Next Article in Journal
Biomarkers as Predictive Factors of Anti-VEGF Response
Previous Article in Journal
Looking for the Holy Grail—Drug Candidates for Glioblastoma Multiforme Chemotherapy
Previous Article in Special Issue
Tumor Marker B7-H6 Bound to the Coiled Coil Peptide-Polymer Conjugate Enables Targeted Therapy by Activating Human Natural Killer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Utility of NK Cells in COVID-19

1
Biotherapy Institute of Japan, Inc., 2-4-8 Edagawa, Koto-ku, Tokyo 135-0051, Japan
2
N2 Clinic Yotsuya, 5F 2-6 Samon-cho, Shinjuku-ku, Tokyo 160-0017, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(5), 1002; https://doi.org/10.3390/biomedicines10051002
Submission received: 28 March 2022 / Revised: 22 April 2022 / Accepted: 25 April 2022 / Published: 26 April 2022
(This article belongs to the Special Issue Recent Therapeutic Advances in Natural Killer Cells)

Abstract

:
Coronavirus disease 2019 (COVID-19) can manifest as acute respiratory distress syndrome and is associated with substantial morbidity and mortality. Extensive data now indicate that immune responses to SARS-CoV-2 infection determine the COVID-19 disease course. A wide range of immunomodulatory agents have been tested for the treatment of COVID-19. Natural killer (NK) cells play an important role in antiviral innate immunity, and anti-SARS-CoV-2 activity and antifibrotic activity are particularly critical for COVID-19 control. Notably, SARS-CoV-2 clearance rate, antibody response, and disease progression in COVID-19 correlate with NK cell status, and NK cell dysfunction is linked with increased SARS-CoV-2 susceptibility. Thus, NK cells function as the key element in the switch from effective to harmful immune responses in COVID-19. However, dysregulation of NK cells has been observed in COVID-19 patients, exhibiting depletion and dysfunction, which correlate with COVID-19 severity; this dysregulation perhaps contributes to disease progression. Given these findings, NK-cell-based therapies with anti-SARS-CoV-2 activity, antifibrotic activity, and strong safety profiles for cancers may encourage the rapid application of functional NK cells as a potential therapeutic strategy to eliminate SARS-CoV-2-infected cells at an early stage, facilitate immune–immune cell interactions, and favor inflammatory processes that prevent and/or reverse over-inflammation and inhibit fibrosis progression, thereby helping in the fight against COVID-19. However, our understanding of the role of NK cells in COVID-19 remains incomplete, and further research on the involvement of NK cells in the pathogenesis of COVID-19 is needed. The rationale of NK-cell-based therapies for COVID-19 has to be based on the timing of therapeutic interventions and disease severity, which may be determined by the balance between beneficial antiviral and potential detrimental pathologic actions. NK cells would be more effective early in SARS-CoV-2 infection and prevent the progression of COVID-19. Immunomodulation by NK cells towards regulatory functions could be useful as an adjunct therapy to prevent the progression of COVID-19.

1. Introduction

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2019 [1], and was declared a pandemic by the World Health Organization (WHO) [2]; it seriously devastated public health and economies worldwide. The spectrum of clinical presentations of COVID-19 individuals ranges from asymptomatic or mild upper respiratory symptoms to severe viral pneumonia causing acute respiratory failure [3,4]. Transmission of SARS-CoV-2 by asymptomatic individuals poses a great public health challenge in containment efforts [5]. The main characteristic symptoms of COVID-19 include fever, fatigue, dry cough, and respiratory distress [3,4]. Severe COVID-19 cases are characterized by pneumonia involving both lungs [6], which can progress to acute respiratory distress syndrome (ARDS), multiorgan failure, and even death [3,4,6,7].
Clinically, the disease symptoms vary depending on the prevalent strain, but most infected patients are asymptomatic. Those who develop pneumonia require hospitalization, a few of whom become severely ill [3]. Approximately 50% of patients with severe COVID-19 had a comorbidity, with hypertension being the most common, followed by diabetes, obesity, and coronary heart disease [3,6,8,9]. A study has shown increasing odds of death associated with older age [9]. Initially, the clinical management of COVID-19 consists of infection prevention and control measures, and supportive care such as oxygenation and mechanical ventilatory support. Current evidence-based treatment guidelines are regularly updated by the National Institutes of Health [10]. The direct-acting antivirals ritonavir-boosted nirmatrelvir (Paxlovid), remdesivir (Veklury), and molnupiravir, and anti-SARS-CoV-2 mAb products (either bamlanivimab plus etesevimab or sasirivimab plus imdevimab or sotrovimab) have recently been approved in the clinical management of COVID-19 patients [10].
Obviously, the pathogenicity of SARS-CoV-2 is largely related to the interplay between the virus and the host [11,12]. Indeed, the interaction between SARS-CoV-2 and host antiviral immunity, including innate and acquired immune responses, have come into the spotlight in this context as well [7,13,14,15]. Severe COVID-19 cases are associated with hypercytokinemia, or the cytokine storm, which is due to exaggerated immune responses leading to severe lung damage [7,15,16]. Analysis of bronchoalveolar immune cells has revealed the roles of macrophages and neutrophils in the pathogenesis of severe COVID-19 [17,18,19]. Autopsy studies of patients who died of COVID-19 provide new insights into the pathophysiology of severe COVID-19: lungs were highly inflamed and had dense infiltrations of aberrantly activated monocyte-derived macrophages and alveolar macrophages, but there was no significant increase in the count of T cells and natural killer (NK) cells in the lungs [20].
To date, it has remained elusive how SARS-CoV-2 infection disrupts the host immune homeostasis and stimulates a hyperactive pro-inflammatory response [13,15]. A proposed model has suggested that the ability of SARS-CoV-2 to evade innate immune responses plays a critical role [21]. SARS-CoV-2 can effectively evade the immune system, causing an inadequate or delayed response [22]. This immune escape gives rise to unrestrained SARS-CoV-2 replication, which eventually results in hyperactivated pro-inflammatory responses [21,23]. NK cells are critical in innate immune responses as essential frontline responders to viral infections [24]. SARS-CoV-2-specific NK cell responses have been observed in vaccinated macaques [25], as well as in patients convalescing from COVID-19 [26]. Human NK cells were found to have anti-SARS-CoV-2 activity [27,28] and antifibrotic activity [27]. Accordingly, NK cell receptors affect the clearance of SARS-CoV-2 [29] and COVID-19 progression [30,31,32,33,34,35]. In addition, a high NK cell count is associated with viral load decline [28], a short time duration of SARS-CoV-2 viral RNA shedding, a stronger antibody response, and a higher survival rate in COVID-19 patients [36,37]. Hence, increasing the count of functional NK cells might help in the management of COVID-19 through the elimination of SARS-CoV-2-infected cells at an early stage and inhibition of disease progression. Consequently, the use of NK cells has been proposed as a suitable therapeutic approach for COVID-19 patients [38]. However, our understanding of the role of NK cells in COVID-19 remains limited. In addition to their beneficial antiviral role, NK cells could also possibly pathologically damage host tissues in viral infections [39,40]. Thus, there is an urgent need to explore the utility of NK cells in SARS-CoV-2 infection by reviewing reported data obtained from COVID-19 patients to deepen our understanding of the role of NK cells in COVID-19, and to improve the therapeutic approaches for the prevention of SARS-CoV-2 infection and treatment of COVID-19 patients.

2. NK Cells

NK cells are innate immune cells that play essential roles in immunosurveillance against viral pathogens and cancer [41,42], while also regulating adaptive immune responses [43,44]. They work through a non-self-strategy in which they act against cells that do not express major histocompatibility complex class I (MHC-I) molecules, which mark cells as “self”. MHC-I-specific inhibitory receptors on NK cells include killer cell immunoglobulin-like receptors (KIRs) and lectin-like CD94–NKG2A heterodimers, which identify self-cells and prevent NK cells from being activated [45,46]. The activating receptors activated by pathogens and cellular stress induce signaling pathways, e.g., NKG2D, NKp30, and NKp46, which trigger NK cell responses [45,46]. NK cells kill target cells through various mechanisms: (i) direct lysis of target cells through cytotoxic degranulation by perforin and granzymes, or death receptors; (ii) production of inflammatory cytokines, such as interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), leading to indirect elimination of target cells; (iii) expression of CD16, which allows for the detection of antibody-coated target cells, thus leading to NK-cell-led antibody-dependent cell cytotoxicity (ADCC); and (iv) by interacting with other immune cells, such as monocytes, inducing IFN-γ production and enhancing cytotoxicity [47,48].
NK cells constitute 10–15% of the total lymphocytes in human peripheral blood [48,49]. NK cells are negative for CD3 and positive for CD16 or CD56, which are cell surface markers [48,49]. There are two major types of NK cell: CD56dim CD16bright/+ (CD56dim) and CD56bright CD16dim/− (CD56bright) NK cells. The CD56dim NK cells are the major subset, constituting 90% of NK cells in peripheral blood, and they have low cytokine production but have high cytotoxicity. They are involved in ADCC. The CD56bright NK cells constitute the remaining 10%, and they produce abundant cytokines but have low natural cytotoxicity [49,50,51]. However, CD56bright NK cells can also become cytotoxic upon appropriate activation, and play roles in different disease states, such as cancers and infections [52].
In the human lung, NK cells comprise up to 20% of all lymphocytes, and are composed of three different subsets: CD56dim, CD56dimCD16, and CD56bright NK cells [53,54]. CD56dim NK cells are the vast majority (up to 80%) of lung NK cells, with the mature NK cells mainly circulating in the lung [55]. The remaining subsets correspond to either an intermediate stage of differentiation or recently activated NK cells that have lost CD16 expression on their surfaces [53]. CD49a+CD69+CD103+ NK cells reside specifically in the lung, representing <3% of the total lung NK cells, most of which are CD56bright NK cells [53,54]. Lung CD56dim NK cells are hypofunctional against target cells in terms of cytotoxicity, ADCC, and following stimulation by IFN-α [55]. By contrast, CD49a+CD56bright lung-resident NK cells show a higher capacity to degranulate and to produce IFN-γ upon contact with virus-infected autologous macrophages in vitro, as compared with matched peripheral blood CD56bright NK cells [54]. As a whole, in the lung, the predominant circulating NK cells are highly differentiated but hypofunctional, whereas lung-resident NK cells are hyperfunctional [53,54,55].

