Immune Netw. 2023 Feb;23(1):e8. English.
Published online Feb 20, 2023.
Copyright © 2023. The Korean Association of Immunologists
Review

Human CD8+ T-Cell Populations That Express Natural Killer Receptors

June-Young Koh,1,2, Dong-Uk Kim,1, Bae-Hyeon Moon,1, and Eui-Cheol Shin1,3
    • 1Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
    • 2Genome Insight, Inc., Daejeon 34051, Korea.
    • 3The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea.
Received December 28, 2022; Revised February 07, 2023; Accepted February 07, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

CD8+ T cells are activated by TCRs that recognize specific cognate Ags, while NK-cell activation is regulated by a balance between signals from germline-encoded activating and inhibitory NK receptors. Through these different processes of Ag recognition, CD8+ T cells and NK cells play distinct roles as adaptive and innate immune cells, respectively. However, some human CD8+ T cells have been found to express activating or inhibitory NK receptors. CD8+ T-cell populations expressing NK receptors straddle the innate-adaptive boundary with their innate-like features. Recent breakthrough technical advances in multi-omics analysis have enabled elucidation of the unique immunologic characteristics of these populations. However, studies have not yet fully clarified the heterogeneity and immunological characteristics of each CD8+ T-cell population expressing NK receptors. Here we aimed to review the current knowledge of various CD8+ T-cell populations expressing NK receptors, and to pave the way for delineating the landscape and identifying the various roles of these T-cell populations.

Keywords
CD8-Positive T-Lymphocytes; Natural Killer Cell Receptors; CD56 Antigen; Receptors, KIR; NKG2A Receptor; NKG2C Receptor

INTRODUCTION

Although both human NK and CD8+ T cells are representative cytotoxic lymphocytes, they have distinct characteristics as innate and adaptive immune cells, respectively. NK-cell activation is regulated by a balance between signals from germline-encoded activating and inhibitory NK receptors. Activating NK receptors—such as NKG2D, NKp44, NKp30, and NKG2C—recognize ligands that are mainly expressed on aberrant cells, e.g., virus-infected, transformed, or stressed cells. On the other hand, inhibitory NK receptors—such as killer cell immunoglobulin-like receptors (KIRs) and NKG2A—recognize MHC ligands that are expressed on normal healthy cells (1, 2). As adaptive immune cells, CD8+ T cells are activated by TCRs that recognizes specific epitopes presented by MHC class I (MHC-I) molecules and exert effector functions (3). The CD8+ T-cell population exhibits a highly diverse TCR repertoire, enabling CD8+ T cells to respond against many different Ags.

Some subsets of T cells such as mucosal-associated invariant T cells, invariant natural killer T cells and γδ T cells express TCRs with limited diversity and exert innate-like functions. Apart from those subsets, CD8+ T cells can express activating or inhibitory NK receptors, including KIRs, NKG2A, and NKG2C. However, studies have not yet comprehensively elucidated the immunological characteristics of CD8+ T-cell subpopulations expressing each NK receptor.

In this review, we provide an overview of CD8+ T-cell subpopulations expressing NK receptors. First, we describe CD8+ T cells expressing CD56, with regard to their innateness and NK receptor expression. Next, we provide a structured review of CD8+ T-cell subpopulations expressing KIR, NKG2A, or NKG2C. Finally, we discuss the physiological and pathological roles of CD8+ T-cell subpopulations expressing NK receptors. We do not describe NKG2D because NKG2D is expressed by all types of human CD8+ T cells, not by a CD8+ T-cell subpopulation.

CD8+ T CELLS EXPRESSING CD56

CD56—also known as neural cell adhesion molecule (NCAM)—is the first characterized immunoglobulin superfamily member engaged in cell adhesion (4). CD56 serves as a classical lineage marker for human NK cells, which are defined as CD3CD56+ lymphocytes (5). Additionally, the CD56 expression level is used to define functionally distinct subsets of NK cells—with a CD56bright NK-cell subset showing elevated cytokine production (6, 7), and a CD56dim NK-cell subset exhibiting enhanced cytotoxicity and a mature differentiation status (8).

In this background, early investigators used the term “natural T cells” to describe CD56-expressing T cells (9, 10, 11). This CD56+ natural T-cell population comprises heterogeneous T-cell subsets, including γδ T cells and CD4+ T cells, but mainly CD8+ T cells with conventional TCRs (11, 12). CD56+ T cells are distinguished from CD56 T cells and invariant T cells, in terms of TCR clonality, surface protein phenotypes, and genome-wide transcriptional patterns. Compared with CD56 T cells, CD56+ T cells exhibit higher expressions of NK-cell-related molecules (e.g., CD16, CD94/NKG2, NKG2D, CD122, and DNAM-1) and granzyme B (12). With regards to TCR diversity, CD56+ T cells exhibit a considerably restricted TCRVβ spectrum compared to CD56 T cells, but a more diverse spectrum than invariant T cells. CD56+ T cells expand in response to IL-2 synergized with IL-12 (13). They also exhibit a potent capacity for T helper 1 cytokine production, and exert TCR-independent cytotoxicity following stimulation with mitogen and IL-2 (10). Based on these innate-like features, it has been suggested that CD56+ T cells may contribute to rapid immune responses against viruses, like innate immune cells. CD56+ T cells have been reported to inhibit hepatitis C virus replication in hepatocytes (14). The frequency of CD56+ T cells in peripheral blood is higher among patients with cytomegalovirus (CMV) infection compared to healthy donors (15).

