Abstract
Natural killer (NK) cells are innate lymphocytes with important roles in immunoregulation, immunosurveillance, and cytokine production. Originally defined on the functional basis of their ‘natural’ ability to lyse tumor targets and thought to be a relatively homogeneous group of lymphocytes, NK cells possess a remarkable degree of phenotypic and functional diversity due to the combinatorial expression of an array of activating and inhibitory receptors. Diversification of NK cells is multifaceted: mechanisms of NK cell education that promote self-tolerance result in a heterogeneous repertoire that further diversifies upon encounters with viral pathogens. Here, we review the genetic, developmental, and environmental sources of NK cell diversity with a particular focus on deep profiling and single-cell technologies that will enable a more thorough and accurate dissection of this intricate and poorly understood lymphocyte lineage.
Keywords: Natural killer cell or NK cell, lymphocyte diversity, innate lymphoid cell, host-pathogen interaction, mass cytometry, single-cell technology
Summary:
The contexts that influence human NK repertoire diversification and their implications for human health and disease are reviewed.
Introduction
Natural killer (NK) cells are a diverse group of innate lymphoid cells (ILCs) that can coordinate and execute the rapid elimination of neoplastic and virus-infected cells [1,2]. ILCs represent a group of largely tissue-resident common lymphoid progenitor-derived cells that do not express somatically-recombined antigen-specific receptors [3–7]. ILCs are now recognized to play significant roles in immune homeostasis, and their characteristics and functions have been thoroughly reviewed elsewhere [4,5,7]. Here, we focus on “conventional” human NK cells, a subset of type I ILCs which have been primarily evaluated in the blood.
NK cells were first identified in both mice and humans in the 1970s on the basis of their ability to kill tumor cells without the requirement of prior priming [8,9]. Primary functions of NK cells include production of IFN-γ and other cytokines, immunoregulation through the perforin-dependent elimination of activated leukocytes, and immunosurveillance through cytotoxicity against tumors or virus-infected cells [2,10,11]. These functions have intriguing clinical implications. For example, NK cells may have protective roles in several autoimmune diseases by eliminating activated autoreactive lymphocytes [12–15]. Further, higher NK cell cytotoxicity is protective against the development of cancer [16]. This latter finding, considered along with the ability of NK cells to eliminate their targets in rapid succession, has led to much interest in the development of NK cell-based cancer therapies, including adoptive NK cell therapy and pharmaceuticals that modulate the activity of NK receptors (NKRs) [17,18]. That NK cells play an important role in the control of early viral infection, particularly by herpesviruses, is evident from severe herpesvirus infections in exceedingly rare cases of primary NK cell deficiency [19], as well as evidence of NK cell dysfunction in immunodeficiencies like X-linked lymphoproliferative disorder [20–22]. Evidence of strong antiviral responses by NK cells, combined with recent findings of memory-like responses by NK cells, has bolstered the argument that future vaccine development should seek to provide long-lasting NK cell immunity [1,23].
In a sense, NK cells can be considered immune intermediaries, with both lifetimes and receptor-to-pathogen ratios between their adaptive lymphocyte and common myeloid progenitor-derived counterparts [24]. NK cells combine diversity, on the order of 104 subpopulations per person, with repertoire flexibility, being capable of altering receptor expression on the timescale of cellular processes [25]. Neither the clinical consequences of this diversity nor the functional characteristics of individual NK cell subsets have been fully elucidated. A better understanding of how this diversity influences NK cell responses and how it can be modulated is necessary for the development of therapeutic strategies employing NK cells. Here, we review the various contexts of NK cell diversification, as well as recent findings and future directions in the deep profiling strategies needed to dissect the functions of this complex group of ILCs.
Single-cell diversity of NK cells
Diversity is an essential characteristic of the immune system, as it must be prepared to respond to innumerable unknown pathogens. Leukocyte diversity is most often attributed to adaptive lymphocytes, which express somatically rearranged antigen-specific receptors, generating on the order of 106-108 distinct specificities per person [26]. NK cell diversity, on the other hand, is determined by the combinatorial expression of an array of germline-encoded inhibitory and activating receptors. While self-tolerance of individual T and B cells is ensured through selection on a single cell surface receptor, the differential expression of NKRs necessitates a complex developmental process to ensure a tunable and self-tolerant NK cell repertoire. These processes are referred to as NK cell education, arming, or licensing and are reviewed extensively elsewhere [27–31].
On a population level, NK cell diversity at the protein expression level arises in three main fashions: genetic diversity of killer immunoglobulin-like receptors (KIRs), stochastic expression of KIRs, and differential expression of other NKRs [32]. Early studies of NK cell diversity relied on multi-color flow cytometry to profile NK cell KIR repertoires [33–40], and a more comprehensive view has emerged in a recent study that employed cytometry by time-of-flight (CyTOF) [41]. CyTOF is a flow cytometry-based platform where, instead of labeling cells with fluorophore-conjugated antibodies, cells are labeled with heavy metal isotope-conjugated antibodies. This allows for the simultaneous assessment of ~40 parameters with a mass cytometry readout without the need for compensation of spectral overlap [42,43]. While this technology vastly improves the number of markers evaluated, downsides include the inefficient capture of events, such that only ~20% of live cells are captured as singlet events, and its slow speed [42–44]. Currently, inroads are being made to add a similar number of parameters to conventional flow cytometry platforms, but any high dimensional analysis involves analytic complexity, often involving multiple parallel analytic techniques to feel confident in results obtained [44].
Recently, our group used CyTOF to evaluate 28 NKRs by CyTOF, identifying between 6,000 and 30,000 distinct NK cell subsets per donor and >100,000 distinct subsets in the 22 donors studied based on a Boolean analysis of subsets based on yes/no expression patterns of the different receptors [41]. Additionally, our study utilized twin donors to demonstrate that inhibitory NKR diversity was more genetically controlled than activating NKR repertoires, which were under more environmental influence [41]. This diversity indicates that some subsets could be better tuned to detect certain types of infections or malignancies, based on NKR expression levels relative to ligand expression patterns during infection or malignancy. While this raises interesting possibilities, there are several important caveats when interpreting these data. There are more NKRs that can be added to this marker panel, and increases in the number of markers evaluated could increase the number of distinct NK cell subsets detected. Further, while this study reveals a high degree of phenotypic diversity among human NK cells, this may not reflect the same degree of functional diversity. It will be important for future studies to evaluate functional markers as well to assess the relationship between phenotypic and functional diversity. Lastly, by interrogating proteomic diversity, this study only analyzes one metric of NK cell diversity. Other methods, including scRNA-seq and scATAC-seq, should be utilized to gain a more comprehensive understanding of NK cell diversity. This study, nonetheless, presents significant insight into the magnitude of NK cell repertoire diversity and provides a methodological foundation for future deep functional profiling of individual NK cell subsets.
Mechanisms of NK cell diversification: genetic and environmental determinants
Development and functional specialization of NK cell subsets
NK cells develop in the bone marrow and secondary lymphoid tissues in discrete stages that have been well characterized [45–51]. After several precursor stages, immature NK cells gain expression of CD56, CD94/NKG2A, and several activating receptors including NKp46, NKp30, and NKG2D. These cells, called CD56bright NK cells, are highly proliferative, capable of producing high amounts of IFN-γ, and express IL-7Rɑ and KIT, two surface markers associated with non-NK ILCs [3,52,53]. In humans, CD56bright NK cells are hypothesized to give rise to CD56dim NK cells during the process of education; this involves a decrease in CD56 expression coupled with increased expression of CD16 and acquisition of KIRs [45,48,54]. CD56dim NK cells are potently cytotoxic, but have substantially lower proliferative and cytokine-producing capacity than their CD56bright counterparts. Further maturation of CD56dim NK cells is generally indicated by the loss of CD62L and acquisition of CD57.
That CD56bright NK cells are precursors to CD56dim NK cells in humans remains unclear. This hypothesis, originally advanced in 1986 [55], is supported both by the observations that CD56bright NK cells have longer telomeres than CD56dim NK cells [52], and that in vitro stimulation of CD56bright NK cells with IL-2 results in acquisition of CD16 and KIR expression and loss of IL-7Rɑ and KIT [52]. However, a recent study used genetic barcoding of hematopoietic stem cells in macaques and demonstrated that CD56bright and CD56dim NK cells have distinct developmental precursors [56]. Considered along with new findings of NK/ILC1 plasticity [57–59], these data suggest that the ontological relationships between human NK cells and ILCs may be more nuanced than originally appreciated.
Outside of these major conventional NK cell subsets, two other classes of NK cells bear mention. First, some NKG2ChiCD57hi NK cells that are expanded by HCMV infection display enhanced IFN-γ production and cytotoxic capacity upon secondary challenge by the homologous pathogen and are called ‘adaptive’ NK cells due to their memory-like properties [1,33,60–63]. Understanding the development and function of adaptive NK cells is of considerable interest as their memory-like properties could be harnessed for therapeutic benefit in vaccines or cancer immunotherapy. Secondly, while we have only discussed the properties of circulating NK cells, there are numerous subsets of NK cells resident in various human tissues. These largely tissue-resident NK cells have diverse functions in reproduction and tissue remodeling, and their ontogeny and diversity have been reviewed elsewhere [64,65].
Below, we discuss the genetic and environmental factors that influence the combinatorial expression of various NKRs. These findings are summarized in Figure 1.
KIR diversity and NK cell education
The KIRs, encoded by 14 genes and 2 pseudogenes clustered on chromosome 19, represent an important family of NKRs. KIRs are HLA class I binding receptors and are the second most polymorphic gene family in the human genome, after HLA [66]. KIRs are named according to the relative length of their cytoplasmic tail, with ‘S’ denoting a short tail that generally confers activating activity, and ‘L’ denoting a long tail that corresponds to inhibitory activity. Importantly, KIRs are expressed stochastically by NK cells and this, combined with the polygenic and polymorphic nature of KIRs, contributes significantly to NK cell diversity [33,35,67].
KIR-HLA interactions represent the main mechanism by which NK cells are tolerized to and sense perturbations in the self-HLA environment [68,69]. Although the majority of NK cells express at least one inhibitory KIR for a self-HLA molecule [31], significant proportions of NK cells do not express any self-specific inhibitory KIR [27,70]. To compensate for this, most KIR-negative NK cells express CD94/NKG2A, an inhibitory receptor that binds to the ubiquitously expressed HLA-E [36,71–73]. Therefore, the vast majority of NK cells retain mechanisms to recognize cells that have downregulated HLA class I due to pathogen infection. Nonetheless, approximately 10% of NK cells in a given individual express neither CD94/NKG2A nor self-specific KIR [35,74]. Some of these inhibitory NKR-deficient NK cells are capable of promoting autoimmunity due to their expression of granzyme and perforin. [75,76]. The process that renders NK cells responsive to ‘missing self’ involves functional calibration to self-HLA, and is called education, licensing, or arming [27–31]. Interestingly, recent data suggest that NK cells can also be educated through CD94/NKG2A, and that this form of education results in higher phenotypic diversity, IFN-γ production, and cytotoxic capacity [77].