3. NK Cells in Viral Infections

NK cells play an important role in innate immune responses to viral infections [24,56,57,58]. The critical role of NK cells in eliminating viral pathogens is highlighted by a case report of a patient lacking NK cells who was highly susceptible to herpesvirus infection [59]. Subsequent studies of NK cell deficiency disorders, which are associated with the complete or partial impairment of NK cells in terms of their number and function, have been associated with an increased susceptibility to viral infections, as well as to more severe and progressive diseases [56]. These studies established the critical role of functional NK cells in the control of viral infections in humans [57,59].
Viral infections trigger a panel of common modifications in their respective target cells, which can be sensed by NK cells; NK cells utilize various recognition modes in viral infections [57]. The functional outcome of NK cells is determined by the integration of both activating and inhibitory signals that regulate NK cell activity [45,46,60]. Virus-infected cells show down-regulation in MHC-I expression [61,62], which decreases the frequency of inhibitory signals on NK cells [44,45,46]. Moreover, virus-infected cells expressing defined ligands can be directly targeted through activating receptors, such as natural cytotoxicity receptors (NKp30, NKp44, NKp46, and NKG2D [57,58,60]); e.g., NKp30 recognizes pp65 of human cytomegalovirus (CMV), NKp46 recognizes hemagglutinin of the influenza virus, and human NKG2D recognizes CMV glycoprotein UL16-binding proteins and the MHC-I-related molecules MICA and MICB [57,58,60]. Additionally, NK cells directly recognize viral moieties via the engagement of pathogen-associated molecular patterns [63] or transmembrane-activating receptors, such as human NKG2C [26,64]. Moreover, NK cells can be activated via CD16-mediated ADCC [48,65] or cytokines, such as interleukin (IL)-2, IL-12, IL-15, IL-18, and type I IFN, which can be produced by virus-infected cells and/or antigen-presenting cells [66]. As a result, NK cells are activated to kill virus-infected cells through the release of cytotoxic granules (containing perforin and granzymes) and death receptors, causing direct lysis and release of cytokines and chemokines, such as IFN-γ and TNF-α, to enhance cytotoxicity, while also regulating the adaptive immune responses [43,44]. These features suggest that NK cells serve as the major antiviral effector cells during the time that the acquired immune system is still being mustered [24,57] and virus-infected cells are developing mechanisms to escape T cell surveillance [62].
However, NK cells are also possibly associated with immunopathology in viral infections, such as influenza A virus (IAV) [39] and hepatitis B virus (HBV) [40] infections. NK cells are activated during viral infections [39,40]. The activated NK cells by IAV infection, which are responsive to the normally NK-cell-resistant lung epithelial cell line A549, could contribute to the killing of non-infected epithelial cells [39]; furthermore, the activation status of NK cells is related to the severity of e-antigen-negative HBV infection [40]. By contrast, in human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections, NK cells appear to act as a rheostat by eliminating activated CD4+ T cells that affect CD8+ T cell function and exhaustion, thus preventing T-cell-mediated pathology [43]. Additionally, NK cells crosstalk with myeloid cells, such as monocytes, macrophages, dendritic cells (DCs), and neutrophils in antiviral responses [67,68]. In particular, NK cells can effectively counteract virus-infected monocytes and macrophages, which produce large amounts of pro-inflammatory cytokines and chemokines, contributing to local inflammation and a dangerous systemic inflammatory response called the cytokine storm [69]. Therefore, when we examine the role of NK cells in SARS-CoV-2 infection, which is causing the ongoing COVID-19 pandemic, we should keep in mind the double-edged roles of NK cells in viral infections and inflammation-driven damage [70].

4. NK Cells in COVID-19 Patients

NK cells have become the focus of interest since the initial report of SARS-CoV-2 infection [71]. Data on NK cells in COVID-19 patients continue to rapidly accumulate [26,27,28,30,31,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90]. Here, we give a detailed description of NK cells in COVID-19 patients.

4.1. Count and Frequency

In association with lymphopenia in COVID-19 patients, the NK cell count in peripheral blood is consistently and significantly decreased [27,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85], and the count is inversely correlated with COVID-19 severity [27,71,73,74,75,77,78,80,81,82,83,84,85]. NK cell frequency is either decreased [74,87,88,90] or not significantly altered [33,34,75,77,78,79,84,85,89]. However, the NK cell count is restored to normal levels after clinical recovery from COVID-19 [71,78]. The depletion of NK cells in the peripheral blood in COVID-19 patients could potentially, in part, be due to their mobilization and homing to different tissue compartments. However, these cells may also, and more likely, be adversely affected by SARS-CoV-2 [27], whereby the virus-infected NK cells are activated, leading to their death during infection. These mechanisms may underlie the depletion of NK cells in COVID-19 patients [27,91,92]. However, these mechanisms need to be investigated further.

4.2. Subsets

NK cells are typically divided into subsets of cytokine-producing CD56bright NK cells and cytotoxic CD56dim NK cells [49]. Most studies have shown that the proportions of CD56bright NK cells are decreased [27,72,77,79,86,87,88], with some exceptions [33,74,90]. However, findings on CD56dim NK cells are conflicting: many results have shown decreased proportions [27,34,74,77,87,88], while some others have shown increased proportions [72,79] or no change [33,86,89,90] in COVID-19 patients. The reason for these conflicting results is currently unknown, and the correlation between NK cell subsets and patient conditions needs to be investigated in the future.

4.3. Immunotypes

NK cells play important roles in viral infections [24,56,57,58]; indeed, viral infections were found to correlate with the immunotypes of NK cells and disease progression [93]. Therefore, the immunotypes of NK cells have been intensively studied in COVID-19 patients [26,27,33,34,74,75,77,79,81,86,88,89,90]. NK cells become activated with increased expression levels of CD38 [27,75,90], HLA-DR [27,75,77,86], CD69 [27,77,79,86,90], and Ksp37 [77]. CD38 is a type II 45 kDa glycoprotein usually present on the surface membrane of NK cells, and physically/functionally clustered with FcγR III/CD16 [94]. HLA-DR-expressing NK cells are functionally activated [95]. CD69 is a triggering molecule [96] associated with early activation [97] and tissue residency [98]. Ksp37-expressing cells also express perforin, indicating their cytotoxic potential [99]. Additionally, NK cells exhibit a skewing to exhaustion due to increased expression levels of NKG2A [71,81,86,88], Tim-3 [75,77,79], CD244 [74], PD-1 [26,74,79], and CD39 [81], or increased proportions of “exhausted” CD56dim CD16 [33,34,74] and CD56CD16+ [74,86] NK cells. The upregulated NKG2A expression in NK cells was found to be mediated by the viral spike protein [100]. However, SARS-CoV-2 encodes the HLA-E-stabilizing peptide Nsp13, which abrogates the inhibition of NKG2A-expressing NK cells [101]. Expansion of the CD56dim CD16 NK cell subset in COVID-19 patients was accompanied by decreased NK activity [33,34] and prolonged recovery of patients with severe COVID-19 [34]. The aforementioned exhausted NK cells have also been demonstrated in other viral infections, such as NKG2A+ and CD56CD16+ NK cells in HBV infection [102,103]; Tim-3+, PD-1+, and CD39+ NK cells in HIV infection [104,105,106]; CD244+ NK cells in Epstein–Barr virus infection [107]; and CD56dim CD16 NK cells in HCV infection [108]. However, the increased expression levels of NKG2A [71], CD69, and Tim-3 [79] in NK cells are restored after clinical recovery from COVID-19. Moreover, the expression levels of NK-cell-activating receptors, such as NKG2D, sialic acid-binding Ig-like lectin 7 (Siglec-7), DNAX accessory molecule-1 (DNAM-1) [79,90], NKp46 [26], and CD16 [33,34,74], are decreased. The reduced expression levels of these activating receptors have been associated with reduced NK cell function in viral infections [109,110,111]. DNAM-1 expression in NK cells is critical to the clearance of SARS-CoV-2 and rapid recovery from infection [29]. In addition, the decreased CD16 expression level might be associated with abnormal NK cell maturation during infection or prolonged cellular activity/activation, driving CD16 shedding, which might be a regulatory mechanism to prevent activation-induced cell death [112]. Moreover, NK cells show increased expression levels of Ki-67 [77,88,90], CD98 [77], CXCR6, CD103 [86], the transcription factor Aiolos [79], caspase-3, and CD95 [27], which are related to cell cycling, metabolism, tissue residency, hematopoietic development, and apoptosis, whereas the expression level of the homing and extravasation marker CD49d is decreased [86]. Thus, the count of tissue-resident CD69+CD103+CXCR6+ NK cells increases in circulation, with a related surge of CD34+DNMA-1brightCXCR4+ inflammatory precursors from the bone marrow [86]. However, the count of CXCR6-expressing NK cells was decreased in another report [79]. Additionally, patients with poor outcomes have shown an expansion of CD56dim CD57+ NKG2C+ [77,89] or CD56+CD57+FcεRIγ [79] adaptive NK cells. The expansion of adaptive NK cells is often associated with human CMV infection [113]. However, most of the patients are negative for CMV DNA in serum, and the expanded adaptive NK cells do not likely depend on CMV reactivation [77]. However, a later study has shown that the expansion of adaptive NK cells occurred mostly, but not exclusively, in individuals who were CMV-seropositive [89], raising the possibility of CMV reaction in COVID-19 patients. However, the expanded CD57+ NKG2C+ NK cells have been detected in convalescent COVID-19 patients with a SARS-CoV-2-specific response, raising the possibility that NK cell memory may also be generated [26]. In addition, perforin- and granzyme B-expressing CD56bright NK cells have also been observed across COVID-19 disease states, which are driven by a defined inflammatory soluble factor, and correlate with disease severity [77,89]. Taken together, NK cells are robustly activated and exhausted in the peripheral blood of COVID-19 patients, and the counts of adaptive NK cells, armed CD56bright NK cells, and bone-marrow-originating inflammatory precursors in circulation are increased in patients with severe COVID-19.

