Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2021 Jun 11;22(2):112–123. doi: 10.1038/s41577-021-00558-3

Natural killer cells in antiviral immunity

Niklas K Björkström 1,, Benedikt Strunz 1, Hans-Gustaf Ljunggren 1
PMCID: PMC8194386  PMID: 34117484

Abstract

Natural killer (NK) cells play an important role in innate immune responses to viral infections. Here, we review recent insights into the role of NK cells in viral infections, with particular emphasis on human studies. We first discuss NK cells in the context of acute viral infections, with flavivirus and influenza virus infections as examples. Questions related to activation of NK cells, homing to infected tissues and the role of tissue-resident NK cells in acute viral infections are also addressed. Next, we discuss NK cells in the context of chronic viral infections with hepatitis C virus and HIV-1. Also covered is the role of adaptive-like NK cell expansions as well as the appearance of CD56 NK cells in the course of chronic infection. Specific emphasis is then placed in viral infections in patients with primary immunodeficiencies affecting NK cells. Not least, studies in this area have revealed an important role for NK cells in controlling several herpesvirus infections. Finally, we address new data with respect to the activation of NK cells and NK cell function in humans infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) giving rise to coronavirus disease 2019 (COVID-19).

Subject terms: Innate lymphoid cells, Infection


This Review covers the role of natural killer cells in acute and chronic viral infections, with emphasis on human infections and insights from patients with primary immunodeficiencies affecting natural killer cells. It also describes the emerging data on how natural killer cells are involved in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.

Introduction

Almost 50 years ago, a small number of research groups started to observe unexpected spontaneous cytotoxic activities among lymphocytes. While these were at first considered to be merely annoying background phenomena, subsequent studies led to the realization that a new subpopulation of lymphocytes was responsible for this activity1. The cells were termed ‘natural killer (NK) cells’. Over the years, insights into the molecular specificity of NK cells in terms of target cell recognition24 and into their role in controlling viral and intracellular bacterial infections and tumours, as well as insights into their ability to regulate other immune cells, have put NK cells at the forefront of modern immunology5. Today, they are used as therapeutic agents in the treatment of human malignant diseases6, and strategies for using them therapeutically against viral diseases, including coronavirus disease 2019 (COVID-19), are currently being explored7.

NK cells are best characterized by their cytotoxic function and their ability to produce cytokines upon stimulation5. With respect to their cytotoxic potential, they target infected, transformed and stressed cells. In this way, they not only eliminate unwanted cells but also contribute to cellular homeostasis8. The conventional known anatomical site of NK cell production is the bone marrow, where interactions with other cellular components, cytokines and soluble molecules support and drive NK cell development (reviewed in ref.9) (Box 1). Most of the current knowledge of human NK cells has arisen from studies of NK cells in peripheral blood and secondary lymphoid organs. Recently, however, there has been increased interest in NK cells residing in specific tissues10 (so-called tissue-resident NK cells) (Box 2). In humans, mature NK cells are traditionally identified as CD3CD56+ lymphocytes. They were long thought to represent a homogeneous population of cells. However, in line with adaptive lymphocytes, we now know that NK cells undergo a well-defined differentiation process involving distinct phenotypic and functional changes (Box 1).

NK cell function is regulated by a large number of germline-encoded receptors11,12. Upon viral infection, host cells may become susceptible to NK cell-mediated recognition through a variety of mechanisms that may include upregulation of self-encoded molecules induced by the infection and/or a concomitant cellular stress response that binds activating NK cell receptors such as natural cytotoxicity receptors (NKp30, NKp44 and NKp46)13, C-type lectin-like receptors (for example, NKG2D and NKp80)14 and co-activating receptors (for example, DNAM1 and CD2)12. In parallel, contributing to increased target cell susceptibility is the often observed downregulation of MHC class I ligands for inhibitory receptors15. Importantly, NK cells can also eliminate virus-infected cells via CD16-mediated antibody-dependent cellular cytotoxicity (ADCC)16. Finally, NK cell activity is modulated by cytokines, including, but not limited to, the activating cytokines IL-2, IL-12, IL-15, IL-18 and type I interferons, which can be produced by virally infected cells or activated antigen-presenting cells17. Many of these cytokines, alone or in combination, promote NK cell survival, proliferation, cytotoxicity and cytokine production, including interferon-γ (IFNγ) production18. Through these mechanisms, NK cells are uniquely equipped to sense and quickly respond to viral infections7. In this respect, they often act in concert with other host immune responses in mediating antiviral immunity.

Here, we review recently obtained knowledge with regard to human NK cells in antiviral immunity. We address NK cell responses to typical acute viral infections. In many situations, these trigger activation of circulating NK cells and their homing to affected tissues. In parallel, tissue-resident NK cells may also become activated. With respect to chronic viral infections, NK cells contribute to viral control. We describe how such infections may lead to ‘adaptations’ in the NK cell repertoire, likely contributing to their maintenance of viral control but possibly also representing a consequence of exhaustion in some cases. Impairment in NK cell-mediated viral control is seen in patients with inborn deficiencies affecting NK cell development and/or function. Finally, we pay specific attention to the very recently developing knowledge with respect to vigorous NK cell activation in human severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19.

Box 1 Human NK cell differentiation.

Natural killer (NK) cells undergo the final steps of their development in secondary lymphoid organs, resulting in mature CD56brightCD16- NK cells that enter the circulation146. CD56bright NK cells are efficient in producing cytokines but have limited cytotoxicity. They can further differentiate into CD56dimCD16+ NK cells, the NK cell population most dominant in peripheral blood. Compared with CD56bright NK cells, peripheral blood CD56dim NK cells are a highly diverse population of immune cells93 that undergo a continuous IL-15-driven differentiation process during their lifespan34. This differentiation is associated with changes in cell-surface phenotype, including loss of expression of CD94, NKG2A and CD62L (also known as L-selectin), as well as induction of killer cell immunoglobulin-like receptor (KIR) and CD57 expression34,36,147,148. On a functional level, less differentiated CD56dim NK cells are more highly proliferative and more responsive to cytokine stimulation, whereas highly differentiated CD56dim NK cells lose their proliferative capacity and instead become more specialized with regard to cellular cytotoxicity and antibody-dependent cellular cytotoxicity34. A parallel pathway of NK cell differentiation occurs in non-lymphoid peripheral tissues (reviewed in ref.10) (Box 2). Here, it is instead the less differentiated CD56bright NK cells that are thought to enter tissue parenchyma and, in response to the local microenvironment, likely differentiate into site-adapted cells50. Understanding NK cell differentiation, the factors driving it and the functional specialization of the discrete differentiation stages provides a framework for subsequently interpreting NK cell antiviral immunity.

Box 2 Human tissue-resident NK cells.

Over the last decade, our knowledge of the presence, development, differentiation and function of natural killer (NK) cells residing in peripheral tissues has increased dramatically (reviewed in ref.10). It is now clear that most human organs, both lymphoid and non-lymphoid, contain tissue-resident NK cells expressing canonical tissue-residency markers such as CD69, CD103 and CD49a, all of which are functionally involved in retaining the cells in tissues50,149151 and have been shown to, de facto, associate with tissue residency in human solid organ transplantation studies152,153. The ontogeny of tissue-resident NK cells remains elusive. However, since their phenotype is in many respects (except for uterine NK cells) similar to that of blood CD56bright NK cells, it is possible that they originate from CD56bright NK cells that get cues to enter tissue microenvironments10. Alternatively, dedicated precursors may exist that seed peripheral tissues154. With their phenotype more resembling less mature CD56bright NK cells, tissue-resident NK cells are in general less cytotoxic but more efficient in producing cytokines such as interferon-γ. While many human peripheral tissues contain a sizeable population of tissue-resident NK cells, mouse peripheral tissues such as the liver, salivary glands and uterus are instead populated by NK1.1+CD49a+DX5 cells referred to as group 1 innate lymphoid cells (ILC1s). Yet, ILC1s likely represent the mouse counterpart of human tissue-resident NK cells155.

NK cells in acute viral infections

NK cells respond vigorously to several acute viral infections, exemplified in the following subsections by acute infections with flavivirus and influenza virus (both RNA viruses). In many situations, this triggers activation of circulating NK cells and their homing to affected tissues. In parallel, specific tissue-resident NK cells may also become activated. Critical to all host responses to acute viral infections is the balance between possible acute antiviral mechanisms and responses contributing to tissue damage and immunopathology.

Flavivirus infections

Flaviviruses, including dengue virus (DENV), West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus, yellow fever virus and Zika virus, are major emerging human pathogens, affecting millions of individuals worldwide. Flaviviruses elicit haemorrhagic fever-like syndromes and central nervous system-associated diseases in infected hosts19. Effective vaccines exist for some of these viral infections. With a few exceptions, there is a lack of knowledge with respect to which host cells are infected by flaviviruses in humans and whether they are targets of NK cell-mediated responses. Innate immune responses elicited by flaviviruses have been studied extensively in animal models, in particular DENV and Zika virus infections20. However, fewer studies have focused specifically on NK cells in these acute viral infections, with influenza virus infections being an exception, as reviewed in the next subsection. In humans, NK cell responses to acute flavivirus infection have been addressed in clinical studies, particularly in DENV infections21,22. While clinical studies performed to date have differed in their set-up, some common observations have been made with respect to NK cell responses in different flavivirus infections. In several studies, both CD56bright NK cells and, in some studies, subpopulations of less differentiated CD56dim NK cells (Box 1) have been reported to respond vigorously to infection2225. Type I and type III interferons likely contribute to the NK cell responses observed in several flavivirus infections, including yellow fever virus infection23. Responses occur irrespective of NK cell education, and no evidence of specific adaptive-like NK cell responses has been observed22,23. Concomitant with the increase in NK cell activation, increased levels of various cytokines, including IL-12, IL-15, IL-18, IFNγ and tumour necrosis factor (TNF), have been detected in flavivirus-infected patients22,23,25. In DENV infections, increased IL-18 levels in plasma and in induced skin blisters, as well as concomitant signalling downstream of IL-18 receptor in NK cells, have suggested an IL-18-dependent mechanism in driving the proliferative NK cell response22. This proliferation might occur via nuclear factor-κB (NF-κB), AKT, activating transcription factor 2 (ATF2) and forkhead box protein O3 (FOXO3) or indirectly via IL-18-induced upregulation of CD98 and consequently amino acid-induced activation of mechanistic target of rapamycin (mTOR)22. NK cell-produced IFNγ, on the other hand, has in other studies been suggested to be important for control of DENV infection26. These and other related studies illustrate the dynamics and specifics of the early NK cell response during prototype acute flavivirus infections in humans. How NK cells contribute to viral clearance in experimental and human flavivirus infections remains to be explored in further detail.