Despite the apparently stochastic nature of KIR expression during NK cell development, certain KIR-expressing NK cells are favored for survival or expansion based on their ability to interact with their cognate HLA molecule. For instance, there is a preponderance of human NK cell repertoires with a bias for expression of the KIRs that are capable of binding to the HLA molecules expressed in that individual (for instance, KIR2DL1 recognizes HLA-C2, and in individuals homozygous for HLA-C2, there is an increase in the frequency of KIR2DL1-expressing NK cells) [33,78–80]. This biased repertoire is dependent on human cytomegalovirus (HCMV) infection, suggesting that the NK cells expressing KIRs specific for the HLA expressed in that individual are selected for during the response to HCMV infection [33,78–80].
In addition to HCMV infection inducing expansions of NK cells expressing self-specific KIRs, other viral infections have been associated with skewing of the KIR repertoire. In the setting of HCMV seropositivity, chronic Epstein-Barr virus (EBV) infection results in a NK repertoire more dramatically skewed towards self-specific KIRs [81]. In the absence of HCMV infection, infectious mononucleosis during primary EBV infection causes transient fluctuations in the KIR repertoire, but does not result in stable KIR skewing [82,83]. Additionally, Chikungunya virus (CHIKV) selectively modulates the NK KIR repertoire by preferentially expanding those cells expressing HLA-C-specific KIRs, such as KIR2DL1, specific for HLA-C2 alleles, and KIR2DL2/DL3, specific for HLA-C1 alleles [84].
Notably, HIV-1 infection results in the expansion of KIR3DS1- and KIR3DL1-expressing NK cells in individuals expressing the cognate HLA-Bw4–80I epitope [85,86]. In these individuals, KIR3DS1/DL1 is associated with slower progression to AIDS and KIR3DS1/DL1 copy number is inversely correlated with the set point viral load [86–92]. It remains unclear how an activating and an inhibitory NKR with virtually identical extracellular domains both correlate with better clinical outcomes, but recent evidence suggests that this paradox may be explained by heretofore unrecognized KIR3DS1 ligands, the involvement of HIV peptides in modulating the KIR3DS1/HLA-Bw4–80I interaction, and the level of education through KIR3DL1 [32,93–95].
CD57 and CD62L: markers of maturation
CD57 is a carbohydrate epitope created by the enzymatic activity of beta-1,3-glucuronyltransferase and has long been appreciated as a marker of differentiation, maturation, and activation on NK and T cells [96–99]. On NK cells, CD57 expression is limited to CD56dimCD16+ NK cells, is correlated with lower proliferative capacity upon exposure to cytokines or neoplastic cells, and is associated with increased cytolysis and IFN-γ production [97,100–102]. CD62L, on the other hand, is inversely associated with NK cell maturation. CD62L (also known as L-selectin) is a homing receptor important in lymphocyte-endothelial cell interactions. During NK cell development, CD62L is first expressed by CD56bright NK cells and is gradually lost during the processes of education and differentiation [103–106]. This process, perhaps analogous to the shedding of CD62L by T cells following activation, could prime NK cells to leave secondary lymphoid tissues and enter the circulation to mediate innate immune responses.
As markers of differentiation, CD57 and CD62L expression by NK cells is modulated by various viruses. Chronic infection with HCMV, EBV, hepatitis B virus (HBV), and hepatitis C virus (HCV) have all been reported to result in increased expression of CD57 by NK cells [82,83,107,108]. Notably, primary EBV infection induces a transient decrease in CD57 expression on NKG2A+ NK cells, likely due to expansion of CD57- NK cells which either contract or acquire CD57 over time [82]. Although CD57 expression is generally considered to correlate with greater functional capacity, a CD57- NK cell subset was identified as the predominant population that degranulates and produces IFN-γ after co-culture with latent EBV-infected lymphoblastoid cell lines (LCLs) [109]. Additionally, loss of CD62L expression is characteristic of a CD56bright subset of tonsilar NK cells that accumulates during chronic EBV infection and restricts EBV-induced B cell transformation [110]. These findings collectively suggest that viral infections and NK cell maturity have a nuanced relationship at various stages of NK cell development.
C-type lectin receptors
NK cells express several members of the NKG2 C-type lectin receptor family. NKG2A and NKG2C form heterodimers with CD94 and bind the non-classical HLA molecule HLA-E [72,111]. NKG2A is an inhibitory receptor, whereas NKG2C is an activating receptor; similar to the role of inhibitory and activating KIR binding the same HLA ligand, the role of HLA-E inducing two ostensibly contradictory signaling pathways remains unclear [112–116]. However, NKG2C and NKG2A are rarely co-expressed on CD56dim NK cells, suggesting that the contradictory functions of NKG2A- and NKG2C-expressing NK populations may promote immune balance [60,117]. Recognition of HLA-E expression through NKG2A/C is thought to be a secondary mechanism by which NK cells can monitor expression of classical HLA class I molecules, as HLA-E binds peptides derived from the leader sequence of classical HLA class I molecules [118]. NKG2A is expressed on all CD56bright NK cells and is gradually lost during ostensible differentiation to CD56dim NK cells [117,119].
NKG2D is an activating receptor that is more distantly related to the NKG2 family, forms a homodimer, and is reported to bind several ligands, including MICA, MICB, and ULBP1–6 [120–122]. As expression of these and other putative NKG2D ligands is associated with DNA damage, cellular stress, and malignant transformation [123–126], NKG2D is a critical NKR in the control of tumorigenesis [127–129].
Among the most dramatic and well-studied virus-induced NK cell expansions is that of NKG2C+ NK cells in HCMV infection. First identified by Gumá and colleagues in 2004, up to 25% of NK cells in HCMV seropositive individuals express NKG2C, compared to ~2% in HCMV seronegative donors [60]. Expanded NKG2C+ NK cells also tend to be CD57+ [62], mediate superior ADCC, and produce higher levels of IFN-γ and TNF. Notably, in NKG2C+ NK cells in HCMV seropositive individuals, IFN-γ and TNF promoters are stably hypomethylated [61,130–133]. This epigenetic reprogramming may provide a mechanistic explanation for the memory-like phenotypes observed in these NKG2C+ NK cells.
Expansions of NKG2C+ NK cells have also been reported in the context of immunodeficiency [134], organ transplantation [63,135–137], hantavirus infection [138], CHIKV infection [84], HIV-1 infection [139], and chronic HBV and HCV infection [107,140,141]. It remains unclear, however, if NKG2C+ NK cell expansions induced by viruses other than HCMV are the result of a direct impact of the virus on NKG2C expression. Unfortunately, most studies that demonstrated expansion of NKG2C+ NK cells during infection by viruses other than HCMV did not stratify donors based on HCMV seropositivity. It is conceivable, therefore, that the observed NKG2C+ expansions are actually the result of a subclinical HCMV reactivation [142]. Primary EBV infection does not alter NKG2C expression in the NK cell repertoire, thus supporting the hypothesis that NKG2C expansions are an HCMV-specific phenomenon [82,83,143].
NKG2A+ NK cells have also been reported to expand in the context of viral infection, particularly by EBV. Primary EBV infection expands a population of early-differentiated CD56dimNKG2A+KIR- NK cells that do not contract and gradually gain CD57 expression and lose CD62L expression over time [82,83,109,144,145]. Accumulation of this subset inversely correlates with EBV DNA levels in peripheral blood mononuclear cells (PBMCs), and is endowed with increased IFN-γ production and cytotoxic degranulation against autologous LCLs [82,109]. EBV-induced NKG2A+ expansions are not limited to CD56dim NK cells, as a distinct CD56brightIFN-γhiNKG2A+ NK cell subset accumulates in the tonsils of EBV seropositive individuals [110]. Although it is unknown whether other viruses cause NKG2A+ NK cell expansions, NKG2A+ NK cells are implicated in better control of HIV-1 infection in in vitro models [146], due to an HLA-E-presented HIV peptide that prevents inhibitory interaction with NKG2A [147].
Several viral infections have been associated with changes in NKG2D expression by NK cells. There is an increase in NKG2D+ NK cell frequency in EBV seropositive individuals, and such an increase is apparent as soon as 10 days after first EBV exposure in an in vitro model of infection [109,143,148]. NKG2D, along with NKG2A, is one marker represented on the predominant NK cell subset that produces IFN-γ and degranulates in response to coculture with EBV-infected LCLs [109]. HCV, on the other hand, appears to have a more nuanced impact on NKG2D expression by NK cells. Acute HCV infection induces an increase in NKG2D expression by both CD56bright and CD56dim NK cells [149]. However, patients with chronic HCV infection have lower frequencies of NKG2D+ NK cells which have impaired cytotoxic potential and lower IFN-γ production; this may be due to increased levels of circulating IL-10 and TGF-β during chronic HCV infection [150]. Importantly, HCV-induced NKG2D downregulation can be reversed by addition of IL-15 in vitro, representing a potential way to bolster current treatment of chronic HCV infection.
Natural cytotoxicity receptors
Natural cytotoxicity receptors (NCRs) are members of the Ig-superfamily and represent a set of recently evolved NKRs [151]. Some NCRs, including NKp30, NKp46, and NKp80, are expressed on both activated and resting NK cells, whereas others, including NKp44, are upregulated after stimulation [152,153]. NCRs have been reported to be critical both in the control of viral infection and tumors. For example, higher NCR expression correlates with enhanced control of and delayed progression during HIV infection [154,155], and deletion of single NCRs impairs NK cell cytotoxic capacity against tumor cells [156,157]. There are many putative NCR ligands that have not been confirmed by independent research groups, and several conflicting reports on the role of particular NCRs in a given viral infection [120,158–162]. Refined studies of NCR signaling and structural analysis of NCR-ligand interactions will be necessary to dissect the complex roles of this receptor family in NK cell biology.
The impact of particular viruses on NK cell expression of NCRs remains poorly studied. During acute HCV infection, both NKp30 and NKp46 are upregulated exclusively on CD56bright NK cells [149]. Additionally, NK cells stimulated with EBV-infected LCLs ex vivo have increased frequencies of NKp30+ cells [143,148]. NCR expression is also altered on CD56neg NK cells in the setting of chronic infection. CD56neg NK cells are an aberrant subset of NK cells that are found in low frequencies in healthy individuals, but expand during chronic HIV and HCV infection. These cells are likely derived from CD56dim NK cells, as they express CD16 and KIRs, but have impaired capacity for cytokine production, cytolysis, and proliferation [163]. Although early studies on CD56neg NK cells may be unreliable as a result of incomplete exclusion of monocytes during flow cytometric gating analysis, more recent studies have confirmed that CD56neg NK cells express lower levels of NKp30 and NKp46 in chronic HIV infection [164–167].