Our research group recently identified a distinct CD8+ T-cell subpopulation showing high CD56 expression without CD161 expression (CD56hiCD161CD8+ T cells), which is characterized by high expression of NK-related molecules and a uniquely restricted TCR repertoire (Fig. 1). The frequency of these cells within the liver sinusoidal CD8+ T-cell population is significantly increased among patients with hepatitis B virus (HBV)-related chronic liver disease (16). CD56hiCD161CD8+ T cells mainly exhibit a CCR7CD45RA effector memory phenotype, and include a higher frequency of CD69+ cells, which are tissue resident memory T (TRM) or TRM-like cells, compared to other effector memory CD8+ T cells. CD56hiCD161CD8+ T cells exhibit weak responsiveness to TCR stimulation, but they show high expression of various NK receptors (e.g., CD94, KIRs, and NKG2C) and exert NKG2C- or NKG2D-mediated effector functions even in the absence of TCR stimulation. Additionally, CD56hiCD161CD8+ T cells are highly responsive to innate cytokines (e.g., IL-12/18 and IL-15) in the absence of TCR stimulation. The CD56hiCD161CD8+ T-cell population resembles previously described CD161CD56+ regulatory CD8+ T cells (17). Further studies are needed to elucidate the roles of CD56hiCD161CD8+ T cells in immune responses to microbial pathogens or immunopathology.

Figure 1
Innate-like features of the CD56hiCD161CD8+ T-cell population. Our group recently reported a CD8+ T-cell population marked with high CD56 expression without CD161 expression (CD56hiCD161CD8+ T cells). These CD56hiCD161CD8+ T cells are distinguished from other CD8+ T cells in terms of their innate-like features. This CD8+ subpopulation exhibits high expressions of various NK receptors, and exerts NK-receptor mediated effector functions in a TCR-independent manner. Additionally, these CD56hiCD161CD8+ T cells show increased responsiveness to stimulation with innate cytokines, including IL-12/18 and IL-15.

CD8+ T CELLS EXPRESSING KIRs

Virus-infected or transformed host cells tend to lose their MHC-I expression (termed “missing-self”). These aberrant cells with MHC-I downregulation are targeted by NK cells. When target cells express sufficient levels of MHC-I, inhibitory KIRs (receptors for MHC-I) deliver inhibitory signals to NK cells, which does not occur when target cells lose MHC-I expression (18). KIRs are polygenic and polymorphic Ig superfamily receptors, which can recognize distinct MHC-I molecules that are also polygenic and polymorphic. Inhibitory KIRs contain immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in their cytoplasmic domains, which suppress signaling delivered by activating receptors (19, 20).

KIRs can be expressed on TCRαβ+CD8+ T cells as well as NK cells (21), and can exert a suppressive function in both cell types. Engagement of KIRs with MHC-I ligands reduces the TCR-mediated phosphorylation of ZAP-70 and LAT and downstream signaling pathways (22). This decreases the TCR-triggered effector functions of CD8+ T cells, in terms of cytokine secretion (23, 24) and cytotoxicity (24, 25, 26). On the other hand, KIRs might contribute to CD8+ T-cell survival by suppressing activation-induced cell death (27, 28). Furthermore, several studies suggest that KIR+CD8+ T cells show intrinsic functional impairment, at least against TCR stimulation. Impairment of proliferation and cytokine secretion have been reported even in the absence of KIR engagement (29, 30, 31).

KIR+CD8+ T cells exhibit the surface phenotypes of CCR7CD45RA+ effector memory T (TEMRA) cells, known as terminally differentiated T cells (32, 33), and CD28CD57+ replicative-senescent T cells (30, 34). They also express high levels of cytotoxic molecules, such as perforin (29) and granzyme B (35). KIR+CD8+ T cells exhibit an increasing frequency with age (31, 36) and have a restricted TCR repertoire (26, 30, 37).

Infection with CMV, one of the most prevalent latent viruses in human beings, is associated with expansion of the KIR+CD8+ T-cell population. Chan et al. (12) demonstrated that CMV-seropositive individuals have higher frequencies of KIR+CD56+ T cells compared to CMV-seronegative individuals. Moreover, CMV reactivation is associated with expansion of the KIR+CD56+ T-cell population in bone marrow transplant recipients. It was hypothesized that the KIR+ T cells in CMV-seropositive individuals are specific to CMV Ags. However, Björkström et al. (30) demonstrated that KIR expression is much lower (or virtually absent) on CMV pp65-specific CD45RA+CD57+CD8+ T cells compared to on the non-specific population. On the other hand, a recent report described KIR expression on the vast majority of HLA-E-restricted CMV UL40-specific CD8+ T cells (38).

Besides in CMV infection, the KIR+CD8+ T-cell population expands in patients with HIV infection (39) and psoriasis (40). Additionally, the MHC allele-dependent expansion of KIR+CD8+ T-cell populations has been reported in cancer patients (41, 42). It remains unknown whether Ag recognition by TCR is required for expansion of the KIR+CD8+ T-cell population in patients with inflammation or infection.