Influenza virus infections

Influenza viruses cause yearly global epidemics, with up to a billion infections annually27. While vaccines limit their spread, they do not prevent all infections. Typically, the virus infects lung epithelial cells. Like the situation in many viral infections, including SARS-CoV-2 infection (discussed in more detail below), infection with influenza virus can lead to excessive inflammation and tissue damage. Although NK cells are part of this response, their contribution to host defence against influenza A virus (IAV) is not fully understood. Upon IAV infection in mice, increased accumulation of NK cells is observed in the lungs28. While local proliferation of pulmonary NK cells may account for part of this response, most of the NK cell accumulation in the lungs and airways during IAV infection has been suggested to be due to NK cell recruitment that is partly dependent on CXC-chemokine receptor 3 (CXCR3) and CC-chemokine receptor 5 (CCR5), respectively28. Several NK cell natural cytotoxicity receptors have been implicated in functional activation of NK cells following influenza virus infection by binding to influenza virus haemagglutinin29. However, the degree to which NK cells contribute (if at all) to influenza virus elimination in this context is less clear. In humans, peripheral blood CD56bright and CD56dim NK cells have been shown to be primed during acute IAV infection30. Furthermore, in peripheral blood, a small subset of CD16CD49a+CXCR3+ NK cells has been identified, with CD49a and CXCR3 potentially promoting homing to and tissue retention in the lungs during acute infection. At the primary site of infection, it is not fully clear how circulating and tissue-resident NK cells contribute to possible antiviral immunity or to pathology, such as by killing bystander cells30. Finally, studies in humans and macaques have demonstrated that cross-reactive antibodies elicited during seasonal influenza virus infections facilitate NK cell-mediated ADCC with regard to infected target cells31,32. This ADCC-mediated response has been correlated with reduced disease severity. In infections such as influenza virus infection, more research is clearly needed to better delineate and discriminate between possible protective antiviral responses and those leading to excessive inflammation and tissue damage.

NK cell activation in acute infections

The acute NK cell response to infection with flaviviruses, including tick-borne encephalitis virus, DENV and yellow fever virus, as well as SARS-CoV-2 (discussed in detail below), is to a large degree confined to less differentiated CD56bright and CD56dim NK cells22,23,25,33 (Box 1). Since these cells are more responsive to cytokines such as IL-12, IL-15, IL-18 and type I interferons, probably in part because of higher expression of receptors for these cytokines22,34,35, it is likely that early NK cell activation is cytokine driven and less reliant on receptor–ligand interactions (Fig. 1). This results in stronger downstream signalling via signal transducer and activator of transcription 1 (STAT1), STAT4 and the PI3K–AKT–mTOR pathways in less differentiated NK cells22,36,37, in which mTOR, via the IRE1α–XBP1–MYC axis, is crucial for NK cell proliferation following acute activation38. Cytokine signalling via JAK–STAT pathways in NK cells was recently comprehensively reviewed elsewhere39. For induction of early NK cell activation, type I interferon signalling via STAT1 and IL-12 plus IL-18 signalling via STAT4 result in strikingly different interactions with the NK cell epigenetic landscape40. More specifically, STAT1 and STAT4 can exhibit antagonistic functions on their downstream networks early after NK cell activation, including STAT4 actively suppressing STAT1 expression40. Furthermore, whereas STAT4 binding resulted in increased chromatin accessibility, STAT1 interacted more with promoter regions40. Although more long-lasting effects of these alterations in the chromatin landscape remain elusive, it is possible that remodelling via IL-12 plus IL-18 and STAT4 could have more permanent effects as STAT4 signalling is important for the generation of both adaptive-like and cytokine-induced memory-like NK cells41,42.

Fig. 1. NK cell differentiation as a framework to understand NK cell antiviral immunity.

Fig. 1

The differentiation status of human natural killer (NK) cells determines their functional response in viral infections. CD56bright NK cells and less differentiated CD56dim NK cells express high levels of NKG2A, natural cytotoxicity receptors (NCRs; NKp30, NKp44 and NKp46) and proinflammatory and antiviral cytokine receptors (IL-12 receptor (IL-12R), IL-18R and type I interferon receptor (IFNAR)). During acute viral infections, with ensuing production of IL-12, IL-18 and type I interferons by other immune cells, less differentiated NK cells are the main NK cell subset to respond. As indicated by black arrows, cytokine stimulation leads to signalling via signal transducer and activator of transcription 1 (STAT1) and STAT4 and PI3K–AKT–mTOR pathways to rapidly induce NK cell cytokine production and proliferation. With increased maturation, circulating CD56bright NK cells undergo a phenotypic shift and their responsiveness during infection becomes altered. As a consequence, more differentiated NK cells are more prone to respond to cytomegalovirus, leading to the expansion of adaptive-like NK cells that express high levels of killer cell immunoglobulin-like receptors (KIRs) and NKG2C. Circulating CD56bright NK cells might also home to tissues, giving rise to tissue-resident NK cells. Their role in the early response to infection remains elusive. However, liver-resident NK cells have been shown to mediate antigen-specific antiviral responses. Alterations in transcription factor expression during NK cell differentiation are indicated within the respective cell nuclei, and a summary of phenotypic changes during NK cell differentiation is depicted in the table. For some receptors, the expression depends on the tissue microenvironment and specific subsets studied and will deviate from what is indicated here (denoted by the asterisk). CLA, cutaneous lymphocyte-associated antigen; HAV, hepatitis A virus; HBV, hepatitis B virus; ISG, interferon-stimulated gene; NA, not available.

A recent study by Sciumè and colleagues looked into the molecular mechanism of early NK cell activation in response to cytokines43. Analysis was focused on regions surrounding genes highly induced on early NK cell activation and revealed de novo chromatin and enhancer remodelling mediated by STAT4. These de novo enhancers were further repurposed to be used not only by STATs but also by the lineage-defining transcription factor T-bet43. Thus, although T-bet and other lineage-defining transcription factors were once considered pioneers for determining the shape of the epigenetic landscape that signal-regulated transcription factors act on, this instead suggests that in acute activation of NK cells STATs will remodel chromatin accessibility and invite lineage-defining transcription factors to participate in novel ways in the acute response.

NK cell homing in acute infections

Since many viruses have narrow tropisms, infections often originate at the site of entry before spreading locally and/or systematically. Alternatively, they may be selectively restricted to a specific organ due to tissue tropism (for example, the liver in the case of hepatitis virus infections). From considerable work in mouse experimental models, it is known that recruitment of circulating NK cells to the site of infection is important for the early inflammatory response and viral containment4447. Chemokine–chemokine receptor interactions are necessary for mouse NK cell homing to tissues early in infection, and certain combinations come into action depending on the affected organ4547. In a similar way, circulating NK cells migrate into lymph nodes in a CXCR5-dependent manner in macaques and help to control simian immunodeficiency virus (SIV) infection48. In humans, however, there is a relative scarcity of studies assessing NK cell homing to tissues during acute viral infections because of the inherent challenges in performing such studies and lack of easy access to infected tissue samples. Nevertheless, in acute DENV infection, responding peripheral blood NK cells show a distinct chemokine receptor imprint, including high expression of the skin-homing marker cutaneous lymphocyte antigen. Because of this, high numbers of NK cells are present in the skin of patients early after infection22. Similarly, skin challenge with varicella zoster virus antigens in previously vaccinated individuals led to rapid recruitment of NK cells to the site of challenge49. However, studies in humans are warranted where NK cell homing to tissues during acute infections can be studied.

Tissue-resident NK cells in infection

Besides homing of NK cells to tissues in response to acute viral infections, we have in recent years learned that most peripheral organs contain tissue-resident CD56bright NK cells (in humans) and/or group 1 innate lymphoid cells (ILC1s) (in mice) in the steady state (reviewed in ref.10) (Box 2). These cells are often strategically positioned at the site of viral infection in organs, such as the liver, female genital tract, salivary glands, kidneys and intestine, poised to respond rapidly10,50. Indeed, in mouse models it has been shown that tissue-resident ILC1s confer early host protection51. Their human counterpart is most likely tissue-resident CD56bright NK cells (hereafter referred to as tissue-resident NK cells), given their functional similarities, enrichment in tissues and shared expression of tissue-residency markers (Box 2). Since tissue-resident NK cells resemble circulating CD56bright NK cells, cytokine and chemokine production is presumably the main function exhibited by these cells in early stages of infection (Fig. 1). Tissue-resident NK cells can also respond with local proliferation in acute and chronic viral infection52. Tissue-resident NK cells have also been shown to more directly influence the magnitude of T cell responses via expression of the inhibitory ligand PDL1, leading to viral persistence but more limited immunopathology after lymphocytic choriomeningitis virus infection52. In humans, a similar regulation of antiviral T cell immunity has been shown for liver-resident NK cells in chronic hepatitis B virus (HBV) infection53. However, compared with what has been reported for conventional NK cell responses to viral infections, much less is known regarding the role of tissue-resident NK cell responses. In this regard, it is striking to note that antigen-specific NK cell responses to vaccination and/or infection appear to be largely confined to tissue-resident NK cells in both humans and mice49,5456 (Box 3). Future work should aim at disentangling the relative contributions of migrating and tissue-resident NK cells in antiviral responses occurring in peripheral organs.