4.4. NK Cell Activity

NK cell activity in COVID-19 patients has been assessed on the basis of flow-cytometry-based CD107a (degranulation) expression. The percentage of CD107a-expressing NK cells is consistently lower in COVID-19 patients than in controls [27,28,71,72,79,86]. There is a statistically significant decrease in expression level of CD107a that is negatively correlated with C-reactive protein levels, indicating that, under conditions of exaggerated systemic inflammation, NK cells are dysfunctional in the peripheral blood of COVID-19 patients [79]. Incubation of NK cells with plasma from COVID-19 patients, especially plasma from patients with severe COVID-19, impaired CD107a expression in response to K562 cells compared with control plasma, suggesting the presence of soluble factors in the plasma that most likely contribute to the impairment [27]. TGF-β is one of these soluble factors that inhibit NK cell function [28].

4.5. Production of Cytokines and Chemokines

The antiviral effects of NK cells can be mediated through the cytokines they produce [24], in addition to crosstalks with other innate immune cells [67,68] and regulation of the adaptive immune response [43,44]. Therefore, the cytokine production ability of NK cells is an important parameter for NK cell function, in addition to NK cell activity. In the peripheral blood of COVID-19 patients, the ability of NK cells to produce cytokines, such as IL-2 [71], IFN-γ [27,71,79], and TNF-α [27,71,73], is reduced. Impaired IFN-γ and TNF-α production may be due to soluble factors in plasma from COVID-19 patients, as shown by a study on NK cell activity in response to K562 cell stimulation [27]. However, some studies do not report a significant decrease in IFN-γ production [72,73]. By contrast, the production of the chemokine MIP-1β (macrophage inflammatory protein-β) is increased [77]. These results indicate that the production of cytokines by NK cells in peripheral blood is impaired and exhibits a skewing towards the redistribution of NK cells to sites in the inflamed lung [77,92], which may be related to the decrease in the count of NK cells in circulation [27,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85].

4.6. Functional Granule Components

The main mechanism by which NK cells eliminate virus-infected cells or transformed cells involves granule exocytosis, with direct release of cytolytic granules containing perforin and granzymes that kill target cells via apoptosis [114]. To explore the functional granule components in NK cells in COVID-19 patients, the expression levels of perforin, granzyme A (GrA), and granzyme B (GrB) in NK cells have been investigated. Data on functional granule components are inconclusive. The perforin expression level is increased [28,77,80,89,90], with some exceptions [73,86]. Regarding the expression levels of granzymes in COVID-19 patients, the results varied among studies: The expression level of GrA increased in [80] and decreased in [73], whereas that of GrB increased in [28,77,89] and decreased in [71,80]. The discrepancies among studies might be caused by the time of sampling during the disease course and disease severity.

4.7. Genetic Variants

NK cells rely on inhibitory and activating receptor signals to determine which target cells will be attacked or tolerated [45,46]. NKG2C is an activating NK cell receptor encoded by KLRC2; it binds to human leukocyte antigen (HLA)-E, leading to NK cell activation [30]. Heterozygous or homozygous KLRC2 deletion may naturally occur, and is associated with a significantly low NKG2C expression level, or the absence of expression [30]. In addition, HLA-E* 0101/0103 genetic variants occur, which are attributable to a single-nucleotide polymorphism, and the cell surface expression level of HLA-E* 0101/0101 is lower than that of HLA-E* 0103/0103 [30]. The deletions of KLRC2 and, at a lower degree, the HLA-E* 0101 allele are independent risk factors for severe COVID-19, suggesting that genetic variants in the NKG2C/HLA-E axis have a significant impact on COVID-19 severity. Detection of these variants my help identify patients at high risk of developing severe COVID-19 [30]. Moreover, KIRs expressed on NK cells are critical in regulating NK cell responsiveness through their binding to HLA class I ligands on cells [45,46]. A study has shown that the reduced gene coding for the activating receptor KIR2DS2 is associated with severe COVID-19 [31]. In particular, the frequency of the KIR2DS2/HLA C1 functional unit was reduced in severe COVID-19, suggesting a protective effect against adverse outcomes of COVID-19, which is probably achieved through the effective activity of NK cells, and thereby contributes to viral clearance in the early stages of SARS-CoV-2 infection [31]. In line with these studies, higher frequencies of the functional A-telomeric activating receptor KIR2DS4 [35] and the inhibitory receptors KIR2DL1 and KIR2DL1/S1 [33] are associated with severe COVID-19, whereas a higher frequency of activating receptor KIR3DS1 in the presence of HLA-B*15:01 is associated with mild and/or moderate COVID-19 [35].

5. Dysregulation of NK Cells in COVID-19 Patients

The immune responses to SARS-CoV-2 are key determinants of COVID-19 severity and outcome [11,12]; therefore, understanding the immunological underpinnings of COVID-19 pathogenesis is critical for the prevention of SARS-CoV-2 infection and COVID-19 treatment. NK cells are important in the immune defense against viral infections [24,56,57,58] and are the critical responders to SARS-CoV-2 infection [26,27,28]. However, dysregulation of NK cells has been observed, the cells exhibiting depletion and dysfunction in COVID-19 patients [26,27,28,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90] (Figure 1), accompanied by expansions of adaptive NK cells [26,77,79,89] and more mature perforin- and granzyme B-armed CD56bright NK cells [77,89], as well as the recruitment of CD34+DNAM-1brightCXCR4+ inflammatory precursors from the bone marrow [86].
The dysregulation of NK cells is associated with the severity of COVID-19. The depletion of NK cells from the peripheral blood may be due to the direct infection of NK cells by SARS-CoV-2 and/or the activation-induced cell death of NK cells during infection [27,91,115], as well as the mobilization/homing of NK cells to affected tissues [77,91,92]. NK cell function may be impaired by SARS-CoV-2 infection through, for example, the upregulation of inhibitory receptor NKG2A expression by the viral spike protein [100]. Additionally, an impaired IFN type I response of NK cells has been demonstrated in SARS-CoV-2 infection [15,27,75], which could be induced by type I IFN deficiency [75], prolonged and excessive IFN-α production [27], and autoantibodies against type I IFNs [116]. The type I IFN response in NK cells is crucial to antiviral defense [15,117], in conjunction with the ability to contain SARS-CoV-2 infection [118]. Moreover, hyperinflammation responses, such as elevated IL-6 levels [27,73,75,78,83,84,89] and an untimely early production of TGF-β [28] or other unknown soluble plasma components [27], might also result in the impairment of NK cells [27,28,119]. Furthermore, analysis of transcriptomic datasets of host responses to viral infections has revealed that the IL-15/IL-15RA axis plays a role in immune dysregulation [120], in which prolonged exposure of NK cells to high circulating IL-15 levels in a COVID-19 patient triggers NK cell dysfunction [121]. Effective immune responses to viral infections mostly depend on the adequate orchestration of both innate and adaptive immune responses to the virus. NK cells, DCs, macrophages, and granulocytes are innate immune cells, whereas T and B cells are adaptive immune cells. Presently, not much is known about the features of immune responses to SARS-CoV-2 that protect against severe COVID-19 because most cohorts profiled to date have included only hospitalized patients. Neutralizing antibodies and SARS-CoV-2-specific T cell responses have been detected in mildly symptomatic patients, providing evidence of an effective immune response across the disease spectrum [122]. Notably, patients with mild COVID-19 have much lower levels of pro-inflammatory plasma cytokines [18,84], suggesting that the immune response in mild COVID-19 can eradicate the virus without triggering the hyperinflammatory state observed in severe cases. NK cells are the first responders to viral infection [24,57]. Upon SARS-CoV-2 infection, local innate immune cells, such as tissue-resident NK cells, DCs, and alveolar macrophages, respond rapidly in SARS-CoV-2-infected tissues to initiate immune responses to eliminate the virus [44,67,68]. Functional NK cells exhibit balanced activation, in which the expression levels of activating receptors are high, whereas those of inhibitory receptors are low, upon recognition of SARS-CoV-2-infected cells [42]. As a consequence, a sufficient count of activated functional NK cells exert enhanced degranulation and cytokine secretion, and initiate innate cytotoxic function, to eliminate SARS-CoV-2-infected cells at an early stage [24,57]. Moreover, crosstalks of NK cells with DCs are facilitated to modulate adaptive immune responses [44,67,68]. DCs recognize the degraded SARS-CoV-2 antigen, and activated NK cells secrete an array of cytokines, such as IFN-γ, that promote DC maturation with the establishment of a cytokine milieu, such as the release of IL-12, and present the SARS-CoV-2-derived peptides to CD4+ T cells and CD8+ T cells through TCR–MHC interactions. Once exposed to the antigen, Th0 CD4+ T cells polarize primarily towards Th1, leading to the release of cytokines, which facilitate DC antigen presentation to cytotoxic CD8+ T cells (CTLs). CTLs detect SARS-CoV-2-infected cells and release cytotoxic granules, including GrB and perforin, to eliminate SARS-CoV-2-infected cells. On the other hand, Th2 CD4 T cells facilitate antigen-recognized B cells to trigger humoral-mediated immune responses and the secretion of antibodies involved in SARS-CoV-2 elimination [123,124,125] (Figure 2). A scenario of effective host anti-SARS-CoV-2 immunity emerges in asymptomatic infection and in mild upper respiratory symptomatic COVID-19 infection.
Lessons learned from genetic syndromes of NK cell cytotoxic defects have implicated NK cells in the quality control of innate immune responses by inhibiting excessive myeloid responses, thereby preventing immunopathology [126,127]. The dysregulation of NK cells in COVID-19 patients results in not only the failure to eliminate SARS-CoV-2-infected cells directly [27,28] but also the indirect impairment of the crosstalks of NK cells with DCs to modulate adaptive immune responses [67,68]. Consequently, failure to provide adequate CTL- and antibody-mediated SARS-CoV-2 elimination allows the virus to persistently replicate and spread, leading to further recruitment of monocytes and granulocytes. As a result, the excessive and prolonged stimulation of the immune system, with the progressive accumulation of SARS-CoV-2-infected cells and inflammatory myeloid cells in sites of viral infection, occurs [17,18,19,20]; this in turn leads to the production of cytokines and chemokines [69], and to the further recruitment of inflammatory myeloid cells. These processes refer to the dysregulated release of pro-inflammatory cytokines at elevated levels, contributing to over-inflammation and tissue injury [15,16,128] (Figure 3). A scenario of dysregulated and exaggerated immune responses emerges in severe and critical COVID-19 patients.
Indeed, the lungs of severe COVID-19 patients are highly infiltrated with aberrantly activated macrophages and neutrophils, without significant increases in the counts of T cells and NK cells [17,18,19,20]. By contrast, more NK cells and T cells are accumulated, with fewer neutrophils and macrophages, in mild COVID-19 patients [129]; this suggests that NK cells do not participate in the exaggerated inflammatory responses observed in ARDS, but may facilitate immune–immune cell interactions to prevent over-inflammation and tissue injury [129]. Thus, NK cells function as key elements in the switch from an effective to a harmful immune response in COVID-19. Therefore, an adequate count of functional NK cells may strengthen the immunosurveillance against SARS-CoV-2 infection [42]. Indeed, dysfunction of NK cells is linked with increased SARS-CoV-2 susceptibility. Genetic variants [30,31], aging, obesity, unhealthy lifestyle [42], and cancer [130] cause NK cell dysfunction, which is associated with increased risk of severe COVID-19 [9,30,31,131,132,133]. Conversely, a higher count of NK cells is associated with a faster decline in viral load [28], a shorter time duration of SARS-CoV-2 viral RNA shedding, a better antibody response, and a higher survival rate in COVID-19 patients [36,37]. In addition, higher expression levels of the activating receptor DNAM1 on NK cells correlate with the rapid clearance of SARS-CoV-2 and recovery from COVID-19 [29].
Since late November 2021, the Omicron variant has emerged as the primary cause of COVID-19, and caused a huge increase in the reported incidences around the world [134]. Immune responses may vary between strains and variants of SARS-CoV-2 [134]. However, for this paper, data were collected before the emergence of the Omicron variant. Therefore, NK cell responses to the Omicron variant, and their differences between strains and variants, are currently not known owing to absence of genomic information. Moreover, NK cell responses to the currently prevalent Delta and Omicron variants may differ. The Omicron variant may initiate more effective immune responses, including NK cell responses, than other variants, especially the Delta variant; as such, variant-dependent responses should to be explored further.