Other NKRs
Expression of several other NKRs have been reported to be influenced by different viral infections. 2B4 (also called CD244 or SLAMF4) is an NKR that interacts with CD48, which is expressed by all hematopoietic cells [168–170]. Although murine 2B4 has predominantly inhibitory function [171,172], human 2B4 is conventionally considered to be an activating receptor that may function as a co-receptor for other activating NKRs like NKp46 [151,173]. Exposure of human NK cells to EBV-infected LCLs or the EBV-producing Akata cell line result in higher frequencies of 2B4+ NK cells [109,143], and a 2B4+ subset is responsible for greater IFN-γ production and cytotoxicity in this setting [109]. Additionally, acute HCV infection induces upregulation of 2B4 on both CD56bright and CD56dim NK cells [149].
Parallel to the Fas/FasL system, TNF-related apoptosis-inducing ligand (TRAIL) is a ligand for five death receptors that mediates apoptosis [174]. As such, TRAIL represents one effector mechanism for NK cells. TRAIL is required for IFN-γ-dependent suppression of tumor growth in vivo [175], and blockade of TRAIL activity in encephalomyocarditis virus (ECMV)-infected mice resulted in higher viral titers and earlier death [176]. TRAIL is upregulated by human NK cells during acute HCMV, EBV, and HCV infection, perhaps representing one mechanism by which NK cells provide early control of infection by these viruses [143,149,177].
DNAM-1 (also called CD226) is an activating NKR and adhesin that binds Nectin-2 (CD112) and Necl-5 (PVR), which are upregulated on several tumors [178-180]. DNAM-1 is also involved in the NK cell response to HCMV, HIV, and HCV infection [149,181–183]. Primary EBV infection has recently been reported to upregulate DNAM-1 on NK cells, although the functional consequences of this upregulation are unknown [143,148].
LILRB1 (also known as LIR1 or ILT2) is an HLA class I-binding inhibitory NKR, although KIRs and CD94/NKG2A are thought to dominate HLA class I-mediated inhibition of NK cells [184–186]. LILRB1 expression correlates with CD57 expression, suggesting that LILRB1 may be a marker of NK cell differentiation [107]. Despite its role as an inhibitory receptor, LILRB1+ NK cells are implicated in better control of HIV-1 infection [187]. However, this may simply reflect the higher cytotoxic potential of more mature NK cells. LILRB1 is also upregulated on NK cells during infection with HBV, HCV, and EBV [107,145].
NK diversification and epigenetic reprogramming
While the NK cell repertoire is remarkably malleable and influenced by viral infections, it remains stable in the absence of environmental perturbations [188]. This suggests that epigenomic modifications may be involved in the processes of NK cell maturation and diversification. Although this ostensible epigenomic reprogramming remains poorly understood in conventional NK cells, there is evidence that epigenetic modifications could represent one mechanism that accounts for the rapidity of NK cell responses. During development, murine NK cells have a long-range acetylation profile across the Ifng locus that is rapidly modifiable by cytokine stimulation [189]. Such hyperacetylation should presumably result in rapid and robust IFN-γ production in appropriate activating contexts. Additionally, HCMV-expanded NKG2Chi NK cells display complete demethylation of the IFNG promoter, which is required to induce stable IFN-γ production after engagement of NKG2C or 2B4 [61]. Clues to epigenetic regulation of NK cell effector function may also be present in studies of T cells. For example, H3K9 acetylation of the GZMB (encoding granzyme B) and PRF1 (encoding perforin) promoters increases granzyme B and perforin expression and enhances CD8+ T cell-mediated cytotoxicity [190]. Stat4-dependent recruitment of Brahma-related gene 1 (Brg1), a chromatin remodeling complex [191], remodels nucleosomes in the IFNG promoter of Th1 cells [192]. IFN-γ expression by CD56dim NK cells may be regulated by a similar mechanism, as CD56dim NK cells express high levels of Stat4 and Brg1 [105]. How CD56bright and CD56dim NK cells tightly and differentially regulate IFN-γ expression would provide valuable understanding of the mechanisms governing functional specialization of NK cell subsets.
Epigenetic reprogramming of adaptive NK cells, on the other hand, has been more thoroughly characterized. A recent pair of studies has demonstrated that HCMV drives the differentiation and diversification of adaptive human NK cells through epigenetic silencing of several signaling proteins and transcription factors [131,132]. These studies have provided a model for adaptive NK cell diversification whereby engagement of DAP12-coupled activating receptors results in epigenetic imprinting of the ZBTB16 locus, resulting in loss of PLZF expression. PLZF-deficient NK cells also display stochastic hypermethylation of promoters encoding signaling proteins SYK, EAT-2, DAB2, and FcεRγ, resulting in diversification of the adaptive NK cell repertoire. Lower expression of these signaling proteins results in lower expression of NCRs NKp30 and NKp46, as well as decreased signaling through SLAM family receptors, including 2B4 [133]. The transcriptional profiles and epigenomic changes induced by other viruses remain unknown, and could represent valuable insights into the diversification and maintenance of the conventional NK cell repertoire.
How does immune experience diversify NK cells?
Although the virus-induced shifts in expression of particular NKRs have been fairly well-characterized, the impact on NK repertoire diversity and the nature of the NK diversification process remain poorly understood. While there are many methods to assess diversity, the Inverse Simpson Index is commonly used to quantify the diversity of leukocytes as well as the microbiome, in part because it does not require normally distributed data [41]. It measures two metrics of a sample to quantify diversity: richness, the number of individual subpopulations per sample, and evenness, the degree to which each subpopulation is represented in the sample. In theory, education, differentiation, and maturation of NK cells should all serve to increase NK repertoire diversity, because all three processes result in the expression of previously un-expressed NKRs (including KIRs, CD57, and CD94/NKG2C), thereby increasing repertoire richness. However, if a lymphocyte undergoes a clonal expansion in response to a particular pathogen, the diversity of that lymphocyte class should decrease by the Inverse Simpson Index, because evenness is decreasing with no apparent change in richness. There is evidence that viruses other than HCMV, like EBV, HIV, and CHIKV, induce clonal-like expansions of NK cells from a well-differentiated NK cell repertoire [81,84–86,193]. Thus, exposure of a mature NK cell repertoire to these viruses might be expected to decrease NK cell diversity [65].
There are suggestions that this is not the case. First, NK cell diversity increases with age. Although differences in the structure of the human NK cell repertoire between cord blood and adult peripheral blood are largely limited to maturity-related markers like CD57 and NKG2A [194], NK cell diversity is higher in adult peripheral blood than in cord blood [188]. NK cell diversification with age could be a physiologic process, or represent the accumulation of diversity induced by exposures to new pathogens. The latter hypothesis is supported by the recent observation that NK cell diversity increases after short-term exposure to cells infected with HIV-1 or West Nile Virus [188]. As it does not seem likely that interacting with virus-infected cells should increase the evenness of NK cell subpopulations, it is reasonable to suspect that these viruses cause a process akin to differentiation where NK cell repertoire richness increases. While this could represent an adaptation to generate a variety of NK cell subsets, some of which might be better tuned to respond to the viral threat, there are several noteworthy considerations when interpreting these data. The viral exposures were performed in vitro, and whether viral exposure in vivo has a similar impact on NK cell diversity remains an open question. Additionally, the Inverse Simpson index represents only one method to measure diversity, may be sensitive to the particular NKRs used in the diversity calculation, and may not reflect different functional phenotypes of samples [32]. Nonetheless, the receptor-ligand interactions and signaling modalities that govern rapid virus-induced NK cell diversification in vitro or maturity-related diversification in vivo could represent important factors influencing NK cell repertoire stability and function, as well as potential viral evasion strategies. The existing data on immune experience largely reflect studies of the effects of viral infection. It will be of interest in future studies to evaluate the impact of malignancy on the phenotypic and functional diversity of the NK cell repertoire.
Importance of NK cell diversity in health and disease: lessons learned from deep profiling
It is appreciated that NK cells have critical roles in the control of early viral infection and malignant transformation [1,2,10,24,195], and the importance of functional specialization of the major NK cell subsets in human health and disease is also well characterized [132,196]. To our knowledge, only one study to date has examined how NK cell repertoire diversity impacts viral susceptibility. This study, performed by our research group, used CyTOF to demonstrate, surprisingly, that NK cell diversity correlated positively with risk of HIV-1 acquisition in a cohort of Kenyan women [188]. Risk of HIV-1 acquisition was correlated neither with CD4 or CD8 T cell diversity nor with expression of specific NK cell receptors. These results are counterintuitive, as lymphocyte diversity is conventionally considered to be beneficial by allowing lymphocytes to recognize and respond to a greater breadth of potential novel pathogens. There are two notable caveats to this study: its data are generated from a small cohort (HIV-1 acquisition, n = 13; matched controls, n = 23) and diversity may be a confounder that correlates with some unidentified metric that itself represents the link to increased viral susceptibility.
The caveats notwithstanding, one way to explain the paradox presented by this study is to posit that NK repertoire diversity is inversely related to repertoire flexibility. Therefore, greater NK cell diversification could impair the ability of the repertoire to respond to and control a novel pathogen, thereby increasing viral susceptibility. Though this hypothesis remains unconfirmed, there are suggestions in the literature that corroborate this assertion. In particular, it appears that murine cytomegalovirus (MCMV)-induced adaptive murine NK cells have decreased capacity to control heterologous infections by influenza virus or Listeria monocytogenes compared to their naive counterparts [197]. This underscores the antigen-specificity of adaptive NK cell responses and supports the notion that virus-induced repertoire diversification may be akin to lineage commitment, where differentiated NK cells are unable to respond to de novo pathogens and thus impair the functional capacity of the repertoire as a whole.
Conclusion: future directions in single-cell technologies
Despite the progress that has been made in deep profiling NK cells to understand the genetic and environmental determinants of their diversification, much uncertainty still remains as to the mechanisms of NK diversification and its role in human health and disease. What epigenetic mechanisms promote NK cell repertoire stability? What are the receptor-ligand interactions and signaling pathways that govern virus-induced NK cell diversification? Which NK cell subsets are signatures of certain viral infections, and do these signatures represent possible viral evasion strategies? Can we identify specific subsets of NK cells that are best adapted to promoting immune homeostasis or controlling particular viral infections or cancers? Does NK cell diversity decrease repertoire flexibility and promote viral susceptibility?
The advent of single-cell technologies for deep profiling of NK cells has made answering such questions imminently possible. Our group and others have recently pioneered the use of mass cytometry to better dissect NK cell diversity [41,109,143,188,198,199]. Single-cell resolution deep sequencing and proteomics methods will also facilitate further complex interrogation [200]. These methods should provide a deeply needed understanding of the links between NK cell phenotype and functional capacity that will be essential for future NK cell-based therapeutic developments.
