engaged by its ligands.

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

The involvement of NKG2D in potentiating CD8+ T-cell to IL-15. Alternatively, the same study reported that IL-15-
responses against low-affinity peptides has been demonstrated activated Jak3 could phosphorylate DAP10 and thus prime the
by our group and others in the context of antitumor immunity NKG2D-DAP10 signaling pathway. In this way, IL-15 was
and vitiligo.62 In the latter, there is an association between early capable of converting effector T cells into NK-like ‘lympho-
inflammatory events (e.g., exposure to UV light, a bleaching kine-activated killers’ (LAK cells) both in vitro and in vivo in
agent, phenols or trauma) and the onset of disease.63 This is celiac patients. Based on these data, it has been proposed that
significant, as the expression of NKG2D ligands is upregulated DAP10 couples with the IL-15 receptor. Notably, in this study,
in response to cell stress, rendering these cells visible to it was shown that the IL-15/NKG2D/DAP10 pathway conferred
NKG2D-expressing immune cells.1,3–5,9 Consistent with these TCR-independent cytolytic functions on T cells in the intestinal
observations, we demonstrated that engagement of NKG2D, epithelium.
but not 2B4 (another activating receptor), resulted in the In chronic inflammatory RA, data from Andersson and co-
exacerbation of CD8+ T-cell-mediated vitiligo using vaccine workers’60 work has demonstrated the expression of NKG2D
mouse models.62 These data indicate that autoimmune vitiligo and its ligands on human RA synovial cells and in the paws of
is limited by insufficient signaling, despite plentiful self-reactive arthritic mice. The authors demonstrated that, in established
T cells in the peripheral immune system. Importantly, studies collagen-induced arthritis, in vivo blockade of NKG2D with an
have also shown a direct role for CD8+ T cells expressing anti-NKG2D mAb ameliorated the disease and protected
NKG2D in another skin-related autoimmune disease, alopecia against pathologic joint damage. In this study, histologic
areata,64–66 indicating strong similarities between both diseases. analysis of arthritic joints from anti-NKG2D-treated mice
A similar situation has been reported for type I diabetes, in demonstrated significant joint protection compared with that
which mouse studies from the Lanier group demonstrated that of control mice. These studies presented compelling data
NKG2D is involved during the onset of the disease. The demonstrating that self-reactive CD8+ T cells can be activated
authors found that Rae-1 is present in prediabetic pancreas by NKG2D-mediated mechanisms that can lead to the
islets of non-obese diabetic mice and that autoreactive CD8+ destruction of host tissues. Considering that tumors are
T cells infiltrating the pancreas express NKG2D, resulting in stressed self-tissues, it would be expected that NKG2D signal-
the destruction of insulin-producing β-cells by T cells.67 ing plays a role in the recognition of NKG2D ligand-expressing
Additionally, treatment with a non-depleting anti-NKG2D cancer cells. Owing to its role in enhancing the cytotoxic
monoclonal antibody (mAb) during the prediabetic stage response of CD8+ T cells (in a TCR-dependent and TCR-
completely prevented disease by impairing the expansion and independent manner) and its pathophysiological role, NKG2D
function of autoreactive CD8+ T cells. may be able to be exploited as a target for autoimmune disease
In chronic inflammatory rheumatoid arthritis (RA), data treatment. Notably, since antitumor T-cell responses can be
from Andersson et al.60 work has demonstrated the expression viewed as a form of autoimmunity, the relationship between
of NKG2D and its ligands on human RA synovial cells and in these two different diseases may provide an opportunity for the
the paws of arthritic mice. The authors demonstrated that, in development of novel cancer therapies.