Box 3 Antigen-specific NK cell responses against viruses.

More than a decade ago, we learned of antigen-specific responses mediated by mouse natural killer (NK) cells54,55,97. These involved antigen-specific recall responses to haptens but also responses against viruses after prior sensitization. Intriguingly, the responses were largely confined to CXC-chemokine receptor 6 (CXCR6)-positive liver-resident NK cells54,55. Similarly, hepatic, but also splenic, NK cells from macaques infected with simian–human immunodeficiency virus and simian immunodeficiency virus or adenovirus vector-vaccinated animals demonstrated long-term antigen specificity in an NKG2-dependent manner156. Recently, such NK cell phenomena have also been reported in the human setting. First, Nikzad and colleagues showed that human NK cells, in humanized mice, exhibited vaccination-dependent, antigen-specific recall responses upon secondary challenge in vitro49. Interestingly, in line with findings from mouse and macaque models, these responses were also confined to hepatic CXCR6+ NK cells49. The human liver is enriched in NK cells and contains a sizeable tissue-resident NK cell subpopulation149. Given this and the association between liver NK cells and antigen-specific NK cell responses from experimental models, it was no surprise that two studies in 2020 reported human liver NK cell antigen-specific responses against hepatitis A virus and hepatitis B virus following vaccination against these viruses56,157. The CD49a+CD16 hepatic NK cells mediating these antigen-specific responses had a primed epigenetic state compared with conventional CD49aCD16+ NK cells, yielding hyper-responsiveness upon IL-2 plus IL-15 stimulation56. However, the mechanism for antigen specificity currently remains elusive and so does the broad usefulness of these cells against other future infections.

NK cells in chronic viral infections

NK cells adapt to chronic viral infection in ways that are only partially understood. These adaptations likely benefit viral control over time. Impairment of the ability to handle chronic viral infections, such as herpesvirus infections, is exemplified by the not seldom lethal consequences of severe inborn deficiencies affecting NK cell development and/or function.

Hepatitis C virus infection

Although hepatitis C virus (HCV) belongs to the family Flaviviridae, it only rarely causes acute symptomatic disease. Instead, HCV is successful in evading the initial immune response and establishing chronic infection. There is strong evidence suggesting that NK cells contribute to HCV infection outcome, as the combination of KIR2DL3 with homozygosity for the killer cell immunoglobulin-like receptor (KIR) ligand gene HLA-C1 has been associated with significantly elevated viral clearance57. Furthermore, certain NK cell phenotypes are linked to reduced acquisition of chronic infection in exposed individuals as well as to the outcome of acute HCV infection58,59. Yet once the virus persists and the infection becomes chronic, the rate of spontaneous clearance is low. In this phase, the immune response against the virus is hampered at several levels. T cells from patients with chronic HCV infection display reduced proliferation and cytokine production60, along with upregulation of inhibitory receptors61,62, a combination of features referred to as ‘exhaustion’. From the lymphocytic choriomeningitis virus infection model, it is known that NK cells can contribute to sustaining the chronic infection by targeting activated CD4+ T cells, which in turn leads to CD8+ T cell exhaustion63, and that NK cell depletion promotes antiviral CD8+ T cell immunity64. A similar mechanism has also been described in chronic HBV infection, in which NK cells mediate depletion of HBV-specific T cells53.

In chronic HCV infection, NK cells display an altered phenotype and functionality. Yet the impact of chronic infection on NK cells appears to be, at least in part, unique to each individual. In assessment of the expression of activating receptors, both elevated and reduced frequencies have been reported59,65,66. Along the same lines, also for NK cell degranulation both reduced levels65,67 and elevated levels68,69 have been reported. By contrast, a consistent effect of chronic HCV infection on NK cells appears to be reduced production of proinflammatory cytokines6870. Thus, although the NK cell compartment is indeed affected in chronic HCV infection, certain aspects of this appear to rely on other factors, such as the duration of infection or the underlying state of liver disease. This effect of chronic HCV infection on the NK cell compartment is likely to be caused by a plethora of mechanisms, but it has been shown that the direct interaction of NK cells with HCV-infected cells leads to reduced expression of NKG2D and decreased functionality71. In line with this, NS5A-mediated production of transforming growth factor-β could also be linked to reduction of NKG2D expression67. In recent years, the development of direct-acting antivirals has revolutionized HCV infection therapy, as these drugs lead to successful viral clearance in nearly all patients, with negligible side effects. This has also made it possible to assess the capacity for reinvigoration of NK cell function after rapid clearance of a chronic infection (Box 4).

Box 4 NK cell reinvigoration after resolution of chronic viral infection.

Since chronic viral infections are cleared only very rarely, it is seldom possible to study immune system reinvigoration after removal of the chronic insult. For instance, antiviral treatment of HIV-1 infection can successfully suppress viral replication but not permanently remove the virus and/or infection. In this regard, the introduction of treatment with direct-acting antivirals for hepatitis C virus (HCV) has presented a unique opportunity for immunologists to address immune system restoration upon pathogen removal. While the viral loads of HCV drop rapidly upon treatment, immune functions appear to be restored only partially. For example, the proliferative capacity of HCV-specific CD8+ T cells is restored158 but mitochondrial alterations persist over time159. Along the same lines, the levels of many, but not all, soluble inflammatory serum proteins that are altered in chronic HCV are normalized upon viral clearance160. Furthermore, the unconventional mucosa-associated invariant T cell subset exhibits phenotypic and functional alterations long after viral clearance161. For natural killer (NK) cells, partial reinvigoration upon viral clearance has been described, although certain NK cell features appear to be affected for a prolonged period. In more detail, a reversal of NK cell activation69 and a rapid restoration of signal transducer and activator of transcription 1 (STAT1) expression and associated type I interferon responsiveness have been described162. On the other hand, the diversity of the NK cell population appears not to be restored after viral clearance163. This might be explained by epigenetic changes caused by chronic infection that are long-lasting in nature. Indeed, in human cytomegalovirus infection, it has been shown that adaptive-like NK cell expansions display epigenetic modifications108 and that chronic antigen stimulation is a driver for such modifications and for related functional alterations164. It is thus plausible that such epigenetic modifications also occur in the context of chronic HCV infection.

HIV-1 infection

The effector capacity of NK cells includes cytotoxic elimination of HIV-1-infected target cells, as well as secretion of IFNγ, TNF and CCL4, influencing antiviral responses and limiting viral spread72,73. Notably, however, a differential role for cytokines, including TNF and IFNγ, has been observed in chronic states of the infection74,75. NK cells also produce β-chemokines, CCL3, CCL4 and CCL5, which function as natural ligands of CCR5, hampering HIV infectivity of target cells76. Additionally, population-level genetic associations between NK cell receptor expression and HIV-1 infection outcome and evolution have clearly revealed the impact of NK cells on HIV-1 disease progression. In HIV, KIRHLA combinations have been associated with the pace of disease progression77 and protection against disease acquisition78. In detail, HLA-B Bw4-80I in combination with KIR3DL1*h but also with KIR3DS1 could be linked to improved viral control77,79. The mechanisms conferring this protection may include both NK cell education through inhibitory receptor ligation80 and the direct interaction of KIRs with HIV-1-derived peptide motifs presented on HLA molecules81,82. Indeed, for KIR3DS1+ NK cells, an HLA-B Bw4-80I-dependent suppression of viral replication has been shown83, further supporting a direct role for NK cells in mediating the observed protective effect. HIV-1-mediated downregulation of HLA molecules, which prevents infected cells from lysis by CD8+ T cells84, may also facilitate NK cell recognition of infected cells. However, this is limited by preservation of expression of HLA-C and HLA-E85. HIV-1 infection has also been shown to trigger the upregulation of ligands for NKG2D and other activating receptors86. However, HIV-1 has enabled mechanisms to limit the recognition of these ligands through the expression of viral accessory proteins87,88. Furthermore, responses characterized by coordinated antibody function, including NK cell-mediated ADCC, are associated with viral controller phenotypes89.

Like for many other viral infections, changes in NK cell receptor repertoire and functionality are observed during HIV-1 infection. For example, treatment-naive HIV-1-infected individuals have a significant loss of CCR7+CD56bright NK cells, and this change has been associated with higher viral loads90. Loss of the CD56dim NK cell subset during HIV-1 infection has also been observed90. Animal models of SIV infection have shown accumulation of CD56CD16+ NK cells in lymph nodes91. Since HIV-1 infection is often subjected to pharmacological intervention, the effects of therapies on NK cells have been studied. Initiation of antiretroviral therapy has been associated with alterations in the CD56bright and CD56dim NK cell populations, with the former increasing after therapy92.

Adaptive-like NK cell expansions

Studies of human peripheral blood NK cells have revealed receptor expression-associated changes throughout life93 that have been linked to an individual’s infection history94. Some of these changes are evident as the appearance of adaptive-like NK cell expansions (reviewed in ref.95) that have been extensively studied in the mouse cytomegalovirus (MCMV) infection model96. In this infection, Ly49H+ NK cells that specifically recognized the MCMV-encoded glycoprotein m157 were expanded on primary infection96. These NK cells were shown to confer protection against MCMV following rechallenge with the virus97. Adaptive-like NK cell expansions have also been identified in humans. The very first indications of adaptive-like NK cells came from observations on the expansion and persistence of CD94+NKG2C+ NK cells in humans infected with human cytomegalovirus (HCMV)98. Adaptive-like NK cell expansions have also been observed following diverse viral infections in humans, including hantavirus infection99, chikungunya virus infection100 and HIV-1 infection101. Notably, all these responses correlated with HCMV co-infection, suggesting that subclinical reactivation of HCMV could be one mechanism underlying the observed responses. By contrast, several other infections, including recurrent herpes simplex virus 2 infection102 and acute Epstein–Barr virus (EBV) infection103, have not been associated with an expansion of NKG2C+ NK cells. Furthermore, adaptive-like NK cell expansions observed in individuals infected with HBV, HCV and/or hepatitis delta virus were shown not to be linked to the chronic hepatitis infections per se but rather to be consequences of an underlying HCMV infection104. Distinct from adaptive-like NK cell expansions characterized by a reshaping of the NK cell repertoire towards certain subpopulations, additional animal models of herpes simplex virus, influenza virus and SIV infection and more recently also work in the human setting assessing vaccination responses have revealed the existence of antigen-specific NK cell responses (Box 3).