Acknowledgments
We would like to thank Kirk Fetters and Drs. Mathieu Le Gars and Laura Simpson for helpful discussion and critical readings of the manuscript. AJW is supported by NIH T32GM007365–42 and CAB by NIH DP2AI112193–01, NIH/NIAID U01AI131302–01, the Burroughs Wellcome Fund Investigators in the Pathogenesis of Infectious Diseases, and NIH/NIAID 5U19AI057229–14 (to Mark M. Davis). CAB is a Chan Zuckerberg Investigator and the Tashia and John Morgridge Faculty Scholar from the Stanford Child Health Research Institute.
Abbreviations
- NK
natural killer
- ILC
innate lymphoid cell
- IFN-γ
interferon gamma
- NKR
natural killer cell receptor
- KIR
killer immunoglobulin-like receptor
- CyTOF
cytometry by time-of-flight
- CD
cluster of differentiation
- IL
interleukin
- HLA
human leukocyte antigen
- HCMV
human cytomegalovirus
- EBV
Epstein-Barr virus
- CHIKV
Chikungunya virus
- AIDS
acquired immunodeficiency syndrome
- HIV
human immunodeficiency virus
- HBV
hepatitis B virus
- HCV
hepatitis C virus
- LCL
lymphoblastoid cell line
- MIC
MHC class I-related protein
- ULBP
UL16 binding protein
- ADCC
antibody-dependent cellular cytotoxicity
- TNF
tumor necrosis factor
- TGF
transforming growth factor
- PBMC
peripheral blood mononuclear cell
- NCR
natural cytotoxicity receptor
- TRAIL
TNF-related apoptosis-inducing ligand
- DNAM
DNAX accessory molecule
- LILRB1
leukocyte immunoglobulin-like receptor B1
- PLZF
promyelocytic leukemia zinc finger protein
- MCMV
murine cytomegalovirus
Footnotes
Conflicts of interest
No conflicts of interest to report.
References
- 1.Cerwenka A, Lanier LL. 2016. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol 16:112–123. [DOI] [PubMed] [Google Scholar]
- 2.Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer cells. Nat. Immunol 9:503–510. [DOI] [PubMed] [Google Scholar]
- 3.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, Powrie F, Vivier E. 2013. Innate lymphoid cells--a proposal for uniform nomenclature. Nat. Rev. Immunol 13:145–149. [DOI] [PubMed] [Google Scholar]
- 4.McKenzie ANJ, Spits H, Eberl G. 2014. Innate lymphoid cells in inflammation and immunity. Immunity. 41:366–374. [DOI] [PubMed] [Google Scholar]
- 5.Artis D, Spits H. 2015. The biology of innate lymphoid cells. Nature. 517:293–301. [DOI] [PubMed] [Google Scholar]
- 6.Spits H, Di Santo JP. 2011. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat. Immunol 12:21–27. [DOI] [PubMed] [Google Scholar]
- 7.Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. 2015. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science. 348:aaa6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Herberman RB, Nunn ME, Lavrin DH. 1975. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int. J. Cancer 16:216–229. [DOI] [PubMed] [Google Scholar]
- 9.Kiessling R, Klein E, Wigzell H. 1975. ‘Natural’ killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur. J. Immunol 5:112–117. [DOI] [PubMed] [Google Scholar]
- 10.Lodoen MB, Lanier LL. 2006. Natural killer cells as an initial defense against pathogens. Curr. Opin. Immunol 18:391–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Waggoner SN, Cornberg M, Selin LK, Welsh RM. 2012. Natural killer cells act as rheostats modulating anti-viral T cells. Nature. Available from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3539796/. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schleinitz N, Vély F, Harlé J-R, Vivier E. 2010. Natural killer cells in human autoimmune diseases. Immunology. 131:451–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bielekova B, Catalfamo M, Reichert-Scrivner S, Packer A, Cerna M, Waldmann TA, McFarland H, Henkart PA, Martin R. 2006. Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. Proc. Natl. Acad. Sci. U. S. A 103:5941–5946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Aramaki T, Ida H, Izumi Y, Fujikawa K, Huang M, Arima K, Tamai M, Kamachi M, Nakamura H, Kawakami A, Origuchi T, Matsuoka N, Eguchi K. 2009. A significantly impaired natural killer cell activity due to a low activity on a per-cell basis in rheumatoid arthritis. Mod. Rheumatol 19:245–252. [DOI] [PubMed] [Google Scholar]
- 15.Park Y-W, Kee S-J, Cho Y-N, Lee E-H, Lee H-Y, Kim E-M, Shin M-H, Park J-J, Kim T-J, Lee S-S, Yoo D-H, Kang H-S. 2009. Impaired differentiation and cytotoxicity of natural killer cells in systemic lupus erythematosus. Arthritis Rheum. 60:1753–1763. [DOI] [PubMed] [Google Scholar]
- 16.Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. 2000. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet. 356:1795–1799. [DOI] [PubMed] [Google Scholar]
- 17.Guillerey C, Smyth MJ. 2016. NK Cells and Cancer Immunoediting. Curr. Top. Microbiol. Immunol 395:115–145. [DOI] [PubMed] [Google Scholar]
- 18.Guillerey C, Huntington ND, Smyth MJ. 2016. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol 17:1025–1036.27540992 [Google Scholar]
- 19.Biron CA, Byron KS, Sullivan JL. 1989. Severe Herpesvirus Infections in an Adolescent without Natural Killer Cells. N. Engl. J. Med 320:1731–1735. [DOI] [PubMed] [Google Scholar]
- 20.Eidenschenk C, Dunne J, Jouanguy E, Fourlinnie C, Gineau L, Bacq D, McMahon C, Smith O, Casanova J-L, Abel L, Feighery C. 2006. A Novel Primary Immunodeficiency with Specific Natural-Killer Cell Deficiency Maps to the Centromeric Region of Chromosome 8. Am. J. Hum. Genet 78:721–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nakajima H, Cella M, Bouchon A, Grierson HL, Lewis J, Duckett CS, Cohen JI, Colonna M. 2000. Patients with X-linked lymphoproliferative disease have a defect in 2B4 receptor-mediated NK cell cytotoxicity. Eur. J. Immunol 30:3309–3318. [DOI] [PubMed] [Google Scholar]
- 22.Parolini S, Bottino C, Falco M, Augugliaro R, Giliani S, Franceschini R, Ochs HD, Wolf H, Bonnefoy JY, Biassoni R, Moretta L, Notarangelo LD, Moretta A. 2000. X-linked lymphoproliferative disease. 2B4 molecules displaying inhibitory rather than activating function are responsible for the inability of natural killer cells to kill Epstein-Barr virus-infected cells. J. Exp. Med 192:337–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rydyznski CE, Waggoner SN. 2015. Boosting vaccine efficacy the natural (killer) way. Trends Immunol. 36:536–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Strauss-Albee DM, Blish CA. 2016. Human NK Cell Diversity in Viral Infection: Ramifications of Ramification. Front. Immunol 7:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hromadnikova I, Pirkova P, Sedlackova L. 2013. Influence of in vitro IL-2 or IL-15 alone or in combination with Hsp-70-derived 14-mer peptide (TKD) on the expression of NK cell activatory and inhibitory receptors. Mediators Inflamm. 2013:405295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nemazee D 2000. Receptor selection in B and T lymphocytes. Annu. Rev. Immunol 18:19–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Fernandez NC, Treiner E, Vance RE, Jamieson AM, Lemieux S, Raulet DH. 2005. A subset of natural killer cells achieves self-tolerance without expressing inhibitory receptors specific for self-MHC molecules. Blood. 105:4416–4423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song Y-J, Yang L, French AR, Sunwoo JB, Lemieux S, Hansen TH, Yokoyama WM. 2005. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature. 436:709–713. [DOI] [PubMed] [Google Scholar]
- 29.Elliott JM, Yokoyama WM. 2011. Unifying concepts of MHC-dependent natural killer cell education. Trends Immunol. 32:364–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Höglund P, Brodin P. 2010. Current perspectives of natural killer cell education by MHC class I molecules. Nat. Rev. Immunol 10:724–734. [DOI] [PubMed] [Google Scholar]
- 31.Goodridge JP, Önfelt B, Malmberg K-JJ. 2015. Newtonian cell interactions shape natural killer cell education. Immunol. Rev 267:197–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Blish CA. 2016. Natural Killer Cell Diversity in Viral Infection: Why and How Much? Pathogens & immunity. 1:165–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Béziat V, Liu LL, Malmberg J-AA, Ivarsson MA, Sohlberg E, Björklund AT, Retière C, Sverremark-Ekström E, Traherne J, Ljungman P, Schaffer M, Price DA, Trowsdale J, Michaëlsson J, Ljunggren H-GG, Malmberg K-JJ. 2013. NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs. Blood. 121:2678–2688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Shilling HG, Guethlein LA, Cheng NW, Gardiner CM, Rodriguez R, Tyan D, Parham P. 2002. Allelic polymorphism synergizes with variable gene content to individualize human KIR genotype. J. Immunol 168:2307–2315. [DOI] [PubMed] [Google Scholar]
- 35.Uhrberg M, Valiante NM, Shum BP, Shilling HG, Lienert-Weidenbach K, Corliss B, Tyan D, Lanier LL, Parham P. 1997. Human diversity in killer cell inhibitory receptor genes. Immunity. 7:753–763. [DOI] [PubMed] [Google Scholar]
- 36.Yawata M, Yawata N, Draghi M, Partheniou F, Little A-M, Parham P. 2008. MHC class I–specific inhibitory receptors and their ligands structure diverse human NK-cell repertoires toward a balance of missing self-response. Blood. 112:2369–2380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yawata M, Yawata N, Draghi M, Little A-M, Partheniou F, Parham P. 2006. Roles for HLA and KIR polymorphisms in natural killer cell repertoire selection and modulation of effector function. J. Exp. Med 203:633–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Martinet L, Ferrari De Andrade L, Guillerey C, Lee JS, Liu J, Souza-Fonseca-Guimaraes F, Hutchinson DS, Kolesnik TB, Nicholson SE, Huntington ND, Smyth MJ. 2015. DNAM-1 expression marks an alternative program of NK cell maturation. Cell Rep. 11:85–97. [DOI] [PubMed] [Google Scholar]
- 39.Cooper MA, Fehniger TA, Caligiuri MA. 2001. The biology of human natural killer-cell subsets. Trends Immunol. 22:633–640.11698225 [Google Scholar]