established collagen-induced arthritis, in vivo blockade of
NKG2D with an anti-NKG2D mAb ameliorated the disease NKG2D AND CANCER IMMUNITY
and protected against pathologic joint damage. In this study, Initial support for a role for NKG2D in tumor immunity was
histologic analysis of arthritic joints from anti-NKG2D-treated provided by studies using tumor cell lines engineered to express
mice demonstrated significant joint protection compared with NKG2D ligands. In these studies, mice were able to reject
that of control mice.60 These studies presented compelling data tumor cells expressing NKG2D ligands but not unmanipulated
demonstrating that self-reactive CD8+ T cells can be activated counterparts.70 The mechanisms mediating rejection were
by NKG2D-mediated mechanisms that can lead to the based on NK cells and CD8+ T cells. Subsequent studies by
destruction of host tissues. Considering that tumors are Smyth et al.71 using methylcholanthrene-treated mice demon-
stressed self-tissues, it would be expected that NKG2D signal- strated that in vivo NKG2D neutralization resulted in a higher
ing plays a role in the recognition of NKG2D ligand-expressing incidence of sarcoma. This observation was followed by studies
cancer cells. conducted by Guerra et al.70 using NKG2D-deficient mice
While NKG2D is a potent costimulator of TCR-mediated crossed with TRAMP mice (SV40 large T-driven prostate
effector functions, its physiological behavior is tightly associated cancer). In this case, the absence of NKG2D in TRAMP mice
with TCR function, and NKG2D is not expected to function resulted in early tumor development and growth compared
independently of TCR signaling.4,30,68 However, in celiac with TRAMP mice expressing NKG2D. These observations
disease, an autoimmune disease elicited by gluten intolerance demonstrated that tumor onset and progression were acceler-
characterized by the destruction of the small intestine, studies ated in the absence of NKG2D or NKG2D ligands.
by Bana Jabri and co-workers61,69 have shown that IL-15 drives Importantly, studies by Liu et al.72 demonstrated dual and
the upregulation of NKG2D, ultimately enabling CD8+ T cells opposing roles for NKG2D in cancer. The authors used mice
to kill in a TCR-independent manner through NKG2D- modified to express human MICB and a shedding-resistant
mediated mechanisms. The authors found that NK cells mutant MICB variant under the control of a prostate-specific
deficient in NKG2D-DAP10 expression were unable to respond promoter. These mice were then crossed to TRAMP mice.

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

TRAMP/MICB mice (MICB is shed) developed prostate NKG2D ligand expression on non-tumor cells, as part of the
tumors at a faster rate than TRAMP mice. However, response to activation of DNA repair mechanisms involving the
TRAMP/shedding-resistant-MICB mice remained tumor-free. ATM/ATR repair pathways, represents a safety concern. This
This interesting dichotomy was explained by elevated levels of could result in ‘on-target, off-tumor’ responses, leading to
soluble MICB and the associated depletion of NK cells in significant and potentially lethal toxicity. Indeed, murine
TRAMP/MICB mice. studies have demonstrated that adoptive transfer of NKG2D-
Challenging several of the conclusions drawn from these CAR T cells resulted in fatal responses.76,77 Importantly,
observations, studies using mice with severe dysfunction of investigators have established that the severity of such toxicity
NKG2D signaling due to systemic expression of MICA revealed varied, depending on the mouse strain used as the recipient.
that these mice were able to reject EG7 tumors with similar Specifically, BALB/c mice were more sensitive to toxicity than
efficacy as their wild-type counterparts.48 However, that EG7 C57BL/6 mice.77 These findings are of significance and must be
cells are a thymoma cell line engineered to express the highly considered when attempting to treat human patients, who are
immunogenic antigen chicken ovalbumin. Thus, it is possible highly heterogeneous. Thus, approaches that target tumor cells
that CD8+ T-cell responses against OVA-derived peptide will expressing NKG2D ligands must be considered with caution.
be mediated by high-affinity TCRs, which would be sufficient Notably, the use of NKG2D-CAR T cells is currently being
to mediate tumor killing in an NKG2D-independent manner. tested in a phase I clinical trial, and safety and toxicity results
Despite their differing conclusions, these studies emphasize are expected soon.