In more in-depth recent work in humans, several characteristics of adaptive-like NK cells have been identified, including clonal expansion, persistence and recall responses105,106. Furthermore, these cells are characterized by low expression of the transcription factor zinc-finger protein PLZF (also known as ZBTB16), silencing of intracellular signalling molecules and commonly also expression of CD57 (refs107,108). Human adaptive-like NK cell expansion depends on expression of the NKG2C ligand HLA-E109. Adaptive-like NKG2C+ NK cells differentially recognize distinct HCMV strains encoding variable UL40 peptides that, in combination with proinflammatory signals, control expansion and differentiation of adaptive-like NKG2C+ NK cells110. Interestingly, HCMV-seropositive individuals possessing a homozygous null allele of KLRC2 (encoding NKG2C) remain healthy. In these individuals, the adaptive-like NK cell expansions instead expressed elevated levels of CD2, which synergized with CD16 to activate NK cells in HCMV infection111. It is not unlikely that there are additional environmental factors that similarly shape NK cell phenotype and function that are even less well understood. Finally, since adaptive-like NK cell expansions decrease NK cell repertoire diversity, the long-term consequences of this for the individual remain elusive.

CD56 NK cells in chronic infections

A particular hallmark of chronic viral infections such as HIV-1 and HCV infections is the expansion of a subset of CD56 NK cells (reviewed in ref.112). More recently, CD56 NK cells have also been observed to be expanded in HCMV-seropositive and EBV-seropositive elderly individuals113. In healthy individuals, the CD56 NK cell subset encompasses only a small proportion of the NK cell compartment in peripheral blood114. In chronic infections, however, it may constitute up to half of all peripheral NK cells, largely at the expense of the CD56dim NK cell subset115117. The CD56 NK cell population displays an exhausted phenotype, with reduced levels of perforin, granzyme B, IFNγ and TNF, and lower cellular cytotoxicity than CD56dim NK cells117,118. It has remained unclear to what extent these expanded CD56 NK cells represent a similar or distinct phenotype as compared with the CD56bright or CD56dim NK cell populations in healthy individuals. However, more recently, an unbiased surface receptor screen and a global proteome analysis were performed to obtain detailed molecular and phenotypic information of the CD56 NK cell subset119. It was revealed that CD56 NK cells have a surface proteome and a total proteome related to, but not identical to, those of CD56dim NK cells. Functional investigations in that study examining degranulation, cytotoxicity and IFNγ production of CD56 NK cells from healthy individuals largely confirmed an, at most, moderate responsiveness of these cells119.

NK cell deficiencies and infections

Studies of patients with more than 400 different primary immunodeficiency diseases (PIDs) have provided key insights into host control of viral infections. NK cells are affected in more than 50 PIDs. At present, at least seven PIDs have an abnormality specifically affecting NK cells (reviewed in ref.120) (Fig. 2). These specific NK cell deficiency disorders include mutations affecting total NK cell numbers, NK cell subsets and/or NK cell functions120. The genes affected in these seven NK cell deficiencies are GATA2 (ref.121), MCM4 (ref.122), RTEL1 (ref.123), GINS1 (ref.124), IRF8 (ref.125), MCM10 (ref.126) and FCGR3A127. The first six genes affect NK cell development or maturation, with resulting low total NK cell numbers and specific defects affecting NK cell subsets. FCGR3A gene mutations affect NK cell function despite normal numbers of NK cells120. This is due to a homozygous mutation in FCGR3A (which encodes CD16)127,128. Intriguingly, despite CD16 functioning as an IgG Fc receptor, patients with mutations in FCGR3A have impaired natural cytotoxicity but intact ADCC function128. The clinical presentation of human NK cell deficiency varies from patient to patient; however, hallmark viral infections affecting these individuals include herpesvirus infections such as HCMV, EBV, herpes simplex virus and varicella zoster virus infections120,126 (Fig. 2). These viruses cause disease in almost 60% of reported NK cell deficiency cases120. On the basis of these findings, one may argue that host defence against herpesvirus infections might be one of the most important functions of NK cells in the context of antiviral immunity. In addition, some other viral infections have been reported in patients with NK cell deficiency, including human papillomavirus infection and some respiratory infections120. Interestingly, these observations suggest an important role for NK cells in keeping these, often latent, infections under control. Notably, of patients with reported NK cell deficiency, almost half have been reported to have died prematurely120, underscoring the severity of these diseases. Corroborating these findings, in studies of patients with a GATA2 deficiency, the frequencies of NK cells were directly associated with increasing complications of disease129.

Fig. 2. NK cell deficiencies and viral infections.

Fig. 2

Different genetic mutations have been linked to specific natural killer (NK) cell deficiencies. Affected genes encode either intranuclear proteins (GATA2, IRF8, RTEL1, GINS1, MCM4 and MCM10) or the surface-expressed Fc receptor for IgG CD16 (encoded by FCGR3A). Defects in the intranuclear proteins lead to disturbed NK cell development and/or differentiation. The mutation affecting CD16 causes altered NK cell functionality. A common denominator for NK cell deficiencies is an inappropriate response to viral infections, as evidenced by severe and/or recurrent herpesvirus infections (human cytomegalovirus (HCMV), Epstein–Barr virus (EBV), herpes simplex virus (HSV) and varicella zoster virus (VZV)), as well as human papillomavirus virus (HPV) infection and respiratory infections.

NK cells in COVID-19

In late 2019, a new zoonotic viral pathogen, SARS-CoV-2, emerged and rapidly spread throughout the world, causing more than 60 million infections and 1.5 million deaths during the first year of the pandemic130. SARS-CoV-2 infection can cause COVID-19, a respiratory and vascular disease which, in severe cases, can lead to acute respiratory distress syndrome, multi-organ failure and death130,131. Although the disease pathogenesis is not yet completely understood, a misdirected and hyperactivated immune system is thought to contribute to severe COVID-19, with a likely contribution from NK cells.

NK cell response to SARS-CoV-2

NK cells are early responders to acute SARS-CoV-2 infection, with recruitment of CD56bright and CD56dim NK cells from the circulation to the lungs, leading to reduced numbers of circulating NK cells in acute infection33,132135 (Fig. 3). A gene module for chemotaxis is induced in lung NK cells33,135, and lungs of patients with COVID-19 contain elevated levels of chemokines such as CCL3, CCL3L1, CCL4, CXCL9, CXCL10 and CXCL11 (refs135,136). Although further work is needed in the area, this chemokine production suggests that CXCR3 and CCR5 are important for homing of NK cells to the lungs in acute SARS-CoV-2 infection137. The early depletion of NK cells from the circulation and their redistribution to the site of infection is in line with what has been observed in other severe acute infections, such as acute hantavirus infection99. Furthermore, NK cells display an activated and cycling phenotype in acute SARS-CoV-2 infection at both the protein level and the transcriptomic level, with upregulation of Ki67, CD69, HLA-DR and CD38 (refs33,132,134). In the CD56dim NK cell compartment, less differentiated NKG2A+CD62L+CD57KIR cells were the main responding cells, suggesting a cytokine-driven mechanism of activation33 (Box 1; Fig. 1). Paralleling the activation of NK cells, regulatory programmes appear to be initiated, as evidenced by the upregulation of inhibitory checkpoint receptors such as LAG3, TIGIT and TIM3 (refs33,132) (Fig. 3). This could possibly explain why peripheral blood NK cell function has been reported to be blunted in acute SARS-CoV-2 infection133,138.

Fig. 3. NK cells in COVID-19.

Fig. 3

In the circulation, natural killer (NK) cells respond strongly to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and display an altered phenotype, including elevated expression of activation markers (HLA-DR, CD69 and CD38), inhibitory molecules (TIM3, LAG3 and possibly PD1) and tissue-homing markers (CCR5, CXCR3 and CD62L). Furthermore, circulating NK cells are highly proliferative and upregulate perforin and granzyme B expression. This response is primarily confined to less differentiated NK cells, suggestive of a cytokine-driven response. NK cells likely home to the lungs, where they exhibit an inflamed transcriptional signature. In severe coronavirus disease 2019 (COVID-19), adaptive-like NK cells are found at higher frequencies in the circulation, but it remains unclear whether these cells home to the lungs and interact with infected epithelia that show increased expression of HLA-E, a ligand for the activating receptor NKG2C. Furthermore, the transcriptional profile of NK cells in the lung microenvironment of patients with severe COVID-19 is even further skewed towards inflammation. This lung microenvironment also contains high numbers of myeloid-derived suppressor cells (MDSCs) and immature neutrophils. However, details on how NK cells might interact with these cells remain elusive. Additional outstanding questions related to NK cells in COVID-19 are highlighted in the figure. CCL, CC-chemokine ligand; CCR, CC-chemokine receptor; CXCL, CXC-chemokine ligand; CXCR, CXC-chemokine receptor; KIR, killer cell immunoglobulin-like receptor; LAG3, lymphocyte activation gene 3; TIGIT, T cell immunoreceptor with immunoglobulin and ITIM domains; TIM3, T cell immunoglobulin mucin receptor 3.