- 40.Angelo LS, Banerjee PP, Monaco-Shawver L, Rosen JB, Makedonas G, Forbes LR, Mace EM, Orange JS. 2015. Practical NK cell phenotyping and variability in healthy adults. Immunol. Res 62:341–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Horowitz A, Strauss-Albee DM, Leipold M, Kubo J, Nemat-Gorgani N, Dogan OC, Dekker CL, Mackey S, Maecker H, Swan GE, Davis MM, Norman PJ, Guethlein LA, Desai M, Parham P, Blish CA. 2013. Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci. Transl. Med 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bendall SC, Simonds EF, Qiu P, Amir E-AD, Krutzik PO, Finck R, Bruggner RV, Melamed R, Trejo A, Ornatsky OI, Balderas RS, Plevritis SK, Sachs K, Pe’er D, Tanner SD, Nolan GP. 2011. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science. 332:687–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Spitzer MH, Nolan GP. 2016. Mass Cytometry: Single Cells, Many Features. Cell. 165:780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kling J. 2015. Cytometry: Measure for measure. Nature. 518:439–443. [DOI] [PubMed] [Google Scholar]
- 45.Scoville SD, Freud AG, Caligiuri MA. 2017. Modeling Human Natural Killer Cell Development in the Era of Innate Lymphoid Cells. Front. Immunol 8:360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yu J, Freud AG, Caligiuri MA. 2013. Location and cellular stages of natural killer cell development. Trends Immunol. 34:573–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Luetke-Eversloh M, Killig M, Romagnani C. 2013. Signatures of human NK cell development and terminal differentiation. Front. Immunol 4:499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, Caligiuri MA. 2006. Evidence for discrete stages of human natural killer cell differentiation in vivo. J. Exp. Med 203:1033–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lim AI, Verrier T, Vosshenrich CA, Di Santo JP. 2017. Developmental options and functional plasticity of innate lymphoid cells. Curr. Opin. Immunol 44:61–68. [DOI] [PubMed] [Google Scholar]
- 50.Zook EC, Kee BL. 2016. Development of innate lymphoid cells. Nat. Immunol 17:775–782. [DOI] [PubMed] [Google Scholar]
- 51.Diefenbach A, Colonna M, Koyasu S. 2014. Development, differentiation, and diversity of innate lymphoid cells. Immunity. 41:354–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Romagnani C, Juelke K, Falco M, Morandi B, D’Agostino A, Costa R, Ratto G, Forte G, Carrega P, Lui G, Conte R, Strowig T, Moretta A, Münz C, Thiel A, Moretta L, Ferlazzo G. 2007. CD56brightCD16- killer Ig-like receptor- NK cells display longer telomeres and acquire features of CD56dim NK cells upon activation. J. Immunol 178:4947–4955. [DOI] [PubMed] [Google Scholar]
- 53.Carson WE, Fehniger TA, Caligiuri MA. 1997. CD56bright natural killer cell subsets: characterization of distinct functional responses to interleukin-2 and the c-kit ligand. Eur. J. Immunol 27:354–360. [DOI] [PubMed] [Google Scholar]
- 54.Freud AG, Yu J, Caligiuri MA. 2014. Human natural killer cell development in secondary lymphoid tissues. Semin. Immunol 26:132–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lanier LL, Le AM, Civin CI, Loken MR, Phillips JH. 1986. The relationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigen expression on human peripheral blood NK cells and cytotoxic T lymphocytes. J. Immunol 136:4480–4486. [PubMed] [Google Scholar]
- 56.Wu C, Li B, Lu R, Koelle SJ, Yang Y, Jares A, Krouse AE, Metzger M, Liang F, Loré K, Wu CO, Donahue RE, Chen ISY, Weissman I, Dunbar CE. 2014. Clonal tracking of rhesus macaque hematopoiesis highlights a distinct lineage origin for natural killer cells. Cell Stem Cell. 14:486–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Silver JS, Humbles AA. 2017. NK cells join the plasticity party. Nat. Immunol 18:959–960. [DOI] [PubMed] [Google Scholar]
- 58.Cortez VS, Ulland TK, Cervantes-Barragan L, Bando JK, Robinette ML, Wang Q, White AJ, Gilfillan S, Cella M, Colonna M. 2017. SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-β signaling. Nat. Immunol 18:995–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gao Y, Souza-Fonseca-Guimaraes F, Bald T, Ng SS, Young A, Ngiow SF, Rautela J, Straube J, Waddell N, Blake SJ, Yan J, Bartholin L, Lee JS, Vivier E, Takeda K, Messaoudene M, Zitvogel L, Teng MWL, Belz GT, Engwerda CR, Huntington ND, Nakamura K, Hölzel M, Smyth MJ. 2017. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol 18:1004–1015. [DOI] [PubMed] [Google Scholar]
- 60.Gumá M, Angulo A, Vilches C, Gómez-Lozano N, Malats N, López-Botet M. 2004. Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. Blood. 104:3664–3671. [DOI] [PubMed] [Google Scholar]
- 61.Luetke-Eversloh M, Hammer Q, Durek P, Nordström K, Gasparoni G, Pink M, Hamann A, Walter J, Chang H-D, Dong J, Romagnani C. 2014. Human cytomegalovirus drives epigenetic imprinting of the IFNG locus in NKG2Chi natural killer cells. PLoS Pathog. 10:e1004441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lopez-Vergès S, Milush JM, Schwartz BS, Pando MJ, Jarjoura J, York VA, Houchins JP, Miller S, Kang S-M, Norris PJ, Nixon DF, Lanier LL. 2011. Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc. Natl. Acad. Sci. U. S. A 108:14725–14732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Foley B, Cooley S, Verneris MR, Pitt M, Curtsinger J, Luo X, Lopez-Vergès S, Lanier LL, Weisdorf D, Miller JS. 2012. Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. Blood. 119:2665–2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sojka DK, Tian Z, Yokoyama WM. 2014. Tissue-resident natural killer cells and their potential diversity. Semin. Immunol 26:127–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Björkström NK, Ljunggren H-GG, Michaëlsson J. 2016. Emerging insights into natural killer cells in human peripheral tissues. Nat. Rev. Immunol 16:310–320. [DOI] [PubMed] [Google Scholar]
- 66.Kulkarni S, Martin MP, Carrington M. 2008. The Yin and Yang of HLA and KIR in human disease. Semin. Immunol 20:343–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Uhrberg M 2005. Shaping the human NK cell repertoire: an epigenetic glance at KIR gene regulation. Mol. Immunol 42:471–475. [DOI] [PubMed] [Google Scholar]
- 68.Rahim MMA, Tu MM, Mahmoud AB, Wight A, Abou-Samra E, Lima PDA, Makrigiannis AP. 2014. Ly49 receptors: innate and adaptive immune paradigms. Front. Immunol 5:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Parham P, Norman PJ, Abi-Rached L, Guethlein LA. 2012. Human-specific evolution of killer cell immunoglobulin-like receptor recognition of major histocompatibility complex class I molecules. Philos. Trans. R. Soc. Lond. B Biol. Sci 367:800–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Fauriat C, Andersson S, Björklund AT, Carlsten M, Schaffer M, Björkström NK, Baumann BC, Michaëlsson J, Ljunggren H-G, Malmberg K-J. 2008. Estimation of the size of the alloreactive NK cell repertoire: studies in individuals homozygous for the group A KIR haplotype. J. Immunol 181:6010–6019. [DOI] [PubMed] [Google Scholar]
- 71.Veinotte LL, Wilhelm BT, Mager DL, Takei F. 2003. Acquisition of MHC-specific receptors on murine natural killer cells. Crit. Rev. Immunol 23:251–266. [DOI] [PubMed] [Google Scholar]
- 72.Braud VM, Allan DS, O’Callaghan CA, Söderström K, D’Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, Lanier LL, McMichael AJ. 1998. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature. 391:795–799. [DOI] [PubMed] [Google Scholar]
- 73.Andersson S, Fauriat C, Malmberg J-A, Ljunggren H-G, Malmberg K-J. 2009. KIR acquisition probabilities are independent of self-HLA class I ligands and increase with cellular KIR expression. Blood. 114:95–104. [DOI] [PubMed] [Google Scholar]
- 74.Andersson S, Malmberg J-A, Malmberg K-J. 2010. Tolerant and diverse natural killer cell repertoires in the absence of selection. Exp. Cell Res 316:1309–1315. [DOI] [PubMed] [Google Scholar]
- 75.Cooley S, Xiao F, Pitt M, Gleason M, McCullar V, Bergemann TL, McQueen KL, Guethlein LA, Parham P, Miller JS. 2007. A subpopulation of human peripheral blood NK cells that lacks inhibitory receptors for self-MHC is developmentally immature. Blood. 110:578–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Anfossi N, André P, Guia S, Falk CS, Roetynck S, Stewart CA, Breso V, Frassati C, Reviron D, Middleton D, Romagné F, Ugolini S, Vivier E. 2006. Human NK cell education by inhibitory receptors for MHC class I. Immunity. 25:331–342. [DOI] [PubMed] [Google Scholar]
- 77.Horowitz A, Djaoud Z, Nemat-Gorgani N, Blokhuis J, Hilton HG, Béziat V, Malmberg K-JJ, Norman PJ, Guethlein LA, Parham P. 2016. Class I HLA haplotypes form two schools that educate NK cells in different ways. Science immunology. 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Held W, Dorfman JR, Wu MF, Raulet DH. 1996. Major histocompatibility complex class I-dependent skewing of the natural killer cell Ly49 receptor repertoire. Eur. J. Immunol 26:2286–2292. [DOI] [PubMed] [Google Scholar]
- 79.Schönberg K, Sribar M, Enczmann J, Fischer JC, Uhrberg M. 2011. Analyses of HLA-C–specific KIR repertoires in donors with group A and B haplotypes suggest a ligand-instructed model of NK cell receptor acquisition. Blood. 117:98–107. [DOI] [PubMed] [Google Scholar]
- 80.Schönberg K, Fischer JC, Kögler G, Uhrberg M. 2011. Neonatal NK-cell repertoires are functionally, but not structurally, biased toward recognition of self HLA class I. Blood. 117:5152–5156. [DOI] [PubMed] [Google Scholar]