the important role of NKG2D in immune responses against NKG2D ligands represent an attractive target for therapies,
tumors and suggest the possibility of exploiting this antitumor and inducing their expression represents a viable approach for
role in the design of novel therapeutic approaches against the induction of antitumor immune responses. Irradiation
cancer. induces activation of the ATM/ATR repair pathways, resulting
in the expression of NKG2D ligands. To take advantage of this,
FINAL REMARKS one could envision the combined use of local (tumor) radiation
Based on the physiological role of NKG2D in immune and immune checkpoint therapies, namely, anti-PD-1 mAbs.
responses against stressed cells (both infected and cancerous Moreover, several drugs can trigger the expression of NKG2D
cells) and the broad expression of its ligands on cancer cells, ligands. Among these are DNA alkylating agents, proteasome
NKG2D is an attractive target for clinical development of novel inhibitors58 and HDAC inhibitors.78–80 Again, however, a key
therapeutic agents. Current strategies are focused on inducing consideration must be that the expression of NKG2D ligands is
NKG2D signaling during CD8+ T-cell priming (i.e., vaccine limited to cancer cells; otherwise, the development of auto-
approaches), engaging NKG2D receptors on effector cells (i.e., immune responses would be a likely outcome.
CD8+ T cells), targeting NKG2D ligands on cancer cells (i.e., In addition to cancer, NKG2D’s role has also been evaluated
NKG2D-CAR T cells), inducing ligands for NKG2D in cancer in anti-viral immunity in various studies. In patients with
cells (i.e., radiotherapy) and preventing the suppressive effects chronic hepatic viral infections, such as HBV and HCV,
of shed-off NKG2D ligands on immune cells (i.e., blocking NKG2D expression was found to be higher on intrahepatic
antibodies). CD8+ T cells and was able to costimulate HBV-specific CD8+
We have shown using a DNA vaccination approach that T cells.81 Similar costimulatory roles of NKG2D have been
genes encoding NKG2D ligands can be incorporated in a reported for virus-specific CD8+ T cells in cytomegalovirus.68
vaccine, along with the desired antigen. In this study, vaccina- Furthermore, in a study focusing on virus-induced encephalitis
tion with genes encoding Rae-1ε or H60 and the melanocyte in mice, blocking NKG2D receptors resulted in a reduction of
shared antigen Trp1 resulted in numerically and functionally the cytolytic capacity of the virus-specific CD8+ T cells and an
enhanced CD8+ T-cell immune responses against Trp1-derived increase in viral titers, as well as an increase in associated
peptides compared to vaccination with Trp1 alone.62 Other mortality.82 Along similar lines, blockade of NKG2D signaling
approaches are being considered, such as in vitro activation of led to reduced clearance of Theiler's murine encephalomyelitis
NKG2D and TCR pathways in CD8+ T cells before adoptive virus in the murine CNS.83 Recently, Kavazović et al.84 showed
cell transfer. This approach would provide transferred CD8+ that a lack of NKG2D on virus-specific murine CD8+ T cells
T cells with enhanced cytolytic and survival capacities optimal attenuated their cytokine production and ability to clear
for antitumor immune responses. One additional benefit may cytomegalovirus infection. Taken together, these data demon-
be that increasing or maintaining the expression of NKG2D in strate the important role of NKG2D in enhancing the effector
CD8+ T cells could result in resistance to immunosuppressive function of CD8+ T cells against viral infections. Based on these
factors in the tumor environment. and our findings, one can speculate that NKG2D-based
An alternative approach is the use of NKG2D-CAR T cells. approaches may be used for the treatment of intracellular
Here, the rationale is that these engineered T cells can eliminate pathogens. For example, in the case of HIV infection, which
tumors that express ligands for NKG2D. Studies in mouse results in low CD4+ T-cell levels, exogenous NKG2D activation
models of lymphoma, ovarian cancer and multiple myeloma would be expected to provide a useful substitute for CD4+
have shown that targeting NKG2D ligands with NKG2D-CAR T-cell assistance to CD8+ T cells. An additional benefit of
T cells results in effective antitumor responses.73–75 However, NKG2D signaling would be that the functionally exhausted

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

phenotype that characterizes CD8+ T cells from HIV patients 14 Ward SG. CD28: a signalling perspective. Biochem J 1996;
318(Part 2): 361–377.
may be reversed. Similar assumptions can also be made 15 Bocko D, Kosmaczewska A, Ciszak L, Teodorowska R, Frydecka I.
regarding the use of NKG2D in other chronic infections, such CD28 costimulatory molecule—expression, structure and function.
as HCV and HBV. Based on our current understanding of Arch Immunol Ther Exp (Warsz) 2002; 50: 169–177.