6. NK-Cell-Based Therapies for COVID-19

Currently, the vaccines and drugs for SARS-CoV-2 have worked in improving the pandemic situation and outcome of COVID-19. We remain cautious, however, because mutant SARS-CoV-2 strains and new viruses will test our pandemic preparedness, requiring accelerated and swift deployment of experimental therapeutics to counter threats to human health. NK cells are important effectors in not only combating SARS-CoV-2 infection directly [27,28,101], but also indirectly by modulating adaptive immune responses to curtail infection through crosstalks with DCs [44,67,68] (Figure 2), and by inhibiting fibrosis progression [27]. Dysregulation of NK cells has been observed in COVID-19 patients, the cells exhibiting depletion and dysfunction [26,27,28,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90]; NK cell dysfunction is linked with increased SARS-CoV-2 susceptibility [9,30,31,131,132,133], suggesting that dysregulation of NK cells may contribute to the worsening of COVID-19 (Figure 3). Therefore, therapies based on functional NK cells may provide potential therapeutic strategies to fight this COVID-19 pandemic. Restoration of the function of NK cells in patients with pathogenic germline mutation(s) in the X-linked IL-2 receptor subunit gamma gene via allogeneic hematopoietic cell transplantation can lead to persistent remission of human-papillomavirus-related diseases [135]. In addition, the function of NK cells is associated with lifestyle [42]. Therefore, adherence to a healthy lifestyle, such as adequate sleep, moderate exercise, forest bathing, and listening to music, and the avoidance of cigarette smoking, alcohol consumption, stress, and obesity, will be favorable for the immunosurveillance of SARS-CoV-2 infection [42].
Additionally, NK-cell-based cancer immunotherapies are well established [136,137]. Sources of therapeutic NK cells are currently being tested clinically, such as autologous NK cells, haploidentical NK cells, umbilical cord blood NK cells, cytokine-induced memory-like NK cells, and chimeric antigen receptor (CAR)-NK cells. Methods designed to augment the cytotoxicity and longevity of NK cells are also under clinical investigation, including cytokine-based agents, NK-cell-engager molecules, and immune-checkpoint inhibitors [137]. Moreover, CAR-NK cells that target SARS-CoV-2 have been developed, and they show superior killing activity and cytokine production [138,139].
Clinical trials of NK cell therapy in COVID-19 patients have begun (Table 1) [36,38,140]. The early results of a Phase I/II clinical trial of NK cell (CYNK-001) therapy for COVID-19 patients (NCT04365101) have been reported recently [141]. CYNK-001 is a cryopreserved, allogeneic, off-the-shelf NK cell product derived from placental CD34+ cells. In the Phase I trial focusing on the safety of administration, patients will receive up to three CYNK-001 infusions. Four of the six patients treated to date have been evaluated at the time of reporting. In all four patients, the three infusions were well tolerated. In three of the four patients, oxygenation improved after the first infusion of CYNK-001 and radiographic improvement was noted. The remaining patient developed progressive hypoxemia prior to the administration of the first infusion of CYNK-001, requiring a larger amount of supplemental oxygen. The patient received three infusions of CYNK-001, but the amount of oxygen required increased. Twelve days after the first CYNK-001 infusion, the patient died of respiratory failure, and it was not possible to rule out the contribution of CYNK-001. The other three patients were discharged with an average follow-up of 16 days after the first infusion [141]. However, more data from clinical trials are required.
Although studies have shown compelling evidence of a less severe disease and lower hospitalization rates associated with Omicron variant infection [142], the high transmissibility of this variant might still impact healthcare system readiness and increase the absolute number of hospitalizations. Nonetheless, the response of NK cells to the Omicron variant is yet to be explored. However, NK-cell-based therapies could contribute to the rapid control and clearance of viral infection via the preserved ADCC mediated by NK cells after vaccination or prior infection [143], thereby attenuating disease severity.
In addition, vaccines and antiviral drugs are invaluable in attenuating infection and accelerating SARS-CoV-2 clearance, as well as lowering mortality caused by the Delta variant [144]. However, individuals with immunocompromised conditions (e.g., organ transplantation and immunodeficient patients) may not be able to mount an effective immune response. NK-cell-based therapies, especially CAR-NK cell therapy [138,139], may provide an alternative strategy for patients unresponsive to current protection and/or treatment strategies, in preparation for future pandemics.
Furthermore, the dysregulation of NK cells upon COVID-19 infection may be due to impaired type I IFN responses, such as type I interferon deficiency [75], or elevated levels of proinflammatory cytokines, such as IL-6 [27,73,75,78,83,84,89] and/or TGF-β [28]. Therefore, the identification of possible populations that respond to immunomodulators [145], such as treatment with INF-α or INF-β, or Tocilizumab, in combination with NK-cell-based therapies, is attractive. Nonetheless, this is yet to be explored.
The rationale of NK-cell-based therapies for COVID-19 has to be based on the timing of therapeutic interventions and disease severity, which is determined by the balance between beneficial antiviral and potential detrimental pathologic actions. NK-cell-based therapies would be more effective early in the infection in order to initiate effective immune responses to prevent the progression of the disease (Figure 2). Immunomodulation of NK cells towards regulatory function by crosstalk with myeloid cells, especially to counteract virus-infected monocytes and macrophages (which produce large amounts of pro-inflammatory cytokines and chemokines, contributing to local inflammation and a dangerous systemic inflammatory response [69]), could be useful as an adjunct therapy for progressive COVID-19, thereby attenuating disease severity.

7. Future Directions and Concluding Remarks

The rapid spread of SARS-CoV-2 and the unprecedented nature of COVID-19 has required an urgent response worldwide [2]. Prevention of infection, vaccines, and antiviral drugs are important for ending this COVID-19 pandemic. Extensive data accumulated thus far have underlined the fact that immune responses to SARS-CoV-2 infection play a deterministic role in shaping the clinical course of COVID-19 [14,15,16,17,18,19,20]. NK cells are important in the control of viral infections [24,56,57,58], including SARS-CoV-2 infection [27,28,101]. Notably, the SARS-CoV-2 clearance rate, antibody responses, and COVID-19 progression correlate with NK cell status [28,29,36,37], and NK cell dysfunction is linked with increased SARS-CoV-2 susceptibility [9,30,31,131,132,133]. Thus, NK cells function as a key element in the switch from effective to harmful immune responses in COVID-19. However, dysregulation of NK cells correlates with COVID-19 severity [26,27,28,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90], which perhaps causes disease progression. Hence, NK-cell-based therapies with sufficient antiviral capacities and a strong safety profiles in oncology may encourage the rapid application of these cells towards improving the management of COVID-19 by suppressing the virus, favoring finely tuned immune responses, and inhibiting fibrosis progression [27,28,38,101] (Table 1). Importantly, our knowledge of the role of NK cells in COVID-19 remains insufficient, such as the differences in their responses to different strains and variants, especially the current Delta and Omicron variants. Moreover, NK-cell-based therapies may potentiate the immunopathology of COVID-19 [70]. Thus, clear proof of protection, alongside increased knowledge regarding the pathology of NK cells in COVID-19, and differences in NK cell responses to different SARS-CoV-2 variants, is required. To this end, it is necessary to comprehensively study SARS-CoV-2-infected individuals with mild symptoms because they have effective immune responses, in addition to patients with severe COVID-19. It is important to study the early stages of SARS-CoV-2 infection to determine the interactions between infected cells and adjacent innate immune cells at primary sites of SARS-CoV-2 infection, lungs, and other tissues, since most studies of COVID-19 patients have been focused on peripheral blood. Additionally, models should help elucidate the role of NK cells in SARS-CoV-2 infection [146,147,148]. Such knowledge will guide us in how to modulate immune responses for maximum benefit and to improve the outcome of COVID-19 patients.