- 81.Saghafian-Hedengren S, Sohlberg E, Theorell J, Carvalho-Queiroz C, Nagy N, Persson J-O, Nilsson C, Bryceson YT, Sverremark-Ekström E. 2013. Epstein-Barr virus coinfection in children boosts cytomegalovirus-induced differentiation of natural killer cells. J. Virol 87:13446–13455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Azzi T, Lünemann A, Murer A, Ueda S, Béziat V, Malmberg K-JJ, Staubli G, Gysin C, Berger C, Münz C, Chijioke O, Nadal D. 2014. Role for early-differentiated natural killer cells in infectious mononucleosis. Blood. 124:2533–2543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hendricks DW, Balfour HH, Dunmire SK, Schmeling DO, Hogquist KA, Lanier LL. 2014. Cutting Edge: NKG2ChiCD57+ NK Cells Respond Specifically to Acute Infection with Cytomegalovirus and Not Epstein–Barr Virus. The Journal of Immunology. 192:4492–4496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Petitdemange C, Becquart P, Wauquier N, Béziat V, Debré P, Leroy EM, Vieillard V. 2011. Unconventional repertoire profile is imprinted during acute chikungunya infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog. 7:e1002268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Alter G, Rihn S, Walter K, Nolting A, Martin M, Rosenberg ES, Miller JS, Carrington M, Altfeld M. 2009. HLA class I subtype-dependent expansion of KIR3DS1+ and KIR3DL1+ NK cells during acute human immunodeficiency virus type 1 infection. J. Virol 83:6798–6805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pelak K, Need AC, Fellay J, Shianna KV, Feng S, Urban TJ, Ge D, De Luca A, Martinez-Picado J, Wolinsky SM, Martinson JJ, Jamieson BD, Bream JH, Martin MP, Borrow P, Letvin NL, McMichael AJ, Haynes BF, Telenti A, Carrington M, Goldstein DB, Alter G, NIAID Center for HIV/AIDS Vaccine Immunology. 2011. Copy number variation of KIR genes influences HIV-1 control. PLoS Biol. 9:e1001208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jennes W, Verheyden S, Mertens JW, Camara M, Seydi M, Dieye TN, Mboup S, Demanet C, Kestens L. 2013. Inhibitory KIR/HLA incompatibility between sexual partners confers protection against HIV-1 transmission. Blood. 121:1157–1164. [DOI] [PubMed] [Google Scholar]
- 88.López-Vázquez A, Miña-Blanco A, Martínez-Borra J, Njobvu PD, Suárez-Alvarez B, Blanco-Gelaz MA, González S, Rodrigo L, López-Larrea C. 2005. Interaction between KIR3DL1 and HLA-B*57 supertype alleles influences the progression of HIV-1 infection in a Zambian population. Hum. Immunol 66:285–289. [DOI] [PubMed] [Google Scholar]
- 89.Martin MP, Gao X, Lee J-H, Nelson GW, Detels R, Goedert JJ, Buchbinder S, Hoots K, Vlahov D, Trowsdale J, Wilson M, O’Brien SJ, Carrington M. 2002. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat. Genet 31:429–434. [DOI] [PubMed] [Google Scholar]
- 90.Martin MP, Qi Y, Gao X, Yamada E, Martin JN, Pereyra F, Colombo S, Brown EE, Shupert WL, Phair J, Goedert JJ, Buchbinder S, Kirk GD, Telenti A, Connors M, O’Brien SJ, Walker BD, Parham P, Deeks SG, McVicar DW, Carrington M. 2007. Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat. Genet 39:733–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Körner C, Altfeld M. 2012. Role of KIR3DS1 in human diseases. Front. Immunol 3:326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Flores-Villanueva PO, Yunis EJ, Delgado JC, Vittinghoff E, Buchbinder S, Leung JY, Uglialoro AM, Clavijo OP, Rosenberg ES, Kalams SA, Braun JD, Boswell SL, Walker BD, Goldfeld AE. 2001. Control of HIV-1 viremia and protection from AIDS are associated with HLA-Bw4 homozygosity. Proc. Natl. Acad. Sci. U. S. A 98:5140–5145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.O’Connor GM, Vivian JP, Gostick E, Pymm P, Lafont BAP, Price DA, Rossjohn J, Brooks AG, McVicar DW. 2015. Peptide-dependent recognition of HLA-B* 57: 01 by KIR3DS1. J. Virol 89:5213–5221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Garcia-Beltran WF, Hölzemer A, Martrus G, Chung AW, Pacheco Y, Simoneau CR, Rucevic M, Lamothe-Molina PA, Pertel T, Kim T-E, Dugan H, Alter G, Dechanet-Merville J, Jost S, Carrington M, Altfeld M. 2016. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat. Immunol 17:1067–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Boudreau JE, Mulrooney TJ, Luduec J-B, Barker E, Hsu KC. 2016. KIR3DL1 and HLA-B Density and Binding Calibrate NK Education and Response to HIV. J. Immunol 196:3398–3410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Shimoda Y, Tajima Y, Nagase T, Harii K, Osumi N, Sanai Y. 1999. Cloning and expression of a novel galactoside beta1, 3-glucuronyltransferase involved in the biosynthesis of HNK-1 epitope. J. Biol. Chem 274:17115–17122. [DOI] [PubMed] [Google Scholar]
- 97.Lopez-Vergès S, Milush JM, Pandey S, York VA, Arakawa-Hoyt J, Pircher H, Norris PJ, Nixon DF, Lanier LL. 2010. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 116:3865–3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Focosi D, Bestagno M, Burrone O, Petrini M. 2010. CD57+ T lymphocytes and functional immune deficiency. J. Leukoc. Biol 87:107–116. [DOI] [PubMed] [Google Scholar]
- 99.Brenchley JM, Karandikar NJ, Betts MR, Ambrozak DR, Hill BJ, Crotty LE, Casazza JP, Kuruppu J, Migueles SA, Connors M, Roederer M, Douek DC, Koup RA. 2003. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood. 101:2711–2720. [DOI] [PubMed] [Google Scholar]
- 100.Nagler A, Lanier LL, Cwirla S, Phillips JH. 1989. Comparative studies of human FcRIII-positive and negative natural killer cells. J. Immunol 143:3183–3191. [PubMed] [Google Scholar]
- 101.Merino J, Martínez-González MA, Rubio M, Inogés S, Sánchez-Ibarrola A, Subirá ML. 1998. Progressive decrease of CD8high+ CD28+ CD57- cells with ageing. Clin. Exp. Immunol 112:48–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chattopadhyay PK, Betts MR, Price DA, Gostick E, Horton H, Roederer M, De Rosa SC. 2009. The cytolytic enzymes granyzme A, granzyme B, and perforin: expression patterns, cell distribution, and their relationship to cell maturity and bright CD57 expression. J. Leukoc. Biol 85:88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Yu J, Mao HC, Wei M, Hughes T, Zhang J, Park I-K, Liu S, McClory S, Marcucci G, Trotta R, Others. 2010. CD94 surface density identifies a functional intermediary between the CD56 bright and CD56 dim human NK-cell subsets. Blood. 115:274–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Juelke K, Killig M, Luetke-Eversloh M, Parente E, Gruen J, Morandi B, Ferlazzo G, Thiel A, Schmitt-Knosalla I, Romagnani C. 2010. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood. 116:1299–1307. [DOI] [PubMed] [Google Scholar]
- 105.Cichocki F, Miller JS, Anderson SK, Bryceson YT. 2013. Epigenetic regulation of NK cell differentiation and effector functions. Front. Immunol 4:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Eller MA, Currier JR. 2012. OMIP-007: Phenotypic analysis of human natural killer cells. Cytometry A. 81:447–449. [DOI] [PubMed] [Google Scholar]
- 107.Béziat V, Dalgard O, Asselah T, Halfon P, Bedossa P, Boudifa A, Hervier B, Theodorou I, Martinot M, Debré P, Björkström NK, Malmberg K-J, Marcellin P, Vieillard V. 2012. CMV drives clonal expansion of NKG2C+ NK cells expressing self-specific KIRs in chronic hepatitis patients. Eur. J. Immunol 42:447–457. [DOI] [PubMed] [Google Scholar]
- 108.Goodier MR, White MJ, Darboe A, Nielsen CM, Goncalves A, Bottomley C, Moore SE, Riley EM. 2014. Rapid NK cell differentiation in a population with near-universal human cytomegalovirus infection is attenuated by NKG2C deletions. Blood. 124:2213–2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hatton O, Strauss-Albee DM, Zhao NQ, Haggadone MD, Pelpola JS, Krams SM, Martinez OM, Blish CA. 2016. NKG2A-Expressing Natural Killer Cells Dominate the Response to Autologous Lymphoblastoid Cells Infected with Epstein-Barr Virus. Front. Immunol 7:607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Lünemann A, Vanoaica LD, Azzi T, Nadal D, Münz C. 2013. A Distinct Subpopulation of Human NK Cells Restricts B Cell Transformation by EBV. The Journal of Immunology. 191:4989–4995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lazetic S, Chang C, Houchins JP, Lanier LL, Phillips JH. 1996. Human natural killer cell receptors involved in MHC class I recognition are disulfide-linked heterodimers of CD94 and NKG2 subunits. J. Immunol 157:4741–4745. [PubMed] [Google Scholar]
- 112.Borrego F, Ulbrecht M, Weiss EH, Coligan JE, Brooks AG. 1998. Recognition of human histocompatibility leukocyte antigen (HLA)-E complexed with HLA class I signal sequence--derived peptides by CD94/NKG2 confers protection from natural killer cell--mediated lysis. J. Exp. Med 187:813–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lanier LL, Corliss B, Wu J, Phillips JH. 1998. Association of DAP12 with activating CD94/NKG2C NK cell receptors. Immunity. 8:693–701. [DOI] [PubMed] [Google Scholar]
- 114.Palmieri G, Tullio V, Zingoni A, Piccoli M, Frati L, Lopez-Botet M, Santoni A. 1999. CD94/NKG2-A inhibitory complex blocks CD16-triggered Syk and extracellular regulated kinase activation, leading to cytotoxic function of human NK cells. J. Immunol 162:7181–7188. [PubMed] [Google Scholar]
- 115.Carretero M, Cantoni C, Bellón T, Bottino C, Biassoni R, Rodríguez A, Pérez-Villar JJ, Moretta L, Moretta A, López-Botet M. 1997. The CD94 and NKG2-AC-type lectins covalently assemble to form a natural killer cell inhibitory receptor for HLA class I molecules. Eur. J. Immunol 27:563–567. [DOI] [PubMed] [Google Scholar]
- 116.Michaëlsson J, Teixeira de Matos C, Achour A, Lanier LL, Kärre K, Söderström K. 2002. A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/NKG2A recognition. J. Exp. Med 196:1403–1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Béziat V, Descours B, Parizot C, Debré P, Vieillard V. 2010. NK cell terminal differentiation: correlated stepwise decrease of NKG2A and acquisition of KIRs. PLoS One. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. 1998. HLA-E surface expression depends on binding of TAP-dependent peptides derived from certain HLA class I signal sequences. J. Immunol 160:4951–4960. [PubMed] [Google Scholar]
- 119.Björkström NK, Riese P, Heuts F, Andersson S, Fauriat C, Ivarsson MA, Björklund AT, Flodström-Tullberg M, Michaëlsson J, Rottenberg ME, Guzmán CA, Ljunggren H-G, Malmberg K-J. 2010. Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. Blood. 116:3853–3864. [DOI] [PubMed] [Google Scholar]
- 120.Morvan MG, Lanier LL. 2016. NK cells and cancer: you can teach innate cells new tricks. Nat. Rev. Cancer 16:7–19. [DOI] [PubMed] [Google Scholar]
- 121.Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T. 1999. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 285:727–729. [DOI] [PubMed] [Google Scholar]