Recent studies have reported that KIR+CD8+ T cells function in regulating immune responses (Fig. 2). In 2021, Pieren et al. (31) reported that KIR+CD45RA+CD8+ T cells are regulatory CD8+ T cells, as was previously described in mice (43). Similar to CD4+ Tregs, KIR+CD45RA+CD8+ T cells exhibit high expressions of Helios and TIGIT. These cells also show upregulation of CD122, which is associated with CD8+ Tregs in mice. Pieren et al. (31) also demonstrated that KIR+CD45RA+CD8+ T cells can dose-dependently regulate the proliferation of KIRNKG2A conventional CD8+ T cells. More recently, Li et al. (37) also reported that KIR+CD8+ cells act as CD8+ Tregs. They found increased frequencies of KIR+CD8+ T cells in patients with autoimmune diseases and infections, such as celiac disease and coronavirus disease 2019 (COVID-19). When gliadin-specific CD4+ T cells from patients with celiac disease were stimulated with Ag, KIR+CD8+ T cells suppressed pathogenic CD4+ T-cell responses by killing pathogenic cells, without harming non-pathogenic CD4+ T cells. RNA sequencing of KIR+CD8+ T cells further revealed that human KIR+CD8+ T cells are the analogous population of mouse Ly49+CD8+ T cells (CD8+ Tregs). Further studies are required to elucidate how the KIR+CD8+ T-cell population size is regulated, and how these cells recognize pathogenic T cells.

Figure 2
KIR+CD8+ T cells as a regulator of immune responses. KIR+CD8+ T cells have been reported to be human CD8+ TREG cells, which can regulate or kill pathogenic T cells. Similar to CD8+ TREG cells of mice, human KIR+CD8+ T cells exhibit high expressions of the transcription factor Helios and IL-15 receptor β chain (CD122). They contain high amounts of cytotoxic molecules (perforin and granzyme B), and show restricted TCR usage. The mechanisms of how KIR+CD8+ T cells originate, and how they recognize pathogenic T cells, remain unknown.

CD8+ T CELLS EXPRESSING NKG2A

A member of the lectin family, NKG2A is an ITIM-bearing inhibitory receptor (44). NKG2A forms a heterodimer with CD94, and binds to HLA-E, which is expressed on most human tissues and complexed with peptides derived from the leader sequence of classical MHC-I (45, 46, 47). NKG2A is typically expressed on NK cells, but can also be expressed on CD8+ T cells (48, 49, 50). TCR stimulation induces NKG2A expression on CD8+ T cells in a manner synergized with cytokines, such as IL-12, IL-15, and TGF (51, 52, 53, 54, 55). NKG2A engagement suppresses effector functions of CD8+ T cells (49), as also observed in NK cells, and blockade of NKG2A or CD94 increases the cytotoxic activity of NKG2A-expressing CD8+ T cells (53).

NKG2A has recently attracted attention as an immune checkpoint, similar to the well-known checkpoint receptors PD-1, TIGIT, and TIM-3 (56, 57). An anti-NKG2A monoclonal Ab, called monalizumab, has been developed (58, 59). Clinical trials investigating concomitant use of anti-PD-1/PD-L1 treatment and monalizumab have shown better clinical results in diverse cancers, including bladder cancer, non-small-cell lung cancer, and colorectal cancer (56, 60, 61). Chronic antigenic stimulation and persistent exposure to various cytokines in the tumor microenvironment reportedly induce NKG2A expression on tumor-infiltrating CD8+ T cells (51). Monalizumab reinvigorates the cytotoxic function of NKG2A+CD8+ T cells by blocking the interaction of NKG2A with HLA-E expressed on cancer cells. It has also been found that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specific CD8+ T cells from patients with severe COVID-19 express high NKG2A levels, and exert reduced effector functions (62).

NKG2A and/or KIRs have been proposed as unique markers of human virtual memory T (TVM) cells, which are T cells featuring memory phenotypes even in neonatal cord blood (63). Human TVM cells express high levels of eomesodermin, CD62L, and CD122; exhibit a CCR7CD45RA+ TEMRA phenotype; and show increased responsiveness to innate cytokines, such as IL-12, IL-15, and IL-18 (63, 64). However, a recent study demonstrated that CD45RA+CD8+ T cells expressing NKG2A versus KIR are distinct subsets (31). Compared to KIR+CD45RA+CD8+ T cells, NKG2A+CD45RA+CD8+ T cells exhibit downregulated transcripts related to senescence, exhaustion, and regulatory functions. Moreover, the relative proportion of NKG2A+CD45RA+CD8+ T cells declines with age.

CD8+ T CELLS EXPRESSING NKG2C

NKG2C also forms a heterodimer with CD94. The NKG2C/CD94 heterodimer binds to HLA-E, similar to NKG2A but with lower affinity, and transduces signals through the immunoreceptor tyrosine activation motif (ITAM)-bearing adaptor molecule DAP12 (65, 66). Like NKG2A, NKG2C is mainly expressed on NK cells, but can also be expressed on some subsets of CD8+ T cells (50, 67). NKG2C+CD8+ T-cell populations have been reported to expand under several pathologic conditions, including CMV infection, Stevens-Johnson syndrome, toxic epidermal necrolysis, and celiac disease (68, 69, 70). Two studies have described NKG2C-mediated CD8+ T-cell activation. Co-stimulation with anti-CD94 and anti-CD3 Abs strengthens the lytic function of NKG2C-expressing CD8+ T cells (50). Even in the absence of TCR stimulation, NKG2C ligation itself can activate T cells to proliferate and kill HLA-E-transfected target cells that do not express the other MHC-I molecules (71). This finding reveals that NKG2C signaling could be a potential alternative to TCR-mediated activation of CD8+ T cells.