Author Contributions

Conceptualization, X.D. and H.T.; literature review, X.D.; writing—original draft preparation, X.D.; writing—review and editing, X.D., H.T. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

X.D., H.T. and M.N. hold patents for feeder-cell-free NK cell expansion culture technology and are stockholders of Biotherapy Institution of Japan Inc., a biotech startup company developing NK-cell-based immunotherapy and Mesenchymal Stromal Cell (MSC)-based products, such as adipose MSCs, MSC-derived conditioned medium (CM) concentrates, and extracellular vesicle (EV) isolates (StemSup®) for clinical research.

References

  1. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.M.; Wang, W.; Song, Z.G.; Hu, Y.; Tao, Z.W.; Tian, J.H.; Pei, Y.Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [Green Version]
  2. Novelli, G.; Biancolella, M.; Mehrian-Shai, R.; Erickson, C.; Godri Pollitt, K.J.; Vasiliou, V.; Watt, J.; Reichardt, J.K.V. COVID-19 update: The first 6 months of the pandemic. Hum. Genom. 2020, 14, 48. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  4. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  5. Yamagishi, T.; Kamiya, H.; Kakimoto, K.; Suzuki, M.; Wakita, T. Descriptive study of COVID-19 outbreak among passengers and crew on Diamond Princess cruise ship, Yokohama Port, Japan, 20 January to 9 February 2020. Eurosurveillance 2020, 25, 2000272. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  7. Cao, X. COVID-19: Immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020, 20, 269–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Richardson, S.; Hirsch, J.S.; Narasimhan, M.; Crawford, J.M.; McGinn, T.; Davidson, K.W.; The Northwell COVID-19 Research Consortium; Barnaby, D.P.; Becker, L.B.; Chelico, J.D.; et al. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA 2020, 323, 2052–2059. [Google Scholar] [CrossRef] [PubMed]
  9. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  10. NIH. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. 2022. Available online: www.covid19treamentguidelines.nih.gov (accessed on 24 March 2022).
  11. Chua, R.L.; Lukassen, S.; Trump, S.; Hennig, B.P.; Wendisch, D.; Pott, F.; Debnath, O.; Thürmann, L.; Kurth, F.; Völker, M.T.; et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 2020, 38, 970–979. [Google Scholar] [CrossRef]
  12. Brodin, P. Immune determinants of COVID-19 disease presentation and severity. Nat. Med. 2021, 27, 28–33. [Google Scholar] [CrossRef] [PubMed]
  13. Ahmadpoor, P.; Rostaing, L. Why the immune system fails to mount an adaptive immune response to a COVID-19 infection. Transpl. Int. 2020, 33, 824–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Vabret, N.; Britton, G.J.; Gruber, C.; Hegde, S.; Kim, J.; Kuksin, M.; Levantovsky, R.; Malle, L.; Moreira, A.; Park, M.D.; et al. Immunology of COVID-19: Current State of the Science. Immunity 2020, 52, 910–941. [Google Scholar] [CrossRef]
  15. Schultze, J.L.; Aschenbrenner, A.C. COVID-19 and the human innate immune system. Cell 2021, 184, 1671–1692. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef] [Green Version]
  17. Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef] [PubMed]
  18. Shaath, H.; Vishnubalaji, R.; Elkord, E.; Alajez, N.M. Single-Cell Transcriptome Analysis Highlights a Role for Neutrophils and Inflammatory Macrophages in the Pathogenesis of Severe COVID-19. Cells 2020, 9, 2374. [Google Scholar] [CrossRef]
  19. Ren, X.; Wen, W.; Fan, X.; Hou, W.; Su, B.; Cai, P.; Li, J.; Liu, Y.; Tang, F.; Zhang, F.; et al. COVID-19 immune features revealed by a large-scale single-cell transcriptome atlas. Cell 2021, 184, 1895–1913.e19. [Google Scholar] [CrossRef]
  20. Melms, J.C.; Biermann, J.; Huang, H.; Wang, Y.; Nair, A.; Tagore, S.; Katsyv, I.; Rendeiro, A.F.; Amin, A.D.; Schapiro, D.; et al. A molecular single-cell lung atlas of lethal COVID-19. Nature 2021, 595, 114–119. [Google Scholar] [CrossRef]
  21. Rodrigues, P.R.S.; Alrubayyi, A.; Pring, E.; Bart, V.M.T.; Jones, R.; Coveney, C.; Lu, F.; Tellier, M.; Maleki-Toyserkani, S.; Richter, F.C.; et al. Innate immunology in COVID-19-a living review. Part II: Dysregulated inflammation drives immunopathology. Oxf Open Immunol. 2020, 1, iqaa005. [Google Scholar] [CrossRef]
  22. Coveney, C.; Tellier, M.; Lu, F.; Maleki-Toyserkani, S.; Jones, R.; Bart, V.M.T.; Pring, E.; Alrubayyi, A.; Richter, F.C.; Scourfield, D.O.; et al. Innate immunology in COVID-19-a living review. Part I: Viral entry, sensing and evasion. Oxf. Open Immunol. 2020, 1, iqaa004. [Google Scholar] [CrossRef] [PubMed]
  23. D’Elia, R.V.; Harrison, K.; Oyston, P.C.; Lukaszewski, R.A.; Clark, G.C. Targeting the “cytokine storm” for therapeutic benefit. Clin. Vaccine Immunol. 2013, 20, 319–327. [Google Scholar] [CrossRef] [PubMed]
  24. Waggoner, S.N.; Reighard, S.D.; Gyurova, I.E.; Cranert, S.A.; Mahl, S.E.; Karmele, E.P.; McNally, J.P.; Moran, M.T.; Brooks, T.R.; Yaqoob, F.; et al. Roles of natural killer cells in antiviral immunity. Curr. Opin. Virol. 2016, 16, 15–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yu, J.; Tostanoski, L.H.; Peter, L.; Mercado, N.B.; McMahan, K.; Mahrokhian, S.H.; Nkolola, J.P.; Liu, J.; Li, Z.; Chandrashekar, A.; et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science 2020, 369, 806–811. [Google Scholar] [CrossRef]
  26. Herrera, L.; Martin-Inaraja, M.; Santos, S.; Inglés-Ferrándiz, M.; Azkarate, A.; Perez-Vaquero, M.A.; Vesga, M.A.; Vicario, J.L.; Soria, B.; Solano, C.; et al. Identifying SARS-CoV-2 ‘memory’ NK cells from COVID-19 convalescent donors for adoptive cell therapy. Immunology 2022, 165, 234–249. [Google Scholar] [CrossRef]
  27. Krämer, B.; Knoll, R.; Bonaguro, L.; ToVinh, M.; Raabe, J.; Astaburuaga-García, R.; Schulte-Schrepping, J.; Kaiser, K.M.; Rieke, G.J.; Bischoff, J.; et al. Early IFN-α signatures and persistent dysfunction are distinguishing features of NK cells in severe COVID-19. Immunity 2021, 54, 2650–2669.e14. [Google Scholar] [CrossRef]
  28. Witkowski, M.; Tizian, C.; Ferreira-Gomes, M.; Niemeyer, D.; Jones, T.C.; Heinrich, F.; Frischbutter, S.; Angermair, S.; Hohnstein, T.; Mattiola, I.; et al. Untimely TGFβ responses in COVID-19 limit antiviral functions of NK cells. Nature 2021, 600, 295–301. [Google Scholar] [CrossRef]
  29. Hsieh, W.C.; Lai, E.Y.; Liu, Y.T.; Wang, Y.F.; Tzeng, Y.S.; Cui, L.; Lai, Y.J.; Huang, H.C.; Huang, J.H.; Ni, H.C.; et al. NK cell receptor and ligand composition influences the clearance of SARS-CoV-2. J. Clin. Investig. 2021, 131, e146408. [Google Scholar] [CrossRef]
  30. Vietzen, H.; Zoufaly, A.; Traugott, M.; Aberle, J.; Aberle, S.W.; Puchhammer-Stöckl, E. Deletion of the NKG2C receptor encoding KLRC2 gene and HLA-E variants are risk factors for severe COVID-19. Genet. Med. 2021, 23, 963–967. [Google Scholar] [CrossRef]
  31. Littera, R.; Chessa, L.; Deidda, S.; Angioni, G.; Campagna, M.; Lai, S.; Melis, M.; Cipri, S.; Firinu, D.; Santus, S.; et al. Natural killer-cell immunoglobulin-like receptors trigger differences in immune response to SARS-CoV-2 infection. PLoS ONE 2021, 16, e0255608. [Google Scholar] [CrossRef]
  32. Saresella, M.; Trabattoni, D.; Marventano, I.; Piancone, F.; La Rosa, F.; Caronni, A.; Lax, A.; Bianchi, L.; Banfi, P.; Navarro, J.; et al. NK Cell Subpopulations and Receptor Expression in Recovering SARS-CoV-2 Infection. Mol. Neurobiol. 2021, 58, 6111–6120. [Google Scholar] [CrossRef] [PubMed]
  33. Casado, J.L.; Moraga, E.; Vizcarra, P.; Velasco, H.; Martín-Hondarza, A.; Haemmerle, J.; Gómez, S.; Quereda, C.; Vallejo, A. Expansion of CD56dimCD16neg NK Cell Subset and Increased Inhibitory KIRs in Hospitalized COVID-19 Patients. Viruses 2021, 14, 46. [Google Scholar] [CrossRef]
  34. Leem, G.; Cheon, S.; Lee, H.; Choi, S.J.; Jeong, S.; Kim, E.S.; Jeong, H.W.; Jeong, H.; Park, S.H.; Kim, Y.S.; et al. Abnormality in the NK-cell population is prolonged in severe COVID-19 patients. J. Allergy Clin. Immunol. 2021, 148, 996–1006.e18. [Google Scholar] [CrossRef] [PubMed]