16 Boomer JS, Green JM. An enigmatic tail of CD28 signaling. Cold
NKG2D in autoimmunity, studies investigating how NKG2D Spring Harb Perspect Biol 2010; 2: a002436.
contributes to the onset of these diseases and its role in 17 Lee KP, Taylor C, Petryniak B, Turka LA, June CH, Thompson CB. The
autoimmune disease perpetuation are warranted. genomic organization of the CD28 gene. Implications for the regulation
of CD28 mRNA expression and heterogeneity. J Immunol 1990; 145:
Therapeutically, one can envision the development of 344–352.
NKG2D blocking reagents that could prevent the progression 18 Carreno BM, Collins M. The B7 family of ligands and its receptors: new
of disease by interfering with the recognition of NKG2D pathways for costimulation and inhibition of immune responses. Annu
Rev Immunol 2002; 20: 29–53.
ligands by CD8+ T cells. Based on the discussed studies, we 19 Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV et al.
strongly believe that continued efforts towards enhancing Biologic activity of cytotoxic T lymphocyte-associated antigen 4 anti-
immunity in cancer treatment and controlling autoimmunity body blockade in previously vaccinated metastatic melanoma and
ovarian carcinoma patients. Proc Natl Acad Sci USA 2003; 100:
will follow NKG2D-based approaches. 4712–4717.
In summary, although much remains to be elucidated 20 Walunas TL, Bakker CY, Bluestone JA. CTLA-4 ligation blocks CD28-
concerning NKG2D’s diverse roles in the immune system, dependent T cell activation. J Exp Med 1996; 183: 2541–2550.
21 Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL et al. An
significant evidence suggests that it may represent a novel activating immunoreceptor complex formed by NKG2D and DAP10.
therapeutic target for multiple historically intransigent diseases. Science 1999; 285: 730–732.
22 Upshaw JL, Arneson LN, Schoon RA, Dick CJ, Billadeau DD, Leibson
PJ. NKG2D-mediated signaling requires a DAP10-bound Grb2-Vav1
CONFLICT OF INTEREST intermediate and phosphatidylinositol-3-kinase in human natural
The authors declare no conflict of interest. killer cells. Nat Immunol 2006; 7: 524–532.
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.
1 Houchins JP, Yabe T, McSherry C, Bach FH. DNA sequence analysis of 24 Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat
NKG2, a family of related cDNA clones encoding type II integral Rev Immunol 2003; 3: 781–790.
membrane proteins on human natural killer cells. J Exp Med 1991; 25 Upshaw JL, Leibson PJ. NKG2D-mediated activation of cytotoxic
173: 1017–1020. lymphocytes: unique signaling pathways and distinct functional out-
2 Brown MG, Fulmek S, Matsumoto K, Cho R, Lyons PA, Levy ER et al. A comes. Semin Immunol 2006; 18: 167–175.
2-Mb YAC contig and physical map of the natural killer gene complex 26 Upshaw JL, Schoon RA, Dick CJ, Billadeau DD, Leibson PJ. The
on mouse chromosome 6. Genomics 1997; 42: 16–25. isoforms of phospholipase C-gamma are differentially used by distinct
3 Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL et al. human NK activating receptors. J Immunol 2005; 175: 213–218.
Activation of NK cells and T cells by NKG2D, a receptor for stress- 27 Irvin BJ, Williams BL, Nilson AE, Maynor HO, Abraham RT. Pleiotropic
inducible MICA. Science 1999; 285: 727–729. contributions of phospholipase C-gamma1 (PLC-gamma1) to T-cell
4 Jamieson AM, Diefenbach A, McMahon CW, Xiong N, Carlyle JR, antigen receptor-mediated signaling: reconstitution studies of a
Raulet DH. The role of the NKG2D immunoreceptor in immune cell PLC-gamma1-deficient Jurkat T-cell line. Mol Cell Biol 2000; 20:
activation and natural killing. Immunity 2002; 17: 19–29. 9149–9161.