Recent studies have also examined and characterized NKG2C-expressing CD8+ T cells (Fig. 3). One study identified NKG2C as an important marker for potent antimicrobial T cells against Mycobacterium leprae (72). Compared with other CD8+ subsets, CD8+ T cells that express granulysin, perforin, and granzyme B exert superior effector functions against M. leprae-infected macrophages, and these cells typically express NKG2C. Functionally, anti-CD94 Ab enhances the release of cytotoxic molecules from anti-CD3-stimulated NKG2C+CD8+ T cells.

Figure 3
Characteristics of CD8+ T cells expressing NKG2C. Balin et al. (72) reported that NKG2C is an important marker for CD8+ T cells expressing granulysin, perforin, and granzyme B (tri-cytotoxic CD8+ T cells), and having greater antimicrobial activity against Mycobacterium leprae. NKG2C ligation activates tri-cytotoxic CD8+ T cells to release cytotoxic proteins. Sottile et al. (67) described downregulation of BCL11B in NKG2C+CD8+ T cells. The loss of BCL11B triggers NK-cell-like reprogramming and induces the generation of NKG2C+CD8+ T cells. Sullivan et al. (38) reported that HLA-E-restricted CD8+ T cells with a TRBV14 repertoire have a low affinity for HLA-E/UL 40 complexes, and express high levels of NKG2C. NKG2C ligation increases the production of IFN-γ and TNF-α from these CD8+ T cells, indicating that NKG2C can compensate for a weak TCR signal.

HLA-E serves as a ligand of both CD94/NKG2C and CD94/NKG2A, complexed with peptides derived from the leader sequence of classical MHC-I (73). In CMV infection, CMV-encoded UL40, which mimics the leader sequence of classical MHC-I, enables CMV-infected cells to evade NK cell-mediated immune responses by engaging NKG2A (74). On the other hand, NKG2C on NK cells can recognize the HLA-E/UL40 complex, and NKG2C+ NK cells exert cytotoxic functions against CMV-infected cells (75). Furthermore, these NKG2C+ NK cells undergo clonal-like expansion against UL40, similar to the memory response of CD8+ T cells. The NKG2C+CD8+ T-cell population also expands in CMV-infected patients (76). Sottile et al. revealed a mechanism for CD8+ T-cell expansion and reprogramming in CMV-infected patients (67). Bulk RNA-sequencing analysis of the NKG2C+CD8+ T-cell population revealed downregulation of BCL11B. The loss of BCL11B triggers NK-cell-like reprogramming of T cells, and induces the generation of NKG2C+CD8+ T cells under HLA-E ligand stimulation. Additionally, TCR analysis reveals that most NKG2C+CD8+ T cells in CMV-seropositive individuals exhibit narrow TCR Vβ-chain usage, mainly TRBV-14, while NKG2C+CD8+ T cells in CMV-seronegative individuals are more polyclonal. This restricted TCR diversity in CMV-seropositive donors indicates that NKG2C+CD8+ T cells undergo clonal expansion.

HLA-E is recognized not only by NKG2A and NKG2C, but also by TCRs of CD8+ T cells that are restricted by HLA-E. HLA-E-restricted CD8+ T cells have been investigated in several diseases, including CMV, HIV, Epstein-Barr virus, Mycobacterium tuberculosis, and Salmonella typhi infection (74, 77, 78). Interestingly, HLA-E-restricted CD8+ T cells reportedly exert regulatory properties in tuberculosis infection by inhibiting the proliferation of CD4+ T cells, and patients with autoimmune type I diabetes exhibit a decreased frequency of HLA-E-restricted CD8+ T cells (79, 80). Sullivan et al. (38) examined whether NKG2C played a role in the activation of HLA-E-restricted CD8+ T cells in CMV infection, and demonstrated that NKG2C on HLA-E-restricted CD8+ T cells could co-stimulate CD8+ T cells by compensating for the relatively weak signal intensity of TCRs.

CONCLUSION

In the late 1990s, researchers reported CD56+ T cells (termed “natural T cells”) and demonstrated that this T-cell subpopulation exhibits distinct immunologic features, in terms of the expressions of many NK receptors and innate-like features, compared to their CD56 T-cell counterparts. Since those initial descriptions, there have been sporadic reports of CD8+ T-cell populations expressing various NK receptors (Table 1). Some act as co-stimulatory or inhibitory molecules, while others stimulate CD8+ T cells to exert effector functions in a TCR-independent manner. Recent breakthrough technical developments in multi-omics analysis have enabled us to explore the heterogenous subpopulations of CD8+ T cells expressing NK receptors, and to reveal the unique immunologic characteristics of these populations (16, 37). The CD8+ T-cell populations expressing NK receptors execute unique functional roles—for example, regulatory roles—distinct from the conventional CD8+ T-cell population. However, studies have not yet fully clarified the heterogeneity of CD8+ T-cell populations expressing NK receptors. Further studies are needed to delineate the heterogeneity of CD8+ T-cell populations expressing NK receptors, and to elucidate their molecular characteristics and roles in physiologic and pathologic conditions.

Table 1
Featured characteristics of human CD8+ T cells expressing NK-associated surface proteins

Notes

Conflict of Interest:The authors declare no potential conflicts of interest.

Author Contributions:

  • Conceptualization: Koh JY, Kim DU, Moon BH, Shin EC.

  • Data curation: Koh JY, Kim DU, Moon BH.

  • Formal analysis: Kim DU, Moon BH.

  • Funding acquisition: Shin EC.

  • Investigation: Koh JY, Kim DU, Moon BH.

  • Methodology: Koh JY, Kim DU, Moon BH.