  35. Bernal, E.; Gimeno, L.; Alcaraz, M.J.; Quadeer, A.A.; Moreno, M.; Martínez-Sánchez, M.V.; Campillo, J.A.; Gomez, J.M.; Pelaez, A.; García, E.; et al. Activating Killer-Cell Immunoglobulin-Like Receptors Are Associated With the Severity of Coronavirus Disease 2019. J. Infect. Dis. 2021, 224, 229–240. [Google Scholar] [CrossRef] [PubMed]
  36. Bao, C.; Tao, X.; Cui, W.; Hao, Y.; Zheng, S.; Yi, B.; Pan, T.; Young, K.H.; Qian, W. Natural killer cells associated with SARS-CoV-2 viral RNA shedding, antibody response and mortality in COVID-19 patients. Exp. Hematol. Oncol. 2021, 10, 5. [Google Scholar] [CrossRef] [PubMed]
  37. Rezaei, M.; Mahmoudi, S.; Mortaz, E.; Marjani, M. Immune cell profiling and antibody responses in patients with COVID-19. BMC Infect. Dis. 2021, 21, 646. [Google Scholar] [CrossRef] [PubMed]
  38. Market, M.; Angka, L.; Martel, A.B.; Bastin, D.; Olanubi, O.; Tennakoon, G.; Boucher, D.M.; Ng, J.; Ardolino, M.; Auer, R.C. Flattening the COVID-19 Curve With Natural Killer Cell Based Immunotherapies. Front. Immunol. 2020, 11, 1512. [Google Scholar] [CrossRef] [PubMed]
  39. Scharenberg, M.; Vangeti, S.; Kekäläinen, E.; Bergman, P.; Al-Ameri, M.; Johansson, N.; Sondén, K.; Falck-Jones, S.; Färnert, A.; Ljunggren, H.G.; et al. Influenza A Virus Infection Induces Hyperresponsiveness in Human Lung Tissue-Resident and Peripheral Blood NK Cells. Front. Immunol. 2019, 10, 1116. [Google Scholar] [CrossRef] [Green Version]
  40. Ghosh, S.; Nandi, M.; Pal, S.; Mukhopadhyay, D.; Chakraborty, B.C.; Khatun, M.; Bhowmick, D.; Mondal, R.K.; Das, S.; Das, K.; et al. Natural killer cells contribute to hepatic injury and help in viral persistence during progression of hepatitis B e-antigen-negative chronic hepatitis B virus infection. Clin. Microbiol. Infect. 2016, 22, 733.e9–733.e19. [Google Scholar] [CrossRef] [Green Version]
  41. Terunuma, H.; Deng, X.; Dewan, Z.; Fujimoto, S.; Yamamoto, N. Potential role of NK cells in the induction of immune responses: Implications for NK cell-based immunotherapy for cancers and viral infections. Int. Rev. Immunol. 2008, 27, 93–110. [Google Scholar] [CrossRef]
  42. Deng, X.; Terunuma, H.; Nieda, M. Immunosurveillance of cancer and viral infections with regard to alterations of human NK cells originating from lifestyle and aging. Biomedicines 2021, 9, 557. [Google Scholar] [CrossRef] [PubMed]
  43. Waggoner, S.N.; Cornberg, M.; Selin, L.K.; Welsh, R.M. Natural killer cells act as rheostats modulating antiviral T cells. Nature 2011, 481, 394–398. [Google Scholar] [CrossRef] [PubMed]
  44. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
  45. Lanier, L.L. Up on the tightrope: Natural killer cell activation and inhibition. Nat. Immunol. 2008, 9, 495–502. [Google Scholar] [CrossRef] [PubMed]
  46. Long, E.O.; Kim, H.S.; Liu, D.; Peterson, M.E.; Rajagopalan, S. Controlling natural killer cell responses: Integration of signals for activation and inhibition. Annu. Rev. Immunol. 2013, 31, 227–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Van Eeden, C.; Khan, L.; Osman, M.S.; Cohen Tervaert, J.W. Natural Killer Cell Dysfunction and Its Role in COVID-19. Int. J. Mol. Sci. 2020, 21, 6351. [Google Scholar] [CrossRef]
  48. Deng, X.; Terunuma, H.; Nieda, M.; Xiao, W.; Nicol, A. Synergistic cytotoxicity of ex vivo expanded natural killer cells in combination with monoclonal antibody drugs against cancer cells. Int. Immunopharmacol. 2012, 14, 593–605. [Google Scholar] [CrossRef]
  49. Cooper, M.A.; Fehniger, T.A.; Caligiuri, M.A. The biology of human natural killer-cell subsets. Trends Immunol. 2001, 22, 633–640. [Google Scholar] [CrossRef]
  50. Fehniger, T.A.; Cooper, M.A.; Nuovo, G.J.; Cella, M.; Facchetti, F.; Colonna, M.; Caligiuri, M.A. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: A potential new link between adaptive and innate immunity. Blood 2003, 101, 3052–3057. [Google Scholar] [CrossRef] [Green Version]
  51. Leibson, P.J. Signal transduction during natural killer cell activation: Inside the mind of a killer. Immunity 1997, 6, 655–661. [Google Scholar] [CrossRef] [Green Version]
  52. Michel, T.; Poli, A.; Cuapio, A.; Briquemont, B.; Iserentant, G.; Ollert, M.; Zimmer, J. Human CD56bright NK Cells: An Update. J. Immunol. 2016, 196, 2923–2931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hervier, B.; Russick, J.; Cremer, I.; Vieillard, V. NK Cells in the Human Lungs. Front. Immunol. 2019, 10, 1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Cooper, G.E.; Ostridge, K.; Khakoo, S.I.; Wilkinson, T.M.A.; Staples, K.J. Human CD49a+ Lung Natural Killer Cell Cytotoxicity in Response to Influenza A Virus. Front. Immunol. 2018, 9, 1671. [Google Scholar] [CrossRef]
  55. Marquardt, N.; Kekäläinen, E.; Chen, P.; Kvedaraite, E.; Wilson, J.N.; Ivarsson, M.A.; Mjösberg, J.; Berglin, L.; Säfholm, J.; Manson, M.L.; et al. Human lung natural killer cells are predominantly comprised of highly differentiated hypofunctional CD69CD56dim cells. J. Allergy Clin. Immunol. 2017, 139, 1321–1330.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Brandstadter, J.D.; Yang, Y. Natural killer cell responses to viral infection. J. Innate Immun. 2011, 3, 274–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Jost, S.; Altfeld, M. Control of human viral infections by natural killer cells. Annu. Rev. Immunol. 2013, 31, 163–194. [Google Scholar] [CrossRef]
  59. Biron, C.A.; Byron, K.S.; Sullivan, J.L. Severe herpesvirus infections in an adolescent without natural killer cells. N. Engl. J. Med. 1989, 320, 1731–1735. [Google Scholar] [CrossRef]
  60. Duev-Cohen, A.; Bar-On, Y.; Glasner, A.; Berhani, O.; Ophir, Y.; Levi-Schaffer, F.; Mandelboim, M.; Mandelboim, O. The human 2B4 and NTB-A receptors bind the influenza viral hemagglutinin and co-stimulate NK cell cytotoxicity. Oncotarget 2016, 7, 13093–13105. [Google Scholar] [CrossRef] [Green Version]
  61. Wiertz, E.J.; Mukherjee, S.; Ploegh, H.L. Viruses use stealth technology to escape from the host immune system. Mol. Med. Today 1997, 3, 116–123. [Google Scholar] [CrossRef]
  62. McMichael, A. T cell responses and viral escape. Cell 1998, 93, 673–676. [Google Scholar] [CrossRef] [Green Version]
  63. Adib-Conquy, M.; Scott-Algara, D.; Cavaillon, J.M.; Souza-Fonseca-Guimaraes, F. TLR-mediated activation of NK cells and their role in bacterial/viral immune responses in mammals. Immunol. Cell Biol. 2014, 92, 256–262. [Google Scholar] [CrossRef]
  64. Hammer, Q.; Rückert, T.; Borst, E.M.; Dunst, J.; Haubner, A.; Durek, P.; Heinrich, F.; Gasparoni, G.; Babic, M.; Tomic, A.; et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018, 19, 453–463. [Google Scholar] [CrossRef] [PubMed]
  65. Della Chiesa, M.; De Maria, A.; Muccio, L.; Bozzano, F.; Sivori, S.; Moretta, L. Human NK Cells and Herpesviruses: Mechanisms of Recognition, Response and Adaptation. Front. Microbiol. 2019, 10, 2297. [Google Scholar] [CrossRef] [PubMed]
  66. Nguyen, K.B.; Salazar-Mather, T.P.; Dalod, M.Y.; Van Deusen, J.B.; Wei, X.Q.; Liew, F.Y.; Caligiuri, M.A.; Durbin, J.E.; Biron, C.A. Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 2002, 169, 4279–4287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Marcenaro, E.; Carlomagno, S.; Pesce, S.; Moretta, A.; Sivori, S. NK/DC crosstalk in anti-viral response. Adv. Exp. Med. Biol. 2012, 946, 295–308. [Google Scholar] [CrossRef] [PubMed]
  68. Molgora, M.; Supino, D.; Mavilio, D.; Santoni, A.; Moretta, L.; Mantovani, A.; Garlanda, C. The yin-yang of the interaction between myelomonocytic cells and NK cells. Scand J. Immunol. 2018, 88, e12705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Jafarzadeh, A.; Chauhan, P.; Saha, B.; Jafarzadeh, S.; Nemati, M. Contribution of monocytes and macrophages to the local tissue inflammation and cytokine storm in COVID-19: Lessons from SARS and MERS, and potential therapeutic interventions. Life Sci. 2020, 257, 118102. [Google Scholar] [CrossRef]