- 122.McFarland BJ, Strong RK. 2003. Thermodynamic analysis of degenerate recognition by the NKG2D immunoreceptor: not induced fit but rigid adaptation. Immunity. 19:803–812. [DOI] [PubMed] [Google Scholar]
- 123.Jolly C, Morimoto RI. 2000. Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J. Natl. Cancer Inst 92:1564–1572. [DOI] [PubMed] [Google Scholar]
- 124.Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. 1996. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc. Natl. Acad. Sci. U. S. A 93:12445–12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. 1999. Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc. Natl. Acad. Sci. U. S. A 96:6879–6884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Jinushi M, Takehara T, Tatsumi T, Kanto T, Groh V, Spies T, Kimura R, Miyagi T, Mochizuki K, Sasaki Y, Hayashi N. 2003. Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int. J. Cancer 104:354–361. [DOI] [PubMed] [Google Scholar]
- 127.Diefenbach A, Jensen ER, Jamieson AM, Raulet DH. 2001. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature. 413:165–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Guerra N, Tan YX, Joncker NT, Choy A, Gallardo F, Xiong N, Knoblaugh S, Cado D, Greenberg NM, Greenberg NR, Raulet DH. 2008. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity. 28:571–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Westwood JA, Kelly JM, Tanner JE, Kershaw MH, Smyth MJ, Hayakawa Y. 2004. Cutting edge: novel priming of tumor-specific immunity by NKG2D-triggered NK cell-mediated tumor rejection and Th1-independent CD4+ T cell pathway. J. Immunol 172:757–761. [DOI] [PubMed] [Google Scholar]
- 130.Zhang T, Scott JM, Hwang I, Kim S. 2013. Cutting Edge: Antibody-Dependent Memory-like NK Cells Distinguished by FcR Deficiency. The Journal of Immunology. 190:1402–1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lee J, Zhang T, Hwang I, Kim A, Nitschke L, Kim M, Scott JM, Kamimura Y, Lanier LL, Kim S. 2015. Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity. 42:431–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Schlums H, Cichocki F, Tesi B, Theorell J, Beziat V, Holmes TD, Han H, Chiang SCC, Foley B, Mattsson K, Larsson S, Schaffer M, Malmberg K-JJ, Ljunggren H-GG, Miller JS, Bryceson YT. 2015. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity. 42:443–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tesi B, Schlums H, Cichocki F, Bryceson YT. 2016. Epigenetic Regulation of Adaptive NK Cell Diversification. Trends Immunol. 37:451–461. [DOI] [PubMed] [Google Scholar]
- 134.Kuijpers TW, Baars PA, Dantin C, van den Burg M, van Lier RAW, Roosnek E. 2008. Human NK cells can control CMV infection in the absence of T cells. Blood. 112:914–915. [DOI] [PubMed] [Google Scholar]
- 135.Della Chiesa M, Falco M, Podestà M, Locatelli F, Moretta L, Frassoni F, Moretta A. 2012. Phenotypic and functional heterogeneity of human NK cells developing after umbilical cord blood transplantation: a role for human cytomegalovirus? Blood. 119:399–410. [DOI] [PubMed] [Google Scholar]
- 136.Foley B, Cooley S, Verneris MR, Curtsinger J, Luo X, Waller EK, Anasetti C, Weisdorf D, Miller JS. 2012. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J. Immunol 189:5082–5088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Achour A, Baychelier F, Besson C, Arnoux A, Marty M, Hannoun L, Samuel D, Debré P, Vieillard V, K-GREF Study Group. 2014. Expansion of CMV-mediated NKG2C+ NK cells associates with the development of specific de novo malignancies in liver-transplanted patients. J. Immunol 192:503–511. [DOI] [PubMed] [Google Scholar]
- 138.Björkström NK, Lindgren T, Stoltz M, Fauriat C, Braun M, Evander M, Michaëlsson J, Malmberg K-J, Klingström J, Ahlm C, Ljunggren H-G. 2011. Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus. J. Exp. Med 208:13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Gumá M, Cabrera C, Erkizia I, Bofill M, Clotet B, Ruiz L, López-Botet M. 2006. Human Cytomegalovirus Infection Is Associated with Increased Proportions of NK Cells That Express the CD94/NKG2C Receptor in Aviremic HIV-1–Positive Patients. J. Infect. Dis 194:38–41. [DOI] [PubMed] [Google Scholar]
- 140.Ahlenstiel G, Titerence RH, Koh C, Edlich B, Feld JJ, Rotman Y, Ghany MG, Hoofnagle JH, Liang TJ, Heller T, Others. 2010. Natural killer cells are polarized toward cytotoxicity in chronic hepatitis C in an interferon-alfa--dependent manner. Gastroenterology. 138:325–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Oliviero B, Varchetta S, Paudice E, Michelone G, Zaramella M, Mavilio D, De Filippi F, Bruno S, Mondelli MU. 2009. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology. 137:1151–60, 1160.e1–7. [DOI] [PubMed] [Google Scholar]
- 142.Della Chiesa M, Sivori S, Carlomagno S, Moretta L, Moretta A. 2015. Activating KIRs and NKG2C in Viral Infections: Toward NK Cell Memory? Front. Immunol 6:573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Djaoud Z, Guethlein LA, Horowitz A, Azzi T, Nemat-Gorgani N, Olive D, Nadal D, Norman PJ, Münz C, Parham P. 2017. Two alternate strategies for innate immunity to Epstein-Barr virus: One using NK cells and the other NK cells and γδ T cells. J. Exp. Med 214:1827–1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chijioke O, Landtwing V, Münz C. 2016. NK Cell Influence on the Outcome of Primary Epstein-Barr Virus Infection. Front. Immunol 7:323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Chijioke O, Müller A, Feederle R, Barros MHM, Krieg C, Emmel V, Marcenaro E, Leung CS, Antsiferova O, Landtwing V, Bossart W, Moretta A, Hassan R, Boyman O, Niedobitek G, Delecluse H-J, Capaul R, Münz C. 2013. Human natural killer cells prevent infectious mononucleosis features by targeting lytic Epstein-Barr virus infection. Cell Rep. 5:1489–1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lisovsky I, Isitman G, Song R, DaFonseca S, Tremblay-McLean A, Lebouché B, Routy J-P, Bruneau J, Bernard NF. 2015. A Higher Frequency of NKG2A+ than of NKG2A- NK Cells Responds to Autologous HIV-Infected CD4 Cells irrespective of Whether or Not They Coexpress KIR3DL1. J. Virol 89:9909–9919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Davis ZB, Cogswell A, Scott H, Mertsching A, Boucau J, Wambua D, Gall S, Planelles V, Campbell KS, Barker E. 2016. A Conserved HIV-1-Derived Peptide Presented by HLA-E Renders Infected T-cells Highly Susceptible to Attack by NKG2A/CD94-Bearing Natural Killer Cells. PLoS Pathog. 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Granzin M, Soltenborn S, Müller S, Kollet J, Berg M, Cerwenka A, Childs RW, Huppert V. 2015. Fully automated expansion and activation of clinical-grade natural killer cells for adoptive immunotherapy. Cytotherapy. 17:621–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Stegmann KA, Björkström NK, Ciesek S, Lunemann S, Jaroszewicz J, Wiegand J, Malinski P, Dustin LB, Rice CM, Manns MP, Pietschmann T, Cornberg M, Ljunggren H-G, Wedemeyer H. 2012. Interferon α-stimulated natural killer cells from patients with acute hepatitis C virus (HCV) infection recognize HCV-infected and uninfected hepatoma cells via DNAX accessory molecule-1. J. Infect. Dis 205:1351–1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Sene D, Levasseur F, Abel M, Lambert M, Camous X, Hernandez C, Pene V, Rosenberg AR, Jouvin-Marche E, Marche PN, Others. 2010. Hepatitis C virus (HCV) evades NKG2D-dependent NK cell responses through NS5A-mediated imbalance of inflammatory cytokines. PLoS Pathog. 6:e1001184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. 2011. Activating and inhibitory receptors of natural killer cells. Immunol. Cell Biol 89:216–224. [DOI] [PubMed] [Google Scholar]
- 152.Fuchs A, Cella M, Kondo T, Colonna M. 2005. Paradoxic inhibition of human natural interferon-producing cells by the activating receptor NKp44. Blood. 106:2076–2082. [DOI] [PubMed] [Google Scholar]
- 153.Vitale M, Falco M, Castriconi R, Parolini S, Zambello R, Semenzato G, Biassoni R, Bottino C, Moretta L, Moretta A. 2001. Identification of NKp80, a novel triggering molecule expressed by human NK cells. Eur. J. Immunol 31:233–242. [DOI] [PubMed] [Google Scholar]
- 154.De Maria A, Fogli M, Costa P, Murdaca G, Puppo F, Mavilio D, Moretta A, Moretta L. 2003. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKp46, NKp30 and NKp44). Eur. J. Immunol 33:2410–2418. [DOI] [PubMed] [Google Scholar]
- 155.Bisio F, Bozzano F, Marras F, Di Biagio A, Moretta L, De Maria A. 2013. Successfully treated HIV-infected patients have differential expression of NK cell receptors (NKp46 and NKp30) according to AIDS status at presentation. Immunol. Lett 152:16–24. [DOI] [PubMed] [Google Scholar]
- 156.Sivori S, Pende D, Bottino C, Marcenaro E, Pessino A, Biassoni R, Moretta L, Moretta A. 1999. NKp46 is the major triggering receptor involved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation between surface density of NKp46 and natural cytotoxicity against autologous, allogeneic or xenogeneic target cells. Eur. J. Immunol 29:1656–1666. [DOI] [PubMed] [Google Scholar]
- 157.Halfteck GG, Elboim M, Gur C, Achdout H, Ghadially H, Mandelboim O. 2009. Enhanced in vivo growth of lymphoma tumors in the absence of the NK-activating receptor NKp46/NCR1. J. Immunol 182:2221–2230. [DOI] [PubMed] [Google Scholar]
- 158.Gazit R, Gruda R, Elboim M, Arnon TI, Katz G, Achdout H, Hanna J, Qimron U, Landau G, Greenbaum E, Zakay-Rones Z, Porgador A, Mandelboim O. 2006. Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. Nat. Immunol 7:517–523. [DOI] [PubMed] [Google Scholar]
- 159.Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A, Mahmood S, Gut M, Heath SC, Estellé J, Bertosio E, Vely F, Gastinel LN, Beutler B, Malissen B, Malissen M, Gut IG, Vivier E, Ugolini S. 2012. Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science. 335:344–348. [DOI] [PubMed] [Google Scholar]