  • Project administration: Koh JY, Shin EC.

  • Resources: Koh JY.

  • Supervision: Shin EC.

  • Validation: Shin EC.

  • Writing - original draft: Koh JY, Kim DU, Moon BH, Shin EC.

Abbreviations

COVID-19 coronavirus disease 2019
CMV cytomegalovirus
ITAM immunoreceptor tyrosine activation motif
ITIM immunoreceptor tyrosine-based inhibitory motif
KIRs killer cell immunoglobulin-like receptors
MHC-I MHC class I
NCAM neural cell adhesion molecule
SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
TEMRA effector memory T
TRM resident memory T
TVM virtual memory T

ACKNOWLEDGEMENTS

This work was supported by the Institute for Basic Science (IBS), Korea, under project code IBS-R801-D2.

References

    1. Lanier LL. NK cell recognition. Annu Rev Immunol 2005;23:225–274.
    1. Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol 2015;33:257–290.
    1. Rha MS, Kim AR, Shin EC. SARS-CoV-2-specific T cell responses in patients with COVID-19 and unexposed individuals. Immune Netw 2021;21:e2
    1. Thiery JP, Brackenbury R, Rutishauser U, Edelman GM. Adhesion among neural cells of the chick embryo. II. Purification and characterization of a cell adhesion molecule from neural retina. J Biol Chem 1977;252:6841–6845.
    1. Nitta T, Yagita H, Sato K, Okumura K. Involvement of CD56 (NKH-1/Leu-19 antigen) as an adhesion molecule in natural killer-target cell interaction. J Exp Med 1989;170:1757–1761.
    1. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, Carson WE, Caligiuri MA. Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood 2001;97:3146–3151.
    1. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol 2001;22:633–640.
    1. Parrish-Novak J, Dillon SR, Nelson A, Hammond A, Sprecher C, Gross JA, Johnston J, Madden K, Xu W, West J, et al. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature 2000;408:57–63.
    1. Deignan T, Curry MP, Doherty DG, Golden-Mason L, Volkov Y, Norris S, Nolan N, Traynor O, McEntee G, Hegarty JE, et al. Decrease in hepatic CD56+ T cells and Vα24+ natural killer T cells in chronic hepatitis C viral infection. J Hepatol 2002;37:101–108.
    1. Doherty DG, Norris S, Madrigal-Estebas L, McEntee G, Traynor O, Hegarty JE, O’Farrelly C. The human liver contains multiple populations of NK cells, T cells, and CD3+CD56+ natural T cells with distinct cytotoxic activities and Th1, Th2, and Th0 cytokine secretion patterns. J Immunol 1999;163:2314–2321.
    1. Norris S, Doherty DG, Collins C, McEntee G, Traynor O, Hegarty JE, O’Farrelly C. Natural T cells in the human liver: cytotoxic lymphocytes with dual T cell and natural killer cell phenotype and function are phenotypically heterogenous and include Vα24-JαQ and γδ T cell receptor bearing cells. Hum Immunol 1999;60:20–31.
    1. Chan WK, Rujkijyanont P, Neale G, Yang J, Bari R, Das Gupta N, Holladay M, Rooney B, Leung W. Multiplex and genome-wide analyses reveal distinctive properties of KIR+ and CD56+ T cells in human blood. J Immunol 2013;191:1625–1636.
    1. Satoh M, Seki S, Hashimoto W, Ogasawara K, Kobayashi T, Kumagai K, Matsuno S, Takeda K. Cytotoxic γδ or αβ T cells with a natural killer cell marker, CD56, induced from human peripheral blood lymphocytes by a combination of IL-12 and IL-2. J Immunol 1996;157:3886–3892.
    1. Ye L, Wang X, Wang S, Wang Y, Song L, Hou W, Zhou L, Li H, Ho W. CD56+ T cells inhibit hepatitis C virus replication in human hepatocytes. Hepatology 2009;49:753–762.
    1. Almehmadi M, Flanagan BF, Khan N, Alomar S, Christmas SE. Increased numbers and functional activity of CD56+ T cells in healthy cytomegalovirus positive subjects. Immunology 2014;142:258–268.
    1. Koh JY, Rha MS, Choi SJ, Lee HS, Han JW, Nam H, Kim DU, Lee JG, Kim MS, Park JY, et al. Identification of a distinct NK-like hepatic T-cell population activated by NKG2C in a TCR-independent manner. J Hepatol 2022;77:1059–1070.
    1. Hu D, Weiner HL, Ritz J. Identification of cytolytic CD161 CD56+ regulatory CD8 T cells in human peripheral blood. PLoS One 2013;8:e59545
    1. Moretta A, Bottino C, Vitale M, Pende D, Biassoni R, Mingari MC, Moretta L. Receptors for HLA class-I molecules in human natural killer cells. Annu Rev Immunol 1996;14:619–648.
    1. Campbell KS, Purdy AK. Structure/function of human killer cell immunoglobulin-like receptors: lessons from polymorphisms, evolution, crystal structures and mutations. Immunology 2011;132:315–325.