5 Diefenbach A, Jamieson AM, Liu SD, Shastri N, Raulet DH. Ligands for 28 Fu G, Chen Y, Schuman J, Wang D, Wen R. Phospholipase Cgamma2
the murine NKG2D receptor: expression by tumor cells and activation
plays a role in TCR signal transduction and T cell selection. J Immunol
of NK cells and macrophages. Nat Immunol 2000; 1: 119–126.
2012; 189: 2326–2332.
6 Cerwenka A, Baron JL, Lanier LL. Ectopic expression of retinoic acid
29 Ting AT, Karnitz LM, Schoon RA, Abraham RT, Leibson PJ. Fc gamma
early inducible-1 gene (RAE-1) permits natural killer cell-mediated
receptor activation induces the tyrosine phosphorylation of both
rejection of a MHC class I-bearing tumor in vivo. Proc Natl Acad Sci
phospholipase C (PLC)-gamma 1 and PLC-gamma 2 in natural
USA 2001; 98: 11521–11526.
7 O'Callaghan CA, Cerwenka A, Willcox BE, Lanier LL, Bjorkman PJ. killer cells. J Exp Med 1992; 176: 1751–1755.
Molecular competition for NKG2D: H60 and RAE1 compete unequally 30 Billadeau DD, Upshaw JL, Schoon RA, Dick CJ, Leibson PJ. NKG2D-
for NKG2D with dominance of H60. Immunity 2001; 15: 201–211. DAP10 triggers human NK cell-mediated killing via a Syk-independent
8 Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W regulatory pathway. Nat Immunol 2003; 4: 557–564.
et al. ULBPs, novel MHC class I-related molecules, bind to CMV 31 Tsuchida M, Manthei ER, Knechtle SJ, Hamawy MM. CD28 ligation
glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D induces rapid tyrosine phosphorylation of the linker molecule LAT
receptor. Immunity 2001; 14: 123–133. in the absence of Syk and ZAP-70 tyrosine phosphorylation. Eur J
9 Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH et Immunol 1999; 29: 2354–2359.
al. Retinoic acid early inducible genes define a ligand family for the 32 Michel F, Attal-Bonnefoy G, Mangino G, Mise-Omata S, Acuto O. CD28
activating NKG2D receptor in mice. Immunity 2000; 12: 721–727. as a molecular amplifier extending TCR ligation and signaling
10 Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. NKG2D capabilities. Immunity 2001; 15: 935–945.
recruits two distinct adapters to trigger NK cell activation and 33 Sutherland CL, Chalupny NJ, Schooley K, VandenBos T, Kubin M,
costimulation. Nat Immunol 2002; 3: 1150–1155. Cosman D. UL16-binding proteins, novel MHC class I-related proteins,
11 Yim D, Jie HB, Sotiriadis J, Kim YS, Kim KS, Rothschild MF et al. bind to NKG2D and activate multiple signaling pathways in primary
Molecular cloning and characterization of pig immunoreceptor DAP10 NK cells. J Immunol 2002; 168: 671–679.
and NKG2D. Immunogenetics 2001; 53: 243–249. 34 Markiewicz MA, Carayannopoulos LN, Naidenko OV, Matsui K, Burack
12 Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte development, WR, Wise EL et al. Costimulation through NKG2D enhances murine
differentiation and activation. Nat Rev Immunol 2003; 3: 317–330. CD8+ CTL function: similarities and differences between NKG2D and
13 Park SG, Schulze-Luehrman J, Hayden MS, Hashimoto N, Ogawa W, CD28 costimulation. J Immunol 2005; 175: 2825–2833.
Kasuga M et al. The kinase PDK1 integrates T cell antigen receptor 35 Maasho K, Opoku-Anane J, Marusina AI, Coligan JE, Borrego F.
and CD28 coreceptor signaling to induce NF-kappaB and activate NKG2D is a costimulatory receptor for human naive CD8+ T cells.