Role of NK cells in COVID-19

Although NK cells appear to be robustly activated and home to the lungs in acute SARS-CoV-2 infection, from the currently available studies, the level of NK cell activation (induction of CD69 and HLA-DR expression) or proliferation is not associated strongly with COVID-19 severity. Instead, Maucourant and colleagues reported that severe/critical COVID-19 was associated with the appearance of adaptive-like NK cell expansions33 (Fig. 3). These expansions where characterized by high NKG2C and CD57 expression as well as by narrow KIR profiles33. Since the first report, similar findings have been reported in two additional studies133,139. It currently remains unclear whether the increased frequency of adaptive-like NK cells in severe COVID-19 is because of a direct effect of SARS-CoV-2 or whether this is a bystander phenomenon driven by HCMV33. Indeed, a history of HCMV infection appears more common in patients with severe COVID-19 (ref.140), and some evidence of local HCMV reactivation in the lungs of patients with COVID-19 receiving ventilator treatment has been reported141. On the other hand, both immune and parenchymal cells in the lungs of patients with COVID-19 display elevated HLA-E expression33, and it has been suggested that the SARS-CoV-2 spike protein can induce surface HLA-E expression by lung epithelial cells142. However, it remains to be determined whether upregulated HLA-E will stimulate NK cells via NKG2C or rather inhibit the NKG2A-expressing NK cells.

Future research should also address whether expansion of adaptive-like NK cells is needed to unleash the full antiviral capacity of NK cells against SARS-CoV-2 infection or whether these cells are instead contributing to COVID-19 pathogenesis. In this regard, it is interesting to note that a recent artificial intelligence-guided big-data approach identified a ‘severe viral pandemic core gene signature’ that highlighted IL-15–IL-15RA and NK cell senescence as important determinants for severe or fatal COVID-19 (ref.143). Corroborating this, it was recently suggested that IL-15 could be linked to NK cell dysfunction in COVID-19 and that this occurs late after symptom onset and only in the most severely ill patients144. Thus, these studies support a model in which misdirected and/or exhausted NK cell responses, rather than hyperactivated NK cells, contribute to severe COVID-19. Future work should explore the contribution of myeloid-derived suppressor cells and immature neutrophils145 in relation to NK cell dysfunction in severe COVID-19 (Fig. 3).

Concluding remarks and outlook

As is evident from studies reviewed herein, it is clear that NK cells can mount vigorous responses to several acute viral infections, including acute flavivirus and influenza virus infections as discussed here. In many situations, this triggers homing of circulating NK cells to affected tissues. In parallel, less well-characterized tissue-resident NK cells may also become activated. Critical in all host responses to acute viral infections is the balance between acute antiviral mechanisms and responses contributing to tissue damage and immunopathology. Clearly, more knowledge is needed with respect to direct NK cell-mediated interactions with viral infected cells and the consequences of possible viral clearance by such interactions. NK cells adapt to chronic viral infections in ways that are only partially understood. These adaptations likely benefit viral control over time, although the detailed mechanisms are far from completely understood. Impairment of the ability to handle chronic viral infections, such as herpesvirus infections, is exemplified by the lethal consequences of severe inborn deficiencies affecting NK cell development and/or function. As discussed, at least seven PIDs have an abnormality specifically affecting NK cells. With the recent COVID-19 pandemic, NK cells have come into the spotlight in this context as well. Although the disease pathogenesis is still being worked out, a misdirected and hyperactivated immune system is thought to contribute to severe COVID-19, with a likely significant contribution from NK cells (Fig. 3).

While many insights with respect to a possible role for NK cells in viral infections have been revealed, from studies in mouse models to studies in humans, there are several outstanding questions and unresolved issues. What is the nature of NK cell viral antigen specificity as observed in, for example, hepatitis A virus and HBV vaccination (Box 3)? Furthermore, regarding antigen-specific NK cell responses, what are specific surface receptor-mediated responses versus what might be novel forms of antigen-specificity ‘dictated’ by changes at an epigenetic level (Box 3)? On the same note, what drives adaptive-like expansions of NK cell populations? What is the function of these cells in the context of viral infection? How are NK cells driven towards sites of infection and what is the role of responses from tissue-resident NK cells in the context of acute infections? Is there a division of labour between infiltrating NK cells and tissue-resident NK cells? What distinguishes functional antiviral NK cell responses from disease-causing responses? This is a question of relevance with regard to both acute responses, like in the SARS-CoV-2 situation (Fig. 3), and chronic infections, such as HIV-1 and HCV infections. With regard to vaccination, a presently hot topic, what is the role of NK cells in the generation of efficient vaccine responses? Finally, while this has been discussed for years, is there a role for NK cells in settings of adoptive immunotherapy for viral diseases such as what we are currently witnessing in the context of human malignant diseases?

In summary, although we have gained a great deal of knowledge of NK cells in the context of human viral infections over the past 10 years, much more remains to be learned.

Acknowledgements

The authors thank present and past members of their groups for their contributions towards the understanding of human natural killer cells in health and disease. Their work was supported by the Swedish Research Council, Sweden’s Innovation Agency, the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 948692), the Swedish Cancer Society, the Swedish Foundation for Strategic Research, the Knut and Alice Wallenberg Foundation, Nordstjernan AB, the Center for Innovative Medicine at Karolinska Institutet, Region Stockholm, SRP Diabetes Karolinska Institutet, StratRegen Karolinska Institutet and Karolinska Institutet. The authors apologize to those colleagues whose work has not been cited owing to space constraints.

Glossary

Antibody-dependent cellular cytotoxicity

(ADCC). A mechanism by which natural killer cells, via their Fc receptor CD16, kill a target cell whose membrane-surface antigens have been bound by a specific antibody.

NK cell education

The process involving acquisition of functional competence in natural killer (NK) cells, often mediated by interactions between inhibitory receptors and the corresponding ligands.

Adaptive-like NK cell

A subset of expanded terminally differentiated natural killer (NK) cells that, in the context of human cytomegalovirus infection, is characterized by expression of NKG2C.

JAK–STAT pathways

Signalling pathways involving Janus kinases (JAKs) and signal transducers and activators of transcription (STATs) downstream of many cytokine receptors, including those for IL-2, IL-12, IL-15 and type I interferons, but not IL-18, important in natural killer cell activation, proliferation and survival.

Killer cell immunoglobulin-like receptor

(KIR). A family of highly polymorphic activating and inhibitory receptors that serve as key regulators of human natural killer cell function.

HLA-E

Non-classical MHC class I molecule characterized by a limited polymorphism that normally binds a restricted subset of peptides derived from signal peptides.

Author contributions

The authors contributed equally to all aspects of the article.

Competing interests

The authors declare no competing interests.