    1. Pende D, Falco M, Vitale M, Cantoni C, Vitale C, Munari E, Bertaina A, Moretta F, Del Zotto G, Pietra G, et al. Killer Ig-like receptors (KIRs): Their role in NK cell modulation and developments leading to their clinical exploitation. Front Immunol 2019;10:1179.
    1. Ferrini S, Cambiaggi A, Meazza R, Sforzini S, Marciano S, Mingari MC, Moretta L. T cell clones expressing the natural killer cell-related p58 receptor molecule display heterogeneity in phenotypic properties and p58 function. Eur J Immunol 1994;24:2294–2298.
    1. Guerra N, Michel F, Gati A, Gaudin C, Mishal Z, Escudier B, Acuto O, Chouaib S, Caignard A. Engagement of the inhibitory receptor CD158a interrupts TCR signaling, preventing dynamic membrane reorganization in CTL/tumor cell interaction. Blood 2002;100:2874–2881.
    1. Bonorino P, Leroy V, Dufeu-Duchesne T, Tongiani-Dashan S, Sturm N, Pernollet M, Vivier E, Zarski JP, Marche PN, Jouvin-Marche E. Features and distribution of CD8 T cells with human leukocyte antigen class I-specific receptor expression in chronic hepatitis C. Hepatology 2007;46:1375–1386.
    1. van der Veken LT, Diez Campelo M, van der Hoorn MA, Hagedoorn RS, van Egmond HM, van Bergen J, Willemze R, Falkenburg JH, Heemskerk MH. Functional analysis of killer Ig-like receptor-expressing cytomegalovirus-specific CD8+ T cells. J Immunol 2009;182:92–101.
    1. Ikeda H, Lethé B, Lehmann F, van Baren N, Baurain JF, de Smet C, Chambost H, Vitale M, Moretta A, Boon T, et al. Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor. Immunity 1997;6:199–208.
    1. Guerra N, Guillard M, Angevin E, Echchakir H, Escudier B, Moretta A, Chouaib S, Caignard A. Killer inhibitory receptor (CD158b) modulates the lytic activity of tumor-specific T lymphocytes infiltrating renal cell carcinomas. Blood 2000;95:2883–2889.
    1. Ugolini S, Arpin C, Anfossi N, Walzer T, Cambiaggi A, Förster R, Lipp M, Toes RE, Melief CJ, Marvel J, et al. Involvement of inhibitory NKRs in the survival of a subset of memory-phenotype CD8+ T cells. Nat Immunol 2001;2:430–435.
    1. Gati A, Guerra N, Gaudin C, Da Rocha S, Escudier B, Lécluse Y, Bettaieb A, Chouaib S, Caignard A. CD158 receptor controls cytotoxic T-lymphocyte susceptibility to tumor-mediated activation-induced cell death by interfering with Fas signaling. Cancer Res 2003;63:7475–7482.
    1. Anfossi N, Doisne JM, Peyrat MA, Ugolini S, Bonnaud O, Bossy D, Pitard V, Merville P, Moreau JF, Delfraissy JF, et al. Coordinated expression of Ig-like inhibitory MHC class I receptors and acquisition of cytotoxic function in human CD8+ T cells. J Immunol 2004;173:7223–7229.
    1. Björkström NK, Béziat V, Cichocki F, Liu LL, Levine J, Larsson S, Koup RA, Anderson SK, Ljunggren HG, Malmberg KJ. CD8 T cells express randomly selected KIRs with distinct specificities compared with NK cells. Blood 2012;120:3455–3465.
    1. Pieren DK, Smits NA, Hoeboer J, Kandiah V, Postel RJ, Mariman R, van Beek J, van Baarle D, de Wit J, Guichelaar T. Regulatory KIR+ RA+ T cells accumulate with age and are highly activated during viral respiratory disease. Aging Cell 2021;20:e13372
    1. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401:708–712.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol 2004;22:745–763.
    1. Brenchley JM, Karandikar NJ, Betts MR, Ambrozak DR, Hill BJ, Crotty LE, Casazza JP, Kuruppu J, Migueles SA, Connors M, et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood 2003;101:2711–2720.
    1. Arlettaz L, Degermann S, De Rham C, Roosnek E, Huard B. Expression of inhibitory KIR is confined to CD8+ effector T cells and limits their proliferative capacity. Eur J Immunol 2004;34:3413–3422.
    1. Anfossi N, Pascal V, Vivier E, Ugolini S. Biology of T memory type 1 cells. Immunol Rev 2001;181:269–278.
    1. Li J, Zaslavsky M, Su Y, Guo J, Sikora MJ, van Unen V, Christophersen A, Chiou SH, Chen L, Li J, et al. KIR+CD8+ T cells suppress pathogenic T cells and are active in autoimmune diseases and COVID-19. Science 2022;376:eabi9591
    1. Sullivan LC, Nguyen THO, Harpur CM, Stankovic S, Kanagarajah AR, Koutsakos M, Saunders PM, Cai Z, Gray JA, Widjaja JML, et al. Natural killer cell receptors regulate responses of HLA-E-restricted T cells. Sci Immunol 2021;6:eabe9057.
    1. Alter G, Rihn S, Streeck H, Teigen N, Piechocka-Trocha A, Moss K, Cohen K, Meier A, Pereyra F, Walker B, et al. Ligand-independent exhaustion of killer immunoglobulin-like receptor-positive CD8+ T cells in human immunodeficiency virus type 1 infection. J Virol 2008;82:9668–9677.