T cells. Nat Immunol 2009; 10: 158–166. J Immunol 2005; 174: 4480–4484.

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

36 Rajasekaran K, Xiong V, Fong L, Gorski J, Malarkannan S. Functional 58 Butler JE, Moore MB, Presnell SR, Chan HW, Chalupny NJ, Lutz CT.
dichotomy between NKG2D and CD28-mediated co-stimulation in Proteasome regulation of ULBP1 transcription. J Immunol 2009; 182:
human CD8+ T cells. PLoS ONE 2010; 5. 6600–6609.
37 Ehrlich LI, Ogasawara K, Hamerman JA, Takaki R, Zingoni A, Allison 59 Venkataraman GM, Suciu D, Groh V, Boss JM, Spies T. Promoter
JP et al. Engagement of NKG2D by cognate ligand or antibody alone is region architecture and transcriptional regulation of the genes for the
insufficient to mediate costimulation of human and mouse CD8+ MHC class I-related chain A and B ligands of NKG2D. J Immunol
T cells. J Immunol 2005; 174: 1922–1931. 2007; 178: 961–969.
38 Barber A, Sentman CL. NKG2D receptor regulates human effector 60 Andersson AK, Sumariwalla PF, McCann FE, Amjadi P, Chang C,
T-cell cytokine production. Blood 2011; 117: 6571–6581. McNamee K et al. Blockade of NKG2D ameliorates disease in mice
39 Serrano-Pertierra E, Cernuda-Morollon E, Lopez-Larrea C. NKG2D- and with collagen-induced arthritis: a potential pathogenic role in chronic
CD28-mediated costimulation regulate CD8+ T cell chemotaxis inflammatory arthritis. Arthritis Rheum 2011; 63: 2617–2629.
through different mechanisms: the role of Cdc42/N-WASp. J Leukocyte 61 Meresse B, Chen Z, Ciszewski C, Tretiakova M, Bhagat G, Krausz TN et
Biol 2014; 95: 487–495. al. Coordinated induction by IL15 of a TCR-independent NKG2D
40 Hu J, Batth IS, Xia X, Li S. Regulation of NKG2D(+)CD8(+) T-cell- signaling pathway converts CTL into lymphokine-activated killer cells
mediated antitumor immune surveillance: identification of a novel in celiac disease. Immunity 2004; 21: 357–366.
CD28 activation-mediated, STAT3 phosphorylation-dependent 62 Zloza A, Lyons GE, Chlewicki LK, Kohlhapp FJ, O'Sullivan JA, Lacek
mechanism. Oncoimmunology 2016; 5: e1252012. AT et al. Engagement of NK receptor NKG2D, but not 2B4, results in
41 Kaech SM, Cui W. Transcriptional control of effector and memory self-reactive CD8+ T cells and autoimmune vitiligo. Autoimmunity
CD8+ T cell differentiation. Nat Rev Immunol 2012; 12: 749–761. 2011; 44: 599–606.
42 Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, 63 Le Poole IC, Wankowicz-Kalinska A, van den Wijngaard RM, Nickoloff
Schoenberger SP. CD4+ T cells are required for secondary expansion BJ, Das PK. Autoimmune aspects of depigmentation in vitiligo. J
and memory in CD8+ T lymphocytes. Nature 2003; 421: 852–856. Invest Dermatol Symp Proc 2004; 9: 68–72.
43 Shedlock DJ, Shen H. Requirement for CD4 T cell help in generating 64 Dai Z, Xing L, Cerise J, Wang EH, Jabbari A, de Jong A et al. CXCR3
functional CD8 T cell memory. Science 2003; 300: 337–339. blockade inhibits T cell migration into the skin and prevents develop-
44 Sun JC, Bevan MJ. Defective CD8 T cell memory following acute ment of alopecia areata. J Immunol 2016; 197: 1089–1099.
infection without CD4 T cell help. Science 2003; 300: 339–342. 65 Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y et
45 Schluns KS, Lefrancois L. Cytokine control of memory T-cell develop- al. Genome-wide association study in alopecia areata implicates both
ment and survival. Nat Rev Immunol 2003; 3: 269–279. innate and adaptive immunity. Nature 2010; 466: 113–117.