Footnotes

Peer review information

Nature Reviews Immunology thanks C. Biron and the other, anonymous, reviewers for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Kiessling R, Klein E, Wigzell H. ‘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. 1975;5:112–117. doi: 10.1002/eji.1830050208. [DOI] [PubMed] [Google Scholar]
  • 2.Kärre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 1986;319:675–678. doi: 10.1038/319675a0. [DOI] [PubMed] [Google Scholar]
  • 3.Ljunggren HG, Kärre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol. Today. 1990;11:237–244. doi: 10.1016/0167-5699(90)90097-s. [DOI] [PubMed] [Google Scholar]
  • 4.Lanier LL. Natural killer cells: from no receptors to too many. Immunity. 1997;6:371–378. doi: 10.1016/s1074-7613(00)80280-0. [DOI] [PubMed] [Google Scholar]
  • 5.Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat. Immunol. 2008;9:503–510. doi: 10.1038/ni1582. [DOI] [PubMed] [Google Scholar]
  • 6.Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2020;16:230–16. doi: 10.1038/s41571-020-0426-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Market M, et al. Flattening the COVID-19 curve with natural killer cell based immunotherapies. Front. Immunol. 2020;11:1512. doi: 10.3389/fimmu.2020.01512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cichocki F, Sitnicka E, Bryceson YT. NK cell development and function-plasticity and redundancy unleashed. Semin. Immunol. 2014;26:114–126. doi: 10.1016/j.smim.2014.02.003. [DOI] [PubMed] [Google Scholar]
  • 9.Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8+ T cells. Nat. Rev. Immunol. 2011;11:645–657. doi: 10.1038/nri3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Björkström NK, Ljunggren H-G, Michaëlsson J. Emerging insights into natural killer cells in human peripheral tissues. Nat. Rev. Immunol. 2016;16:310–320. doi: 10.1038/nri.2016.34. [DOI] [PubMed] [Google Scholar]
  • 11.Lanier LL. NK cell recognition. Annu. Rev. Immunol. 2005;23:225–274. doi: 10.1146/annurev.immunol.23.021704.115526. [DOI] [PubMed] [Google Scholar]
  • 12.Bryceson YT, March ME, Ljunggren H-G, Long EO. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol. Rev. 2006;214:73–91. doi: 10.1111/j.1600-065X.2006.00457.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Biassoni R, et al. Human natural killer cell receptors and co-receptors. Immunol. Rev. 2001;181:203–214. doi: 10.1034/j.1600-065x.2001.1810117.x. [DOI] [PubMed] [Google Scholar]
  • 14.Bartel Y, Bauer B, Steinle A. Modulation of NK cell function by genetically coupled C-type lectin-like receptor/ligand pairs encoded in the human natural killer gene complex. Front. Immunol. 2013;4:362. doi: 10.3389/fimmu.2013.00362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Halenius A, Gerke C, Hengel H. Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets — but how many arrows in the quiver? Cell. Mol. Immunol. 2015;12:139–153. doi: 10.1038/cmi.2014.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ochoa MC, et al. Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol. Cell Biol. 2017;95:347–355. doi: 10.1038/icb.2017.6. [DOI] [PubMed] [Google Scholar]
  • 17.Wu Y, Tian Z, Wei H. Developmental and functional control of natural killer cells by cytokines. Front. Immunol. 2017;8:930. doi: 10.3389/fimmu.2017.00930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood. 2010;115:2167–2176. doi: 10.1182/blood-2009-08-238469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gould EA, Solomon T. Pathogenic flaviviruses. Lancet. 2008;371:500–509. doi: 10.1016/S0140-6736(08)60238-X. [DOI] [PubMed] [Google Scholar]
  • 20.Nelemans T, Kikkert M. Viral innate immune evasion and the pathogenesis of emerging RNA virus infections. Viruses. 2019;11:961. doi: 10.3390/v11100961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mathew A. Defining the role of NK cells during dengue virus infection. Immunology. 2018;154:557–562. doi: 10.1111/imm.12928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zimmer CL, et al. NK cells are activated and primed for skin-homing during acute dengue virus infection in humans. Nat. Commun. 2019;10:3897. doi: 10.1038/s41467-019-11878-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Marquardt N, et al. The human NK cell response to yellow fever virus 17D is primarily governed by NK cell differentiation independently of NK cell education. J. Immunol. 2015;195:3262–3272. doi: 10.4049/jimmunol.1401811. [DOI] [PubMed] [Google Scholar]
  • 24.Yao Y, et al. The natural killer cell response to West Nile virus in young and old individuals with or without a prior history of infection. PLoS ONE. 2017;12:e0172625. doi: 10.1371/journal.pone.0172625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Blom K, et al. NK cell responses to human tick-borne encephalitis virus infection. J. Immunol. 2016;197:2762–2771. doi: 10.4049/jimmunol.1600950. [DOI] [PubMed] [Google Scholar]
  • 26.Costa VV, et al. Dengue virus-infected dendritic cells, but not monocytes, activate natural killer cells through a contact-dependent mechanism involving adhesion molecules. mBio. 2017;8:e00741–17. doi: 10.1128/mBio.00741-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Iuliano AD, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391:1285–1300. doi: 10.1016/S0140-6736(17)33293-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Carlin LE, Hemann EA, Zacharias ZR, Heusel JW, Legge KL. Natural killer cell recruitment to the lung during influenza a virus infection is dependent on CXCR3, CCR5, and virus exposure dose. Front. Immunol. 2018;9:781. doi: 10.3389/fimmu.2018.00781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Luczo JM, Ronzulli SL, Tompkins SM. Influenza a virus hemagglutinin and other pathogen glycoprotein interactions with nk cell natural cytotoxicity receptors NKp46, NKp44, and NKp30. Viruses. 2021;13:156. doi: 10.3390/v13020156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Scharenberg M, et al. Influenza a virus infection induces hyperresponsiveness in human lung tissue-resident and peripheral blood NK cells. Front. Immunol. 2019;10:1116. doi: 10.3389/fimmu.2019.01116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jegaskanda S, Weinfurter JT, Friedrich TC, Kent SJ. Antibody-dependent cellular cytotoxicity is associated with control of pandemic H1N1 influenza virus infection of macaques. J. Virol. 2013;87:5512–5522. doi: 10.1128/JVI.03030-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jegaskanda S, et al. Cross-reactive influenza-specific antibody-dependent cellular cytotoxicity in intravenous immunoglobulin as a potential therapeutic against emerging influenza viruses. J. Infect. Dis. 2014;210:1811–1822. doi: 10.1093/infdis/jiu334. [DOI] [PubMed] [Google Scholar]
  • 33.Maucourant C, et al. Natural killer cell immunotypes related to COVID-19 disease severity. Sci. Immunol. 2020;5:eabd6832. doi: 10.1126/sciimmunol.abd6832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Björkström NK, et al. Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. Blood. 2010;116:3853–3864. doi: 10.1182/blood-2010-04-281675. [DOI] [PubMed] [Google Scholar]
  • 35.Lunemann S, et al. Effects of HDV infection and pegylated interferon α treatment on the natural killer cell compartment in chronically infected individuals. Gut. 2015;64:469–482. doi: 10.1136/gutjnl-2014-306767. [DOI] [PubMed] [Google Scholar]
  • 36.Yu J, et al. CD94 surface density identifies a functional intermediary between the CD56bright and CD56dim human NK-cell subsets. Blood. 2010;115:274–281. doi: 10.1182/blood-2009-04-215491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wagner JA, et al. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J. Clin. Invest. 2017;127:4042–4058. doi: 10.1172/JCI90387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dong H, et al. The IRE1 endoplasmic reticulum stress sensor activates natural killer cell immunity in part by regulating c-Myc. Nat. Immunol. 2019;20:865–878. doi: 10.1038/s41590-019-0388-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gotthardt D, Trifinopoulos J, Sexl V, Putz EM. JAK/STAT cytokine signaling at the crossroad of NK cell development and maturation. Front. Immunol. 2019;10:2590. doi: 10.3389/fimmu.2019.02590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lau CM, et al. Epigenetic control of innate and adaptive immune memory. Nat. Immunol. 2018;19:963–972. doi: 10.1038/s41590-018-0176-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Madera S, Sun JC. Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion. J. Immunol. 2015;194:1408–1412. doi: 10.4049/jimmunol.1402001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Romee R, et al. Cytokine activation induces human memory-like NK cells. Blood. 2012;120:4751–4760. doi: 10.1182/blood-2012-04-419283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sciumè G, et al. Rapid enhancer remodeling and transcription factor repurposing enable high magnitude gene induction upon acute activation of NK cells. Immunity. 2020;53:745–758.e4. doi: 10.1016/j.immuni.2020.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Salazar-Mather TP, Orange JS, Biron CA. Early murine cytomegalovirus (MCMV) infection induces liver natural killer (NK) cell inflammation and protection through macrophage inflammatory protein 1alpha (MIP-1alpha)-dependent pathways. J. Exp. Med. 1998;187:1–14. doi: 10.1084/jem.187.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Trifilo MJ, et al. CXC chemokine ligand 10 controls viral infection in the central nervous system: evidence for a role in innate immune response through recruitment and activation of natural killer cells. J. Virol. 2004;78:585–594. doi: 10.1128/JVI.78.2.585-594.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Thapa M, Kuziel WA, Carr DJJ. Susceptibility of CCR5-deficient mice to genital herpes simplex virus type 2 is linked to NK cell mobilization. J. Virol. 2007;81:3704–3713. doi: 10.1128/JVI.02626-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Gazit R, et al. Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. Nat. Immunol. 2006;7:517–523. doi: 10.1038/ni1322. [DOI] [PubMed] [Google Scholar]
  • 48.Huot N, et al. Natural killer cells migrate into and control simian immunodeficiency virus replication in lymph node follicles in African green monkeys. Nat. Med. 2017;23:1277–1286. doi: 10.1038/nm.4421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nikzad R, et al. Human natural killer cells mediate adaptive immunity to viral antigens. Sci. Immunol. 2019;4:eaat8116. doi: 10.1126/sciimmunol.aat8116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dogra P, et al. Tissue determinants of human NK cell development, function, and residence. Cell. 2020;180:749–763.e13. doi: 10.1016/j.cell.2020.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Weizman O-E, et al. ILC1 confer early host protection at initial sites of viral infection. Cell. 2017;171:795–808.e12. doi: 10.1016/j.cell.2017.09.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhou J, et al. Liver-resident NK cells control antiviral activity of hepatic T cells via the PD-1-PD-L1 axis. Immunity. 2019;50:403–417.e4. doi: 10.1016/j.immuni.2018.12.024. [DOI] [PubMed] [Google Scholar]
  • 53.Peppa D, et al. Up-regulation of a death receptor renders antiviral T cells susceptible to NK cell-mediated deletion. J. Exp. Med. 2013;210:99–114. doi: 10.1084/jem.20121172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.O’Leary JG, Goodarzi M, Drayton DL, Andrian, von UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat. Immunol. 2006;7:507–516. doi: 10.1038/ni1332. [DOI] [PubMed] [Google Scholar]