    1. Liao YH, Jee SH, Sheu BC, Huang YL, Tseng MP, Hsu SM, Tsai TF. Increased expression of the natural killer cell inhibitory receptor CD94/NKG2A and CD158b on circulating and lesional T cells in patients with chronic plaque psoriasis. Br J Dermatol 2006;155:318–324.
    1. Campillo JA, Martínez-Escribano JA, Moya-Quiles MR, Marín LA, Muro M, Guerra N, Parrado A, Campos M, Frías JF, Minguela A, et al. Natural killer receptors on CD8 T cells and natural killer cells from different HLA-C phenotypes in melanoma patients. Clin Cancer Res 2006;12:4822–4831.
    1. Gimeno L, Serrano-López EM, Campillo JA, Cánovas-Zapata MA, Acuña OS, García-Cózar F, Martínez-Sánchez MV, Martínez-Hernández MD, Soto-Ramírez MF, López-Cubillana P, et al. KIR+ CD8+ T lymphocytes in cancer immunosurveillance and patient survival: gene expression profiling. Cancers (Basel) 2020;12:2991.
    1. Mishra S, Srinivasan S, Ma C, Zhang N. CD8+ regulatory T cell - a mystery to be revealed. Front Immunol 2021;12:708874
    1. Vivier E, Daëron M. Immunoreceptor tyrosine-based inhibition motifs. Immunol Today 1997;18:286–291.
    1. Brooks AG, Posch PE, Scorzelli CJ, Borrego F, Coligan JE. NKG2A complexed with CD94 defines a novel inhibitory natural killer cell receptor. J Exp Med 1997;185:795–800.
    1. Braud VM, Allan DS, O’Callaghan CA, Söderström K, D’Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 1998;391:795–799.
    1. Houchins JP, Lanier LL, Niemi EC, Phillips JH, Ryan JC. Natural killer cell cytolytic activity is inhibited by NKG2-A and activated by NKG2-C. J Immunol 1997;158:3603–3609.
    1. Mahapatra S, Mace EM, Minard CG, Forbes LR, Vargas-Hernandez A, Duryea TK, Makedonas G, Banerjee PP, Shearer WT, Orange JS. High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults. PLoS One 2017;12:e0181134
    1. Bertone S, Schiavetti F, Bellomo R, Vitale C, Ponte M, Moretta L, Mingari MC. Transforming growth factor-β-induced expression of CD94/NKG2A inhibitory receptors in human T lymphocytes. Eur J Immunol 1999;29:23–29.
    1. Arlettaz L, Villard J, de Rham C, Degermann S, Chapuis B, Huard B, Roosnek E. Activating CD94:NKG2C and inhibitory CD94:NKG2A receptors are expressed by distinct subsets of committed CD8+ TCR αβ lymphocytes. Eur J Immunol 2004;34:3456–3464.
    1. Borst L, van der Burg SH, van Hall T. The NKG2A-HLA-E axis as a novel checkpoint in the tumor microenvironment. Clin Cancer Res 2020;26:5549–5556.
    1. Gunturi A, Berg RE, Crossley E, Murray S, Forman J. The role of TCR stimulation and TGF-β in controlling the expression of CD94/NKG2A receptors on CD8 T cells. Eur J Immunol 2005;35:766–775.
    1. Sheu BC, Chiou SH, Lin HH, Chow SN, Huang SC, Ho HN, Hsu SM. Up-regulation of inhibitory natural killer receptors CD94/NKG2A with suppressed intracellular perforin expression of tumor-infiltrating CD8+ T lymphocytes in human cervical carcinoma. Cancer Res 2005;65:2921–2929.
    1. Jabri B, Selby JM, Negulescu H, Lee L, Roberts AI, Beavis A, Lopez-Botet M, Ebert EC, Winchester RJ. TCR specificity dictates CD94/NKG2A expression by human CTL. Immunity 2002;17:487–499.
    1. Derre L, Corvaisier M, Pandolfino MC, Diez E, Jotereau F, Gervois N. Expression of CD94/NKG2-A on human T lymphocytes is induced by IL-12: implications for adoptive immunotherapy. J Immunol 2002;168:4864–4870.
    1. Salomé B, Sfakianos JP, Ranti D, Daza J, Bieber C, Charap A, Hammer C, Banchereau R, Farkas AM, Ruan DF, et al. NKG2A and HLA-E define an alternative immune checkpoint axis in bladder cancer. Cancer Cell 2022;40:1027–1043.e9.
    1. Creelan BC, Antonia SJ. The NKG2A immune checkpoint - a new direction in cancer immunotherapy. Nat Rev Clin Oncol 2019;16:277–278.
    1. André P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, Bléry M, Bonnafous C, Gauthier L, Morel A, et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 2018;175:1731–1743.e13.
    1. van Montfoort N, Borst L, Korrer MJ, Sluijter M, Marijt KA, Santegoets SJ, van Ham VJ, Ehsan I, Charoentong P, André P, et al. NKG2A blockade potentiates CD8 T cell immunity induced by cancer vaccines. Cell 2018;175:1744–1755.e15.
    1. Herbst RS, Majem M, Barlesi F, Carcereny E, Chu Q, Monnet I, Sanchez-Hernandez A, Dakhil S, Camidge DR, Winzer L, et al. Coast: An open-label, phase II, multidrug platform study of durvalumab alone or in combination with oleclumab or monalizumab in patients with unresectable, stage III non-small-cell lung cancer. J Clin Oncol 2022;40:3383–3393.
    1. Ducoin K, Oger R, Bilonda Mutala L, Deleine C, Jouand N, Desfrançois J, Podevin J, Duchalais E, Cruard J, Benlalam H, et al. Targeting NKG2A to boost anti-tumor CD8 T-cell responses in human colorectal cancer. OncoImmunology 2022;11:2046931