46 Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL, Embers M et 66 Xing L, Dai Z, Jabbari A, Cerise JE, Higgins CA, Gong W et al. Alopecia
al. Reversible defects in natural killer and memory CD8 T cell lineages areata is driven by cytotoxic T lymphocytes and is reversed by JAK
in interleukin 15-deficient mice. J Exp Med 2000; 191: 771–780. inhibition. Nat Med 2014; 20: 1043–1049.
47 Schluns KS, Williams K, Ma A, Zheng XX, Lefrancois L. Cutting edge: 67 Ogasawara K, Hamerman JA, Ehrlich LR, Bour-Jordan H, Santamaria
requirement for IL-15 in the generation of primary and memory P, Bluestone JA et al. NKG2D blockade prevents autoimmune diabetes
antigen-specific CD8 T cells. J Immunol 2002; 168: 4827–4831. in NOD mice. Immunity 2004; 20: 757–767.
48 Andre MC, Sigurdardottir D, Kuttruff S, Pommerl B, Handgretinger R, 68 Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR,
Rammensee HG et al. Impaired tumor rejection by memory CD8 T cells Spies T. Costimulation of CD8alphabeta T cells by NKG2D via
engagement by MIC induced on virus-infected cells. Nat Immunol
in mice with NKG2D dysfunction. Int J Cancer 2012; 131:
2001; 2: 255–260.
1601–1610.
69 Tang F, Chen Z, Ciszewski C, Setty M, Solus J, Tretiakova M et al.
49 McQueen B, Trace K, Whitman E, Bedsworth T, Barber A. Natural
Cytosolic PLA2 is required for CTL-mediated immunopathology of
killer group 2D and CD28 receptors differentially activate mammalian/
celiac disease via NKG2D and IL-15. J Exp Med 2009; 206:
mechanistic target of rapamycin to alter murine effector CD8+ T-cell
707–719.
differentiation. Immunology 2016; 147: 305–320.
70 Guerra N, Tan YX, Joncker NT, Choy A, Gallardo F, Xiong N et al.
50 Soderquest K, Walzer T, Zafirova B, Klavinskis LS, Polic B, Vivier E et
NKG2D-deficient mice are defective in tumor surveillance in models of
al. Cutting edge: CD8+ T cell priming in the absence of NK cells leads
spontaneous malignancy. Immunity 2008; 28: 571–580.
to enhanced memory responses. J Immunol 2011; 186: 3304–3308. 71 Smyth MJ, Swann J, Cretney E, Zerafa N, Yokoyama WM, Hayakawa Y.
51 Wensveen FM, Lenartic M, Jelencic V, Lemmermann NA, ten Brinke A,
NKG2D function protects the host from tumor initiation. J Exp Med
Jonjic S et al. NKG2D induces Mcl-1 expression and mediates survival 2005; 202: 583–588.
of CD8 memory T cell precursors via phosphatidylinositol 3-kinase. J 72 Liu G, Lu S, Wang X, Page ST, Higano CS, Plymate SR et al.
Immunol 2013; 191: 1307–1315. Perturbation of NK cell peripheral homeostasis accelerates prostate
52 Zloza A, Kohlhapp FJ, Lyons GE, Schenkel JM, Moore TV, Lacek AT et carcinoma metastasis. J Clin Invest 2013; 123: 4410–4422.
al. NKG2D signaling on CD8(+) T cells represses T-bet and rescues 73 Barber A, Zhang T, Sentman CL. Immunotherapy with chimeric
CD4-unhelped CD8(+) T cell memory recall but not effector responses. NKG2D receptors leads to long-term tumor-free survival and develop-
Nat Med 2012; 18: 422–428. ment of host antitumor immunity in murine ovarian cancer. J Immunol
53 Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway 2008; 180: 72–78.