  • 55.Paust S, et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat. Immunol. 2010;11:1127–1135. doi: 10.1038/ni.1953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stary V, et al. A discrete subset of epigenetically primed human NK cells mediates antigen-specific immune responses. Sci. Immunol. 2020;5:eaba6232. doi: 10.1126/sciimmunol.aba6232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Khakoo SI, et al. HLA and NK cell inhibitory receptor genes in resolving hepatitis C virus infection. Science. 2004;305:872–874. doi: 10.1126/science.1097670. [DOI] [PubMed] [Google Scholar]
  • 58.Golden-Mason L, Cox AL, Randall JA, Cheng L, Rosen HR. Increased natural killer cell cytotoxicity and NKp30 expression protects against hepatitis C virus infection in high-risk individuals and inhibits replication in vitro. Hepatology. 2010;52:1581–1589. doi: 10.1002/hep.23896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Alter G, et al. Reduced frequencies of NKp30+NKp46+, CD161+, and NKG2D+ NK cells in acute HCV infection may predict viral clearance. J. Hepatol. 2011;55:278–288. doi: 10.1016/j.jhep.2010.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Semmo N, et al. Preferential loss of IL-2-secreting CD4+ T helper cells in chronic HCV infection. Hepatology. 2005;41:1019–1028. doi: 10.1002/hep.20669. [DOI] [PubMed] [Google Scholar]
  • 61.Shen T, et al. PD-1 expression on peripheral CD8+ TEM/TEMRA subsets closely correlated with HCV viral load in chronic hepatitis C patients. Virol. J. 2010;7:310. doi: 10.1186/1743-422X-7-310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bengsch B, et al. Coexpression of PD-1, 2B4, CD160 and KLRG1 on exhausted HCV-specific CD8+ T cells is linked to antigen recognition and T cell differentiation. PLoS Pathog. 2010;6:e1000947. doi: 10.1371/journal.ppat.1000947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Waggoner SN, Cornberg M, Selin LK, Welsh RM. Natural killer cells act as rheostats modulating antiviral T cells. Nature. 2012;481:394–398. doi: 10.1038/nature10624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lang PA, et al. Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. Proc. Natl Acad. Sci. USA. 2012;109:1210–1215. doi: 10.1073/pnas.1118834109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nattermann J, et al. Surface expression and cytolytic function of natural killer cell receptors is altered in chronic hepatitis C. Gut. 2006;55:869–877. doi: 10.1136/gut.2005.076463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.De Maria A, et al. Increased natural cytotoxicity receptor expression and relevant IL-10 production in NK cells from chronically infected viremic HCV patients. Eur. J. Immunol. 2007;37:445–455. doi: 10.1002/eji.200635989. [DOI] [PubMed] [Google Scholar]
  • 67.Sène D, et al. Hepatitis C virus (HCV) evades NKG2D-dependent NK cell responses through NS5A-mediated imbalance of inflammatory cytokines. PLoS Pathog. 2010;6:e1001184. doi: 10.1371/journal.ppat.1001184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Oliviero B, et al. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology. 2009;137:1151–1160. doi: 10.1053/j.gastro.2009.05.047. [DOI] [PubMed] [Google Scholar]
  • 69.Serti E, et al. Successful interferon-free therapy of chronic hepatitis C virus infection normalizes natural killer cell function. Gastroenterology. 2015;149:190–200.e2. doi: 10.1053/j.gastro.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lunemann S, et al. Compromised function of natural killer cells in acute and chronic viral hepatitis. J. Infect. Dis. 2014;209:1362–1373. doi: 10.1093/infdis/jit561. [DOI] [PubMed] [Google Scholar]
  • 71.Yoon JC, Lim J-B, Park JH, Lee JM. Cell-to-cell contact with hepatitis C virus-infected cells reduces functional capacity of natural killer cells. J. Virol. 2011;85:12557–12569. doi: 10.1128/JVI.00838-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Scully E, Alter G. NK cells in HIV disease. Curr. HIV AIDS Rep. 2016;13:85–94. doi: 10.1007/s11904-016-0310-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Flórez-Álvarez L, Hernández JC, Zapata W. NK cells in HIV-1 infection: from basic science to vaccine strategies. Front. Immunol. 2018;9:2290. doi: 10.3389/fimmu.2018.02290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gallitano SM, McDermott L, Brar K, Lowenstein E. Use of tumor necrosis factor (TNF) inhibitors in patients with HIV/AIDS. J. Am. Acad. Dermatol. 2016;74:974–980. doi: 10.1016/j.jaad.2015.11.043. [DOI] [PubMed] [Google Scholar]
  • 75.Roff SR, Noon-Song EN, Yamamoto JK. The significance of Interferon-γ in HIV-1 pathogenesis, therapy, and prophylaxis. Front. Immunol. 2014;4:498. doi: 10.3389/fimmu.2013.00498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zapata W, et al. Influence of CCR5 and CCR2 genetic variants in the resistance/susceptibility to HIV in serodiscordant couples from Colombia. AIDS Res. Hum. Retroviruses. 2013;29:1594–1603. doi: 10.1089/aid.2012.0299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Martin MP, et al. Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat. Genet. 2007;39:733–740. doi: 10.1038/ng2035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Boulet S, et al. Increased proportion of KIR3DS1 homozygotes in HIV-exposed uninfected individuals. AIDS. 2008;22:595–599. doi: 10.1097/QAD.0b013e3282f56b23. [DOI] [PubMed] [Google Scholar]
  • 79.Martin MP, et al. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat. Genet. 2002;31:429–434. doi: 10.1038/ng934. [DOI] [PubMed] [Google Scholar]
  • 80.Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu. Rev. Immunol. 2013;31:227–258. doi: 10.1146/annurev-immunol-020711-075005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Alter G, et al. HIV-1 adaptation to NK-cell-mediated immune pressure. Nature. 2011;476:96–100. doi: 10.1038/nature10237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hölzemer A, et al. Selection of an HLA-C*03:04-restricted HIV-1 p24 Gag sequence variant is associated with viral escape from KIR2DL3+natural killer cells: data from an observational cohort in South Africa. PLoS Med. 2015;12:e1001900. doi: 10.1371/journal.pmed.1001900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Alter G, et al. Differential natural killer cell-mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. J. Exp. Med. 2007;204:3027–3036. doi: 10.1084/jem.20070695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature. 1998;391:397–401. doi: 10.1038/34929. [DOI] [PubMed] [Google Scholar]
  • 85.Specht A, et al. Selective downmodulation of HLA-A and -B by Nef alleles from different groups of primate lentiviruses. Virology. 2008;373:229–237. doi: 10.1016/j.virol.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 86.Richard J, Sindhu S, Pham TNQ, Belzile J-P, Cohen EA. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood. 2010;115:1354–1363. doi: 10.1182/blood-2009-08-237370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Norman JM, et al. The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat. Immunol. 2011;12:975–983. doi: 10.1038/ni.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Shah AH, et al. Degranulation of natural killer cells following interaction with HIV-1-infected cells is hindered by downmodulation of NTB-A by Vpu. Cell Host Microbe. 2010;8:397–409. doi: 10.1016/j.chom.2010.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ackerman ME, et al. Polyfunctional HIV-specific antibody responses are associated with spontaneous HIV control. PLoS Pathog. 2016;12:e1005315. doi: 10.1371/journal.ppat.1005315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hong HS, et al. Loss of CCR7 expression on CD56bright NK cells is associated with a CD56dimCD16+ NK cell-like phenotype and correlates with HIV viral load. PLoS ONE. 2012;7:e44820. doi: 10.1371/journal.pone.0044820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Schafer JL, Li H, Evans TI, Estes JD, Reeves RK. Accumulation of cytotoxic CD16+ NK cells in simian immunodeficiency virus-infected lymph nodes associated with in situ differentiation and functional anergy. J. Virol. 2015;89:6887–6894. doi: 10.1128/JVI.00660-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ripa M, et al. Dynamics of adaptive and innate immunity in patients treated during primary human immunodeficiency virus infection: results from Maraviroc in HIV Acute Infection (MAIN) randomized clinical trial. Clin. Microbiol. Infect. 2015;21:876.e1–4. doi: 10.1016/j.cmi.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 93.Horowitz A, et al. Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci. Transl Med. 2013;5:208ra145. doi: 10.1126/scitranslmed.3006702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Strauss-Albee DM, et al. Human NK cell repertoire diversity reflects immune experience and correlates with viral susceptibility. Sci. Transl Med. 2015;7:297ra115. doi: 10.1126/scitranslmed.aac5722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Adams NM, Grassmann S, Sun JC. Clonal expansion of innate and adaptive lymphocytes. Nat. Rev. Immunol. 2020;20:694–707. doi: 10.1038/s41577-020-0307-4. [DOI] [PubMed] [Google Scholar]
  • 96.Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296:1323–1326. doi: 10.1126/science.1070884. [DOI] [PubMed] [Google Scholar]
  • 97.Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457:557–561. doi: 10.1038/nature07665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gumá M, et al. Expansion of CD94/NKG2C+ NK cells in response to human cytomegalovirus-infected fibroblasts. Blood. 2006;107:3624–3631. doi: 10.1182/blood-2005-09-3682. [DOI] [PubMed] [Google Scholar]
  • 99.Björkström NK, et al. Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus. J. Exp. Med. 2011;208:13–21. doi: 10.1084/jem.20100762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Petitdemange C, et al. Unconventional repertoire profile is imprinted during acute chikungunya infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog. 2011;7:e1002268. doi: 10.1371/journal.ppat.1002268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gumá M, et al. 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. 2006;194:38–41. doi: 10.1086/504719. [DOI] [PubMed] [Google Scholar]
  • 102.Björkström NK, Svensson A, Malmberg K-J, Eriksson K, Ljunggren H-G. Characterization of natural killer cell phenotype and function during recurrent human HSV-2 infection. PLoS ONE. 2011;6:e27664. doi: 10.1371/journal.pone.0027664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hendricks DW, et al. Cutting edge: NKG2ChiCD57+ NK cells respond specifically to acute infection with cytomegalovirus and not Epstein-Barr virus. J. Immunol. 2014;192:4492–4496. doi: 10.4049/jimmunol.1303211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Malone DFG, et al. Cytomegalovirus-driven adaptive-like natural killer cell expansions are unaffected by concurrent chronic hepatitis virus infections. Front. Immunol. 2017;8:14725–7. doi: 10.3389/fimmu.2017.00525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Schlums H, et al. Adaptive NK cells can persist in patients with GATA2 mutation depleted of stem and progenitor cells. Blood. 2017;129:1927–1939. doi: 10.1182/blood-2016-08-734236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Cichocki F, et al. ARID5B regulates metabolic programming in human adaptive NK cells. J. Exp. Med. 2018;215:2379–2395. doi: 10.1084/jem.20172168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lee J, et al. Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity. 2015;42:431–442. doi: 10.1016/j.immuni.2015.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Schlums H, et al. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity. 2015;42:443–456. doi: 10.1016/j.immuni.2015.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Rölle A, et al. IL-12-producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion. J. Clin. Invest. 2014;124:5305–5316. doi: 10.1172/JCI77440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hammer Q, et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018;19:453–463. doi: 10.1038/s41590-018-0082-6. [DOI] [PubMed] [Google Scholar]