    1. Gangaev A, Ketelaars SL, Isaeva OI, Patiwael S, Dopler A, Hoefakker K, De Biasi S, Gibellini L, Mussini C, Guaraldi G, et al. Identification and characterization of a SARS-CoV-2 specific CD8+ T cell response with immunodominant features. Nat Commun 2021;12:2593.
    1. Jacomet F, Cayssials E, Basbous S, Levescot A, Piccirilli N, Desmier D, Robin A, Barra A, Giraud C, Guilhot F, et al. Evidence for eomesodermin-expressing innate-like CD8+ KIR/NKG2A+ T cells in human adults and cord blood samples. Eur J Immunol 2015;45:1926–1933.
    1. White JT, Cross EW, Burchill MA, Danhorn T, McCarter MD, Rosen HR, O’Connor B, Kedl RM. Virtual memory T cells develop and mediate bystander protective immunity in an IL-15-dependent manner. Nat Commun 2016;7:11291.
    1. Valés-Gómez M, Reyburn HT, Erskine RA, López-Botet M, Strominger JL. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J 1999;18:4250–4260.
    1. Lanier LL, Corliss B, Wu J, Phillips JH. Association of DAP12 with activating CD94/NKG2C NK cell receptors. Immunity 1998;8:693–701.
    1. Sottile R, Panjwani MK, Lau CM, Daniyan AF, Tanaka K, Barker JN, Brentjens RJ, Sun JC, Le Luduec JB, Hsu KC. Human cytomegalovirus expands a CD8+ T cell population with loss of BCL11B expression and gain of NK cell identity. Sci Immunol 2021;6:eabe6968
    1. Ishiyama K, Arakawa-Hoyt J, Aguilar OA, Damm I, Towfighi P, Sigdel T, Tamaki S, Babdor J, Spitzer MH, Reed EF, et al. Mass cytometry reveals single-cell kinetics of cytotoxic lymphocyte evolution in CMV-infected renal transplant patients. Proc Natl Acad Sci U S A 2022;119:e2116588119
    1. Morel E, Escamochero S, Cabañas R, Díaz R, Fiandor A, Bellón T. CD94/NKG2C is a killer effector molecule in patients with Stevens-Johnson syndrome and toxic epidermal necrolysis. J Allergy Clin Immunol 2010;125:703–710. 710.e1–710.e8.
    1. Meresse B, Curran SA, Ciszewski C, Orbelyan G, Setty M, Bhagat G, Lee L, Tretiakova M, Semrad C, Kistner E, et al. Reprogramming of CTLs into natural killer-like cells in celiac disease. J Exp Med 2006;203:1343–1355.
    1. Gumá M, Busch LK, Salazar-Fontana LI, Bellosillo B, Morte C, García P, López-Botet M. The CD94/NKG2C killer lectin-like receptor constitutes an alternative activation pathway for a subset of CD8+ T cells. Eur J Immunol 2005;35:2071–2080.
    1. Balin SJ, Pellegrini M, Klechevsky E, Won ST, Weiss DI, Choi AW, Hakimian J, Lu J, Ochoa MT, Bloom BR, et al. Human antimicrobial cytotoxic T lymphocytes, defined by NK receptors and antimicrobial proteins, kill intracellular bacteria. Sci Immunol 2018;3:eaat7668
    1. Grant EJ, Nguyen AT, Lobos CA, Szeto C, Chatzileontiadou DS, Gras S. The unconventional role of HLA-E: the road less traveled. Mol Immunol 2020;120:101–112.
    1. Joosten SA, Sullivan LC, Ottenhoff TH. Characteristics of HLA-E restricted T-cell responses and their role in infectious diseases. J Immunol Res 2016;2016:2695396
    1. Hammer Q, Rückert T, Borst EM, Dunst J, Haubner A, Durek P, Heinrich F, Gasparoni G, Babic M, Tomic A, et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat Immunol 2018;19:453–463.
    1. Kovalenko EI, Zvyagin IV, Streltsova MA, Mikelov AI, Erokhina SA, Telford WG, Sapozhnikov AM, Lebedev YB. Surface NKH2C identifies differentiated αβT-cell clones expanded in peripheral blood. Front Immunol 2021;11:613882
    1. Jouand N, Bressollette-Bodin C, Gérard N, Giral M, Guérif P, Rodallec A, Oger R, Parrot T, Allard M, Cesbron-Gautier A, et al. HCMV triggers frequent and persistent UL40-specific unconventional HLA-E-restricted CD8 T-cell responses with potential autologous and allogeneic peptide recognition. PLoS Pathog 2018;14:e1007041
    1. Voogd L, Ruibal P, Ottenhoff TH, Joosten SA. Antigen presentation by MHC-E: a putative target for vaccination? Trends Immunol 2022;43:355–365.
    1. Joosten SA, van Meijgaarden KE, van Weeren PC, Kazi F, Geluk A, Savage ND, Drijfhout JW, Flower DR, Hanekom WA, Klein MR, et al. Mycobacterium tuberculosis peptides presented by HLA-E molecules are targets for human CD8 T-cells with cytotoxic as well as regulatory activity. PLoS Pathog 2010;6:e1000782
    1. Jiang H, Canfield SM, Gallagher MP, Jiang HH, Jiang Y, Zheng Z, Chess L. HLA-E-restricted regulatory CD8+ T cells are involved in development and control of human autoimmune type 1 diabetes. J Clin Invest 2010;120:3641–3650.

Metrics
Share
Figures

1 / 3

Tables

1 / 1

PERMALINK