  70. Masselli, E.; Vaccarezza, M.; Carubbi, C.; Pozzi, G.; Presta, V.; Mirandola, P.; Vitale, M. NK cells: A double edge sword against SARS-CoV-2. Adv. Biol. Regul. 2020, 77, 100737. [Google Scholar] [CrossRef]
  71. Zheng, M.; Gao, Y.; Wang, G.; Song, G.; Liu, S.; Sun, D.; Xu, Y.; Tian, Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell. Mol. Immunol. 2020, 17, 533–535. [Google Scholar] [CrossRef] [Green Version]
  72. Osman, M.; Faridi, R.M.; Sligl, W.; Shabani-Rad, M.T.; Dharmani-Khan, P.; Parker, A.; Kalra, A.; Tripathi, M.B.; Storek, J.; Cohen Tervaert, J.W.; et al. Impaired natural killer cell counts and cytolytic activity in patients with severe COVID-19. Blood Adv. 2020, 4, 5035–5039. [Google Scholar] [CrossRef] [PubMed]
  73. Mazzoni, A.; Salvati, L.; Maggi, L.; Capone, M.; Vanni, A.; Spinicci, M.; Mencarini, J.; Caporale, R.; Peruzzi, B.; Antonelli, A.; et al. Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J. Clin. Investig. 2020, 130, 4694–4703. [Google Scholar] [CrossRef] [PubMed]
  74. Li, M.; Guo, W.; Dong, Y.; Wang, X.; Dai, D.; Liu, X.; Wu, Y.; Li, M.; Zhang, W.; Zhou, H.; et al. Elevated Exhaustion Levels of NK and CD8(+) T Cells as Indicators for Progression and Prognosis of COVID-19 Disease. Front. Immunol. 2020, 11, 580237. [Google Scholar] [CrossRef] [PubMed]
  75. Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Péré, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
  76. Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L.; Song, S.; Ma, Z.; Mo, P.; Zhang, Y. Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia. J. Infect. Dis. 2020, 221, 1762–1769. [Google Scholar] [CrossRef] [Green Version]
  77. Maucourant, C.; Filipovic, I.; Ponzetta, A.; Aleman, S.; Cornillet, M.; Hertwig, L.; Strunz, B.; Lentini, A.; Reinius, B.; Brownlie, D.; et al. Natural killer cell immunotypes related to COVID-19 disease severity. Sci. Immunol. 2020, 5, eabd6832. [Google Scholar] [CrossRef]
  78. Chen, X.; Huang, J.; Huang, Y.; Chen, J.; Huang, Y.; Jiang, X.; Shi, Y. Characteristics of immune cells and cytokines in patients with coronavirus disease 2019 in Guangzhou, China. Hum. Immunol. 2020, 81, 702–708. [Google Scholar] [CrossRef]
  79. Varchetta, S.; Mele, D.; Oliviero, B.; Mantovani, S.; Ludovisi, S.; Cerino, A.; Bruno, R.; Castelli, A.; Mosconi, M.; Vecchia, M.; et al. Unique immunological profile in patients with COVID-19. Cell. Mol. Immunol. 2021, 18, 604–612. [Google Scholar] [CrossRef]
  80. Jiang, Y.; Wei, X.; Guan, J.; Qin, S.; Wang, Z.; Lu, H.; Qian, J.; Wu, L.; Chen, Y.; Chen, Y.; et al. COVID-19 pneumonia: CD8(+) T and NK cells are decreased in number but compensatory increased in cytotoxic potential. Clin. Immunol. 2020, 218, 108516. [Google Scholar] [CrossRef]
  81. Demaria, O.; Carvelli, J.; Batista, L.; Thibult, M.L.; Morel, A.; André, P.; Morel, Y.; Vély, F.; Vivier, E. Identification of druggable inhibitory immune checkpoints on Natural Killer cells in COVID-19. Cell. Mol. Immunol. 2020, 17, 995–997. [Google Scholar] [CrossRef]
  82. Carsetti, R.; Zaffina, S.; Piano Mortari, E.; Terreri, S.; Corrente, F.; Capponi, C.; Palomba, P.; Mirabella, M.; Cascioli, S.; Palange, P.; et al. Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases. Front. Immunol. 2020, 11, 610300. [Google Scholar] [CrossRef] [PubMed]
  83. Song, C.Y.; Xu, J.; He, J.Q.; Lu, Y.Q. Immune dysfunction following COVID-19, especially in severe patients. Sci. Rep. 2020, 10, 15838. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, Y.; Huang, X.; Sun, J.; Xie, T.; Lei, Y.; Muhammad, J.; Li, X.; Zeng, X.; Zhou, F.; Qin, H.; et al. Clinical Characteristics and Immune Injury Mechanisms in 71 Patients with COVID-19. mSphere 2020, 5, e00362-20. [Google Scholar] [CrossRef] [PubMed]
  85. Song, J.W.; Zhang, C.; Fan, X.; Meng, F.P.; Xu, Z.; Xia, P.; Cao, W.J.; Yang, T.; Dai, X.P.; Wang, S.Y.; et al. Immunological and inflammatory profiles in mild and severe cases of COVID-19. Nat. Commun. 2020, 11, 3410. [Google Scholar] [CrossRef] [PubMed]
  86. Bozzano, F.; Dentone, C.; Perrone, C.; Di Biagio, A.; Fenoglio, D.; Parodi, A.; Mikulska, M.; Bruzzone, B.; Giacobbe, D.R.; Vena, A.; et al. Extensive activation, tissue trafficking, turnover and functional impairment of NK cells in COVID-19 patients at disease onset associates with subsequent disease severity. PLoS Pathog. 2021, 17, e1009448. [Google Scholar] [CrossRef] [PubMed]
  87. Wilk, A.J.; Rustagi, A.; Zhao, N.Q.; Roque, J.; Martínez-Colón, G.J.; McKechnie, J.L.; Ivison, G.T.; Ranganath, T.; Vergara, R.; Hollis, T.; et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26, 1070–1076. [Google Scholar] [CrossRef]
  88. Bouadma, L.; Wiedemann, A.; Patrier, J.; Surénaud, M.; Wicky, P.H.; Foucat, E.; Diehl, J.L.; Hejblum, B.P.; Sinnah, F.; de Montmollin, E.; et al. Immune Alterations in a Patient with SARS-CoV-2-Related Acute Respiratory Distress Syndrome. J. Clin. Immunol. 2020, 40, 1082–1092. [Google Scholar] [CrossRef]
  89. Zenarruzabeitia, O.; Astarloa-Pando, G.; Terrén, I.; Orrantia, A.; Pérez-Garay, R.; Seijas-Betolaza, I.; Nieto-Arana, J.; Imaz-Ayo, N.; Pérez-Fernández, S.; Arana-Arri, E.; et al. T Cell Activation, Highly Armed Cytotoxic Cells and a Shift in Monocytes CD300 Receptors Expression Is Characteristic of Patients With Severe COVID-19. Front. Immunol. 2021, 12, 655934. [Google Scholar] [CrossRef]
  90. Wilk, A.J.; Lee, M.J.; Wei, B.; Parks, B.; Pi, R.; Martínez-Colón, G.J.; Ranganath, T.; Zhao, N.Q.; Taylor, S.; Becker, W.; et al. Multi-omic profiling reveals widespread dysregulation of innate immunity and hematopoiesis in COVID-19. J. Exp. Med. 2021, 218, e20210582. [Google Scholar] [CrossRef]
  91. Jewett, A. The Potential Effect of Novel Coronavirus SARS-CoV-2 on NK Cells; A Perspective on Potential Therapeutic Interventions. Front. Immunol. 2020, 11, 1692. [Google Scholar] [CrossRef]
  92. Xiong, Y.; Liu, Y.; Cao, L.; Wang, D.; Guo, M.; Jiang, A.; Guo, D.; Hu, W.; Yang, J.; Tang, Z.; et al. Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg. Microbes Infect. 2020, 9, 761–770. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, C.; Wang, X.M.; Li, S.R.; Twelkmeyer, T.; Wang, W.H.; Zhang, S.Y.; Wang, S.F.; Chen, J.Z.; Jin, X.; Wu, Y.Z.; et al. NKG2A is a NK cell exhaustion checkpoint for HCV persistence. Nat. Commun. 2019, 10, 1507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zambello, R.; Barilà, G.; Manni, S.; Piazza, F.; Semenzato, G. NK cells and CD38: Implication for (Immuno)Therapy in Plasma Cell Dyscrasias. Cells 2020, 9, 768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Erokhina, S.A.; Streltsova, M.A.; Kanevskiy, L.M.; Grechikhina, M.V.; Sapozhnikov, A.M.; Kovalenko, E.I. HLA-DR-expressing NK cells: Effective killers suspected for antigen presentation. J. Leukoc. Biol. 2021, 109, 327–337. [Google Scholar] [CrossRef] [PubMed]
  96. Moretta, A.; Poggi, A.; Pende, D.; Tripodi, G.; Orengo, A.M.; Pella, N.; Augugliaro, R.; Bottino, C.; Ciccone, E.; Moretta, L. CD69-mediated pathway of lymphocyte activation: Anti-CD69 monoclonal antibodies trigger the cytolytic activity of different lymphoid effector cells with the exception of cytolytic T lymphocytes expressing T cell receptor alpha/beta. J. Exp. Med. 1991, 174, 1393–1398. [Google Scholar] [CrossRef] [Green Version]
  97. Romagnani, C.; Della Chiesa, M.; Kohler, S.; Moewes, B.; Radbruch, A.; Moretta, L.; Moretta, A.; Thiel, A. Activation of human NK cells by plasmacytoid dendritic cells and its modulation by CD4+ T helper cells and CD4+ CD25hi T regulatory cells. Eur. J. Immunol. 2005, 35, 2452–2458. [Google Scholar] [CrossRef]
  98. Marquardt, N.; Kekäläinen, E.; Chen, P.; Lourda, M.; Wilson, J.N.; Scharenberg, M.; Bergman, P.; Al-Ameri, M.; Hård, J.; Mold, J.E.; et al. Unique transcriptional and protein-expression signature in human lung tissue-resident NK cells. Nat. Commun. 2019, 10, 3841. [Google Scholar] [CrossRef] [Green Version]