- 160.Mendelson M, Tekoah Y, Zilka A, Gershoni-Yahalom O, Gazit R, Achdout H, Bovin NV, Meningher T, Mandelboim M, Mandelboim O, David A, Porgador A. 2010. NKp46 O-glycan sequences that are involved in the interaction with hemagglutinin type 1 of influenza virus. J. Virol 84:3789–3797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Draghi M, Pashine A, Sanjanwala B, Gendzekhadze K, Cantoni C, Cosman D, Moretta A, Valiante NM, Parham P. 2007. NKp46 and NKG2D recognition of infected dendritic cells is necessary for NK cell activation in the human response to influenza infection. J. Immunol 178:2688–2698. [DOI] [PubMed] [Google Scholar]
- 162.Bar-On Y, Glasner A, Meningher T, Achdout H, Gur C, Lankry D, Vitenshtein A, Meyers AFA, Mandelboim M, Mandelboim O. 2013. Neuraminidase-mediated, NKp46-dependent immune-evasion mechanism of influenza viruses. Cell Rep. 3:1044–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Björkström NK, Ljunggren H-G, Sandberg JK. 2010. CD56 negative NK cells: origin, function, and role in chronic viral disease. Trends Immunol. 31:401–406. [DOI] [PubMed] [Google Scholar]
- 164.Eller MA, Eller LA, Ouma BJ, Thelian D, Gonzalez VD, Guwatudde D, McCutchan FE, Marovich MA, Michael NL, de Souza MS, Wabwire-Mangen F, Robb ML, Currier JR, Sandberg JK. 2009. Elevated natural killer cell activity despite altered functional and phenotypic profile in Ugandans with HIV-1 clade A or clade D infection. J. Acquir. Immune Defic. Syndr 51:380–389. [DOI] [PubMed] [Google Scholar]
- 165.Mavilio D, Lombardo G, Benjamin J, Kim D, Follman D, Marcenaro E, O’Shea MA, Kinter A, Kovacs C, Moretta A, Fauci AS. 2005. Characterization of CD56-/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proceedings of the National Academy of Sciences. 102:2886–2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Mavilio D, Benjamin J, Daucher M, Lombardo G, Kottilil S, Planta MA, Marcenaro E, Bottino C, Moretta L, Moretta A, Fauci AS. 2003. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc. Natl. Acad. Sci. U. S. A 100:15011–15016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Milush JM, López-Vergès S, York VA, Deeks SG, Martin JN, Hecht FM, Lanier LL, Nixon DF. 2013. CD56negCD16+ NK cells are activated mature NK cells with impaired effector function during HIV-1 infection. Retrovirology. 10:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Brown MH, Boles K, van der Merwe PA, Kumar V, Mathew PA, Barclay AN. 1998. 2B4, the natural killer and T cell immunoglobulin superfamily surface protein, is a ligand for CD48. J. Exp. Med 188:2083–2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Garni-Wagner BA, Purohit A, Mathew PA, Bennett M, Kumar V. 1993. A novel function-associated molecule related to non-MHC-restricted cytotoxicity mediated by activated natural killer cells and T cells. J. Immunol 151:60–70. [PubMed] [Google Scholar]
- 170.Bryceson YT, March ME, Ljunggren H-G, Long EO. 2006. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol. Rev 214:73–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Vaidya SV, Stepp SE, McNerney ME, Lee J-K, Bennett M, Lee K-M, Stewart CL, Kumar V, Mathew PA. 2005. Targeted disruption of the 2B4 gene in mice reveals an in vivo role of 2B4 (CD244) in the rejection of B16 melanoma cells. J. Immunol 174:800–807. [DOI] [PubMed] [Google Scholar]
- 172.Lee K-M, McNerney ME, Stepp SE, Mathew PA, Schatzle JD, Bennett M, Kumar V. 2004. 2B4 acts as a non-major histocompatibility complex binding inhibitory receptor on mouse natural killer cells. J. Exp. Med 199:1245–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Sivori S, Falco M, Marcenaro E, Parolini S, Biassoni R, Bottino C, Moretta L, Moretta A. 2002. Early expression of triggering receptors and regulatory role of 2B4 in human natural killer cell precursors undergoing in vitro differentiation. Proc. Natl. Acad. Sci. U. S. A 99:4526–4531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Falschlehner C, Schaefer U, Walczak H. 2009. Following TRAIL’s path in the immune system. Immunology. 127:145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Takeda K, Smyth MJ, Cretney E, Hayakawa Y, Yamaguchi N, Yagita H, Okumura K. 2001. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in NK cell-mediated and IFN-gamma-dependent suppression of subcutaneous tumor growth. Cell. Immunol 214:194–200. [DOI] [PubMed] [Google Scholar]
- 176.Sato K, Hida S, Takayanagi H, Yokochi T, Kayagaki N, Takeda K, Yagita H, Okumura K, Tanaka N, Taniguchi T, Ogasawara K. 2001. Antiviral response by natural killer cells through TRAIL gene induction by IFN–α/β. Eur. J. Immunol 31:3138–3146. [DOI] [PubMed] [Google Scholar]
- 177.Sedger LM, Shows DM, Blanton RA, Peschon JJ, Goodwin RG, Cosman D, Wiley SR. 1999. IFN-gamma mediates a novel antiviral activity through dynamic modulation of TRAIL and TRAIL receptor expression. J. Immunol 163:920–926. [PubMed] [Google Scholar]
- 178.Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. 2003. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J. Exp. Med 198:557–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, Kitamura T, Nicholl J, Sutherland GR, Lanier LL, Phillips JH. 1996. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity. 4:573–581. [DOI] [PubMed] [Google Scholar]
- 180.Masson D, Jarry A, Baury B, Blanchardie P, Laboisse C, Lustenberger P, Denis MG. 2001. Overexpression of the CD155 gene in human colorectal carcinoma. Gut. 49:236–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Magri G, Muntasell A, Romo N, Sáez-Borderías A, Pende D, Geraghty DE, Hengel H, Angulo A, Moretta A, López-Botet M. 2011. NKp46 and DNAM-1 NK-cell receptors drive the response to human cytomegalovirus-infected myeloid dendritic cells overcoming viral immune evasion strategies. Blood. 117:848–856. [DOI] [PubMed] [Google Scholar]
- 182.Zingoni A, Ardolino M, Santoni A, Cerboni C. 2012. NKG2D and DNAM-1 activating receptors and their ligands in NK-T cell interactions: role in the NK cell-mediated negative regulation of T cell responses. Front. Immunol 3:408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Matusali G, Potestà M, Santoni A, Cerboni C, Doria M. 2012. The human immunodeficiency virus type 1 Nef and Vpu proteins downregulate the natural killer cell-activating ligand PVR. J. Virol 86:4496–4504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Cosman D, Fanger N, Borges L, Kubin M, Chin W, Peterson L, Hsu ML. 1997. A novel immunoglobulin superfamily receptor for cellular and viral MHC class I molecules. Immunity. 7:273–282. [DOI] [PubMed] [Google Scholar]
- 185.Prod’homme V, Griffin C, Aicheler RJ, Wang ECY, McSharry BP, Rickards CR, Stanton RJ, Borysiewicz LK, López-Botet M, Wilkinson GWG, Tomasec P. 2007. The human cytomegalovirus MHC class I homolog UL18 inhibits LIR-1+ but activates LIR-1- NK cells. J. Immunol 178:4473–4481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Navarro F, Llano M, Bellón T, Colonna M, Geraghty DE, López-Botet M. 1999. The ILT2(LIR1) and CD94/NKG2A NK cell receptors respectively recognize HLA-G1 and HLA-E molecules co-expressed on target cells. Eur. J. Immunol 29:277–283. [DOI] [PubMed] [Google Scholar]
- 187.Scott-Algara D, Arnold V, Didier C, Kattan T, Pirozzi G, Barré-Sinoussi F, Pancino G. 2008. The CD85j+ NK cell subset potently controls HIV-1 replication in autologous dendritic cells. PLoS One. 3:e1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Strauss-Albee DM, Fukuyama J, Liang EC, Yao Y, Jarrell JA, Drake AL, Kinuthia J, Montgomery RR, John-Stewart G, Holmes S, Blish CA. 2015. Human NK cell repertoire diversity reflects immune experience and correlates with viral susceptibility. Sci. Transl. Med 7:297ra115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Chang S, Aune TM. 2005. Histone hyperacetylated domains across the Ifng gene region in natural killer cells and T cells. Proc. Natl. Acad. Sci. U. S. A 102:17095–17100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Araki Y, Fann M, Wersto R, Weng N-P. 2008. Histone acetylation facilitates rapid and robust memory CD8 T cell response through differential expression of effector molecules (eomesodermin and its targets: perforin and granzyme B). J. Immunol 180:8102–8108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Khavari PA, Peterson CL, Tamkun JW, Mendel DB, Crabtree GR. 1993. BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature. 366:170–174. [DOI] [PubMed] [Google Scholar]
- 192.Zhang F, Boothby M. 2006. T helper type 1-specific Brg1 recruitment and remodeling of nucleosomes positioned at the IFN-gamma promoter are Stat4 dependent. J. Exp. Med 203:1493–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Marcus A, Raulet DH. 2013. Evidence for natural killer cell memory. Curr. Biol 23:R817–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Strauss-Albee DM, Liang EC, Ranganath T, Aziz N, Blish CA. 2017. The newborn human NK cell repertoire is phenotypically formed but functionally reduced. Cytometry B Clin. Cytom 92:33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Jost S, Altfeld M. 2013. Control of human viral infections by natural killer cells. Annu. Rev. Immunol 31:163–194. [DOI] [PubMed] [Google Scholar]
- 196.Cichocki F, Schlums H, Theorell J, Tesi B, Miller JS, Ljunggren H-G, Bryceson YT. 2015. Diversification and Functional Specialization of Human NK Cell Subsets Natural Killer Cells. Springer, Cham, pp 63–93. [DOI] [PubMed] [Google Scholar]
- 197.Min-Oo G, Lanier LL. 2014. Cytomegalovirus generates long-lived antigen-specific NK cells with diminished bystander activation to heterologous infection. J. Exp. Med 211:2669–2680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Strauss-Albee DM, Horowitz A, Parham P, Blish CA. 2014. Coordinated regulation of NK receptor expression in the maturing human immune system. J. Immunol 193:4871–4879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Vendrame E, Fukuyama J, Strauss-Albee DM, Holmes S, Blish CA. 2017. Mass Cytometry Analytical Approaches Reveal Cytokine-Induced Changes in Natural Killer Cells. Cytometry B Clin. Cytom 92:57–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Chattopadhyay PK, Gierahn TM, Roederer M, Love JC. 2014. Single-cell technologies for monitoring immune systems. Nat. Immunol 15:128–135. [DOI] [PMC free article] [PubMed] [Google Scholar]