  • 111.Liu LL, et al. Critical role of CD2 co-stimulation in adaptive natural killer cell responses revealed in NKG2C-deficient humans. Cell Rep. 2016;15:1088–1099. doi: 10.1016/j.celrep.2016.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Björkström NK, Ljunggren H-G, Sandberg JK. CD56 negative NK cells: origin, function, and role in chronic viral disease. Trends Immunol. 2010;31:401–406. doi: 10.1016/j.it.2010.08.003. [DOI] [PubMed] [Google Scholar]
  • 113.Müller-Durovic B, Grählert J, Devine OP, Akbar AN, Hess C. CD56-negative NK cells with impaired effector function expand in CMV and EBV co-infected healthy donors with age. Aging. 2019;11:724–740. doi: 10.18632/aging.101774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mavilio D, et al. Characterization of CD56-/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc. Natl Acad. Sci. USA. 2005;102:2886–2891. doi: 10.1073/pnas.0409872102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Eller MA, et al. 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. 2009;51:380–389. doi: 10.1097/QAI.0b013e3181aa256e. [DOI] [PubMed] [Google Scholar]
  • 116.Mavilio D, et al. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc. Natl Acad. Sci. USA. 2003;100:15011–15016. doi: 10.1073/pnas.2336091100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Alter G, et al. Sequential deregulation of NK cell subset distribution and function starting in acute HIV-1 infection. Blood. 2005;106:3366–3369. doi: 10.1182/blood-2005-03-1100. [DOI] [PubMed] [Google Scholar]
  • 118.Gonzalez VD, et al. Expansion of functionally skewed CD56-negative NK cells in chronic hepatitis C virus infection: correlation with outcome of pegylated IFN-alpha and ribavirin treatment. J. Immunol. 2009;183:6612–6618. doi: 10.4049/jimmunol.0901437. [DOI] [PubMed] [Google Scholar]
  • 119.Voigt J, et al. Proteome analysis of human CD56neg NK cells reveals a homogeneous phenotype surprisingly similar to CD56dim NK cells. Eur. J. Immunol. 2018;48:1456–1469. doi: 10.1002/eji.201747450. [DOI] [PubMed] [Google Scholar]
  • 120.Mace EM, Orange JS. Emerging insights into human health and NK cell biology from the study of NK cell deficiencies. Immunol. Rev. 2019;287:202–225. doi: 10.1111/imr.12725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N. Engl. J. Med. 1989;320:1731–1735. doi: 10.1056/NEJM198906293202605. [DOI] [PubMed] [Google Scholar]
  • 122.Gineau L, et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J. Clin. Invest. 2012;122:821–832. doi: 10.1172/JCI61014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hanna S, Béziat V, Jouanguy E, Casanova J-L, Etzioni A. A homozygous mutation of RTEL1 in a child presenting with an apparently isolated natural killer cell deficiency. J. Allergy Clin. Immunol. 2015;136:1113–1114. doi: 10.1016/j.jaci.2015.04.021. [DOI] [PubMed] [Google Scholar]
  • 124.Cottineau J, et al. Inherited GINS1 deficiency underlies growth retardation along with neutropenia and NK cell deficiency. J. Clin. Invest. 2017;127:1991–2006. doi: 10.1172/JCI90727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Mace EM, et al. Biallelic mutations in IRF8 impair human NK cell maturation and function. J. Clin. Invest. 2017;127:306–320. doi: 10.1172/JCI86276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Mace EM, et al. Human NK cell deficiency as a result of biallelic mutations in MCM10. J. Clin. Invest. 2020;130:5272–5286. doi: 10.1172/JCI134966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Grier JT, et al. Human immunodeficiency-causing mutation defines CD16 in spontaneous NK cell cytotoxicity. J. Clin. Invest. 2012;122:3769–3780. doi: 10.1172/JCI64837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.de Vries E, et al. Identification of an unusual Fc gamma receptor IIIa (CD16) on natural killer cells in a patient with recurrent infections. Blood. 1996;88:3022–3027. [PubMed] [Google Scholar]
  • 129.Spinner MA, et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood. 2014;123:809–821. doi: 10.1182/blood-2013-07-515528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Guan W-J, et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020;382:1708–1720. doi: 10.1056/NEJMoa2002032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wilk AJ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020;26:1070–1076. doi: 10.1038/s41591-020-0944-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Varchetta S, et al. Unique immunological profile in patients with COVID-19. Cell. Mol. Immunol. 2020;395:497. doi: 10.1038/s41423-020-00557-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Xu G, et al. The differential immune responses to COVID-19 in peripheral and lung revealed by single-cell RNA sequencing. Cell Discov. 2020;6:73–14. doi: 10.1038/s41421-020-00225-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Liao M, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020;26:842–844. doi: 10.1038/s41591-020-0901-9. [DOI] [PubMed] [Google Scholar]
  • 136.Chua RL, et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 2020;38:970–979. doi: 10.1038/s41587-020-0602-4. [DOI] [PubMed] [Google Scholar]
  • 137.Brownlie D, et al. Distinct lung-homing receptor expression and activation profiles on NK cell and T cell subsets in COVID-19 and influenza. bioRxiv. 2021 doi: 10.1101/2021.01.13.426553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Osman M, et al. Impaired natural killer cell counts and cytolytic activity in patients with severe COVID-19. Blood Adv. 2020;4:5035–5039. doi: 10.1182/bloodadvances.2020002650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Rendeiro AF, et al. Longitudinal immune profiling of mild and severe COVID-19 reveals innate and adaptive immune dysfunction and provides an early prediction tool for clinical progression. medRxiv. 2020 doi: 10.1101/2020.09.08.20189092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shrock E, et al. Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity. Science. 2020;370:eabd4250. doi: 10.1126/science.abd4250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Le Balc’h P, et al. Herpes simplex virus and cytomegalovirus reactivations among severe COVID-19 patients. Crit. Care. 2020;24:530–3. doi: 10.1186/s13054-020-03252-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bortolotti D, Gentili V, Rizzo S, Rotola A, Rizzo R. SARS-CoV-2 spike 1 protein controls natural killer cell activation via the HLA-E/NKG2A pathway. Cells. 2020;9:1975. doi: 10.3390/cells9091975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sahoo D, et al. AI-guided discovery of the invariant host response to viral pandemics. bioRxiv. 2020 doi: 10.1101/2020.09.21.305698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Liu C, et al. Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19. Cell. 2021;184:1836–1857.e22. doi: 10.1016/j.cell.2021.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kvedaraite E, et al. Major alterations in the mononuclear phagocyte landscape associated with COVID-19 severity. Proc. Natl Acad. Sci. USA. 2021;118:e2018587118. doi: 10.1073/pnas.2018587118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Freud AG, et al. A human CD34+ subset resides in lymph nodes and differentiates into CD56bright natural killer cells. Immunity. 2005;22:295–304. doi: 10.1016/j.immuni.2005.01.013. [DOI] [PubMed] [Google Scholar]
  • 147.Juelke K, et al. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood. 2010;116:1299–1307. doi: 10.1182/blood-2009-11-253286. [DOI] [PubMed] [Google Scholar]
  • 148.Lopez-Vergès S, et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010;116:3865–3874. doi: 10.1182/blood-2010-04-282301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Marquardt N, et al. Cutting edge: identification and characterization of human intrahepatic CD49a+ NK cells. J. Immunol. 2015;194:2467–2471. doi: 10.4049/jimmunol.1402756. [DOI] [PubMed] [Google Scholar]
  • 150.Marquardt N, et al. Unique transcriptional and protein-expression signature in human lung tissue-resident NK cells. Nat. Commun. 2019;10:3841–12. doi: 10.1038/s41467-019-11632-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ivarsson MA, et al. Composition and dynamics of the uterine NK cell KIR repertoire in menstrual blood. Mucosal Immunol. 2017;10:322–331. doi: 10.1038/mi.2016.50. [DOI] [PubMed] [Google Scholar]
  • 152.Snyder ME, et al. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci. Immunol. 2019;4:eaav5581. doi: 10.1126/sciimmunol.aav5581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zuber J, et al. Bidirectional intragraft alloreactivity drives the repopulation of human intestinal allografts and correlates with clinical outcome. Sci. Immunol. 2016;1:eaah3732. doi: 10.1126/sciimmunol.aah3732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lim AI, et al. Systemic human ILC precursors provide a substrate for tissue ILC differentiation. Cell. 2017;168:1086–1100.e10. doi: 10.1016/j.cell.2017.02.021. [DOI] [PubMed] [Google Scholar]
  • 155.Seillet C, Brossay L, Vivier E. Natural killers or ILC1s? That is the question. Curr. Opin. Immunol. 2021;68:48–53. doi: 10.1016/j.coi.2020.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Reeves RK, et al. Antigen-specific NK cell memory in rhesus macaques. Nat. Immunol. 2015;16:927–932. doi: 10.1038/ni.3227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wijaya RS, et al. HBV vaccination and HBV infection induces HBV-specific natural killer cell memory. Gut. 2021;70:357–369. doi: 10.1136/gutjnl-2019-319252. [DOI] [PubMed] [Google Scholar]
  • 158.Martin B, et al. Restoration of HCV-specific CD8+ T cell function by interferon-free therapy. J. Hepatol. 2014;61:538–543. doi: 10.1016/j.jhep.2014.05.043. [DOI] [PubMed] [Google Scholar]
  • 159.Aregay A, et al. Elimination of hepatitis C virus has limited impact on the functional and mitochondrial impairment of HCV-specific CD8+ T cell responses. J. Hepatol. 2019;71:889–899. doi: 10.1016/j.jhep.2019.06.025. [DOI] [PubMed] [Google Scholar]
  • 160.Hengst J, et al. Direct-acting antiviral-induced hepatitis C virus clearance does not completely restore the altered cytokine and chemokine milieu in patients with chronic hepatitis C. J. Infect. Dis. 2016;214:1965–1974. doi: 10.1093/infdis/jiw457. [DOI] [PubMed] [Google Scholar]
  • 161.Hengst J, et al. Nonreversible MAIT cell-dysfunction in chronic hepatitis C virus infection despite successful interferon-free therapy. Eur. J. Immunol. 2016;46:2204–2210. doi: 10.1002/eji.201646447. [DOI] [PubMed] [Google Scholar]
  • 162.Serti E, et al. Rapid decrease in hepatitis C viremia by direct acting antivirals improves the natural killer cell response to IFNα. Gut. 2017;66:724–735. doi: 10.1136/gutjnl-2015-310033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Strunz B, et al. Chronic hepatitis C virus infection irreversibly impacts human natural killer cell repertoire diversity. Nat. Commun. 2018;9:2275. doi: 10.1038/s41467-018-04685-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Merino A, et al. Chronic stimulation drives human NK cell dysfunction and epigenetic reprograming. J. Clin. Invest. 2019;129:3770–3785. doi: 10.1172/JCI125916. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Nature Reviews. Immunology are provided here courtesy of Nature Publishing Group

RESOURCES