regulates innate immune system ligands of the NKG2D receptor. 74 Zhang T, Barber A, Sentman CL. Chimeric NKG2D modified T cells
Nature 2005; 436: 1186–1190. inhibit systemic T-cell lymphoma growth in a manner involving multi-
54 Eissmann P, Evans JH, Mehrabi M, Rose EL, Nedvetzki S, Davis DM. ple cytokines and cytotoxic pathways. Cancer Res 2007; 67:
Multiple mechanisms downstream of TLR-4 stimulation allow expres- 11029–11036.
sion of NKG2D ligands to facilitate macrophage/NK cell crosstalk. J 75 Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma
Immunol 2010; 184: 6901–6909. with adoptively transferred chimeric NKG2D receptor-expressing
55 Schwinn N, Vokhminova D, Sucker A, Textor S, Striegel S, Moll I et al. T cells. Gene Ther 2011; 18: 509–516.
Interferon-gamma down-regulates NKG2D ligand expression and 76 VanSeggelen H, Hammill JA, Dvorkin-Gheva A, Tantalo DG, Kwiecien
impairs the NKG2D-mediated cytolysis of MHC class I-deficient JM, Denisova GF et al. T cells engineered with chimeric antigen
melanoma by natural killer cells. Int J Cancer 2009; 124: receptors targeting NKG2D ligands display lethal toxicity in mice. Mol
1594–1604. Ther 2015; 23: 1600–1610.
56 Bui JD, Carayannopoulos LN, Lanier LL, Yokoyama WM, Schreiber RD. 77 Lynn RC, Powell DJ Jr.. Strain-dependent lethal toxicity in NKG2D
IFN-dependent down-regulation of the NKG2D ligand H60 on tumors. ligand-targeted CAR T-cell therapy. Mol Ther 2015; 23: 1559–1561.
J Immunol 2006; 176: 905–913. 78 Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N.
57 Eisele G, Wischhusen J, Mittelbronn M, Meyermann R, Waldhauer I, Cancer cells become susceptible to natural killer cell killing after
Steinle A et al. TGF-beta and metalloproteinases differentially suppress exposure to histone deacetylase inhibitors due to glycogen synthase
NKG2D ligand surface expression on malignant glioma cells. Brain kinase-3-dependent expression of MHC class I-related chain A and B.
2006; 129: 2416–2425. Cancer Res 2005; 65: 11136–11145.

Cellular and Molecular Immunology


Functions of NKG2D in CD8+ T cells
JA Guevara-Patino et al

10

79 Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M et al. 82 Walsh KB, Lanier LL, Lane TE. NKG2D receptor signaling enhances
Natural killer cell-mediated lysis of hepatoma cells via specific cytolytic activity by virus-specific CD8+ T cells: evidence for a
induction of NKG2D ligands by the histone deacetylase inhibitor protective role in virus-induced encephalitis. J Virol 2008; 82:
sodium valproate. Cancer Res 2005; 65: 6321–6329. 3031–3044.
80 Berghuis D, Schilham MW, Vos HI, Santos SJ, Kloess S, Buddingh EP 83 Deb C, Howe CL. NKG2D contributes to efficient clearance of
et al. Histone deacetylase inhibitors enhance expression of NKG2D picornavirus from the acutely infected murine brain. J Neurovirol
ligands in Ewing sarcoma and sensitize for natural killer cell-mediated 2008; 14: 261–266.
cytolysis. Clin Sarcoma Res 2012; 2: 8. 84 Kavazovic I, Lenartic M, Jelencic V, Jurkovic S, Lemmermann NAW,
81 Kennedy PT, Gehring AJ, Nowbath A, Selden C, Quaglia A, Dhillon A et Jonjic S et al. NKG2D stimulation of CD8(+) T cells during
al. The expression and function of NKG2D molecule on intrahepatic priming promotes their capacity to produce cytokines in
CD8+ T cells in chronic viral hepatitis. J Viral Hepatol 2008; 15: response to viral infection in mice. Eur J Immunol 2017; 47:
901–909. 1123–1135.

Cellular and Molecular Immunology

You might also like