  99. Ogawa, K.; Tanaka, K.; Ishii, A.; Nakamura, Y.; Kondo, S.; Sugamura, K.; Takano, S.; Nakamura, M.; Nagata, K. A novel serum protein that is selectively produced by cytotoxic lymphocytes. J. Immunol. 2001, 166, 6404–6412. [Google Scholar] [CrossRef] [Green Version]
  100. 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. [Google Scholar] [CrossRef]
  101. Hammer, Q.; Dunst, J.; Christ, W.; Picarazzi, F.; Wendorff, M.; Momayyezi, P.; Huhn, O.; Netskar, H.K.; Maleki, K.T.; García, M.; et al. SARS-CoV-2 Nsp13 encodes for an HLA-E-stabilizing peptide that abrogates inhibition of NKG2A-expressing NK cells. Cell Rep. 2022, 21, 110503. [Google Scholar] [CrossRef]
  102. Li, F.; Wei, H.; Wei, H.; Gao, Y.; Xu, L.; Yin, W.; Sun, R.; Tian, Z. Blocking the natural killer cell inhibitory receptor NKG2A increases activity of human natural killer cells and clears hepatitis B virus infection in mice. Gastroenterology 2013, 144, 392–401. [Google Scholar] [CrossRef] [PubMed]
  103. Wijaya, R.S.; Read, S.A.; Schibeci, S.; Han, S.; Azardaryany, M.K.; van der Poorten, D.; Lin, R.; Yuen, L.; Lam, V.; Douglas, M.W.; et al. Expansion of dysfunctional CD56-CD16+ NK cells in chronic hepatitis B patients. Liver Int. 2021, 41, 969–981. [Google Scholar] [CrossRef] [PubMed]
  104. Finney, C.A.; Ayi, K.; Wasmuth, J.D.; Sheth, P.M.; Kaul, R.; Loutfy, M.; Kain, K.C.; Serghides, L. HIV infection deregulates Tim-3 expression on innate cells: Combination antiretroviral therapy results in partial restoration. J. Acquir. Immune Defic. Syndr. 2013, 63, 161–167. [Google Scholar] [CrossRef] [PubMed]
  105. Norris, S.; Coleman, A.; Kuri-Cervantes, L.; Bower, M.; Nelson, M.; Goodier, M.R. PD-1 expression on natural killer cells and CD8(+) T cells during chronic HIV-1 infection. Viral Immunol. 2012, 25, 329–332. [Google Scholar] [CrossRef]
  106. Dierks, P.; Wroblewski, R.; Eberhard, J.M.; Martrus, G.; Degen, O.; Hertling, S.; Schmiedel, S.; Lunemann, S.; Hüfner, A.; Lohse, A.W.; et al. Brief Report: Increased Frequency of CD39+ CD56bright Natural Killer Cells in HIV-1 Infection Correlates With Immune Activation and Disease Progression. J. Acquir. Immune Defic. Syndr. 2017, 74, 467–472. [Google Scholar] [CrossRef]
  107. Parolini, S.; Bottino, C.; Falco, M.; Augugliaro, R.; Giliani, S.; Franceschini, R.; Ochs, H.D.; Wolf, H.; Bonnefoy, J.Y.; Biassoni, R.; et al. X-linked lymphoproliferative disease. 2B4 molecules displaying inhibitory rather than activating function are responsible for the inability of natural killer cells to kill Epstein-Barr virus-infected cells. J. Exp. Med. 2000, 192, 337–346. [Google Scholar] [CrossRef]
  108. Oliviero, B.; Mantovani, S.; Varchetta, S.; Mele, D.; Grossi, G.; Ludovisi, S.; Nuti, E.; Rossello, A.; Mondelli, M.U. Hepatitis C virus-induced NK cell activation causes metzincin-mediated CD16 cleavage and impaired antibody-dependent cytotoxicity. J. Hepatol. 2017, 66, 1130–1137. [Google Scholar] [CrossRef]
  109. Desimio, M.G.; Covino, D.A.; Doria, M. Potential of the NKG2D/NKG2DL Axis in NK Cell-Mediated Clearance of the HIV-1 Reservoir. Int. J. Mol. Sci. 2019, 20, 4490. [Google Scholar] [CrossRef] [Green Version]
  110. Varchetta, S.; Mele, D.; Lombardi, A.; Oliviero, B.; Mantovani, S.; Tinelli, C.; Spreafico, M.; Prati, D.; Ludovisi, S.; Ferraioli, G.; et al. Lack of Siglec-7 expression identifies a dysfunctional natural killer cell subset associated with liver inflammation and fibrosis in chronic HCV infection. Gut 2016, 65, 1998–2006. [Google Scholar] [CrossRef]
  111. Cifaldi, L.; Doria, M.; Cotugno, N.; Zicari, S.; Cancrini, C.; Palma, P.; Rossi, P. DNAM-1 Activating Receptor and Its Ligands: How Do Viruses Affect the NK Cell-Mediated Immune Surveillance during the Various Phases of Infection? Int. J. Mol. Sci. 2019, 20, 3715. [Google Scholar] [CrossRef] [Green Version]
  112. Romee, R.; Foley, B.; Lenvik, T.; Wang, Y.; Zhang, B.; Ankarlo, D.; Luo, X.; Cooley, S.; Verneris, M.; Walcheck, B.; et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 2013, 121, 3599–3608. [Google Scholar] [CrossRef] [PubMed]
  113. Lopez-Vergès, S.; Milush, J.M.; Schwartz, B.S.; Pando, M.J.; Jarjoura, J.; York, V.A.; Houchins, J.P.; Miller, S.; Kang, S.M.; Norris, P.J.; et al. Expansion of a unique CD57⁺NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc. Natl. Acad. Sci. USA 2011, 108, 14725–14732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Trapani, J.A.; Smyth, M.J. Functional significance of the perforin/granzyme cell death pathway. Nat. Rev. Immunol. 2002, 2, 735–747. [Google Scholar] [CrossRef] [PubMed]
  115. Ramos da Silva, S.; Ju, E.; Meng, W.; Paniz Mondolfi, A.E.; Dacic, S.; Green, A.; Bryce, C.; Grimes, Z.; Fowkes, M.; Sordillo, E.M.; et al. Broad Severe Acute Respiratory Syndrome Coronavirus 2 Cell Tropism and Immunopathology in Lung Tissues from Fatal Coronavirus Disease 2019. J. Infect. Dis. 2021, 223, 1842–1854. [Google Scholar] [CrossRef]
  116. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef]
  117. Gill, N.; Chenoweth, M.J.; Verdu, E.F.; Ashkar, A.A. NK cells require type I IFN receptor for antiviral responses during genital HSV-2 infection. Cell. Immunol. 2011, 269, 29–37. [Google Scholar] [CrossRef]
  118. Pekayvaz, K.; Leunig, A.; Kaiser, R.; Joppich, M.; Brambs, S.; Janjic, A.; Popp, O.; Nixdorf, D.; Fumagalli, V.; Schmidt, N.; et al. Protective immune trajectories in early viral containment of non-pneumonic SARS-CoV-2 infection. Nat. Commun. 2022, 13, 1018. [Google Scholar] [CrossRef]
  119. Cifaldi, L.; Prencipe, G.; Caiello, I.; Bracaglia, C.; Locatelli, F.; De Benedetti, F.; Strippoli, R. Inhibition of natural killer cell cytotoxicity by interleukin-6: Implications for the pathogenesis of macrophage activation syndrome. Arthritis Rheumatol. 2015, 67, 3037–3046. [Google Scholar] [CrossRef]
  120. Sahoo, D.; Katkar, G.D.; Khandelwal, S.; Behroozikhah, M.; Claire, A.; Castillo, V.; Tindle, C.; Fuller, M.; Taheri, S.; Rogers, T.F.; et al. AI-guided discovery of the invariant host response to viral pandemics. EBioMedicine 2021, 68, 103390. [Google Scholar] [CrossRef]
  121. Liu, C.; Martins, A.J.; Lau, W.W.; Rachmaninoff, N.; Chen, J.; Imberti, L.; Mostaghimi, D.; Fink, D.L.; Burbelo, P.D.; Dobbs, K.; et al. Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19. Cell 2021, 184, 1836–1857.e22. [Google Scholar] [CrossRef]
  122. Rodda, L.B.; Netland, J.; Shehata, L.; Pruner, K.B.; Morawski, P.A.; Thouvenel, C.D.; Takehara, K.K.; Eggenberger, J.; Hemann, E.A.; Waterman, H.R.; et al. Functional SARS-CoV-2-Specific Immune Memory Persists after Mild COVID-19. Cell 2021, 184, 169–183.e17. [Google Scholar] [CrossRef]
  123. Braciale, T.J.; Sun, J.; Kim, T.S. Regulating the adaptive immune response to respiratory virus infection. Nat. Rev. Immunol. 2012, 12, 295–305. [Google Scholar] [CrossRef] [PubMed]
  124. Newton, A.H.; Cardani, A.; Braciale, T.J. The host immune response in respiratory virus infection: Balancing virus clearance and immunopathology. Semin. Immunopathol. 2016, 38, 471–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rydyznski Moderbacher, C.; Ramirez, S.I.; Dan, J.M.; Grifoni, A.; Hastie, K.M.; Weiskopf, D.; Belanger, S.; Abbott, R.K.; Kim, C.; Choi, J.; et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 2020, 183, 996–1012.e19. [Google Scholar] [CrossRef] [PubMed]
  126. Pachlopnik Schmid, J.; Côte, M.; Ménager, M.M.; Burgess, A.; Nehme, N.; Ménasché, G.; Fischer, A.; de Saint Basile, G. Inherited defects in lymphocyte cytotoxic activity. Immunol. Rev. 2010, 235, 10–23. [Google Scholar] [CrossRef] [PubMed]
  127. Van Dommelen, S.L.; Sumaria, N.; Schreiber, R.D.; Scalzo, A.A.; Smyth, M.J.; Degli-Esposti, M.A. Perforin and granzymes have distinct roles in defensive immunity and immunopathology. Immunity 2006, 25, 835–848. [Google Scholar] [CrossRef] [Green Version]
  128. Schulte-Schrepping, J.; Reusch, N.; Paclik, D.; Baßler, K.; Schlickeiser, S.; Zhang, B.; Krämer, B.; Krammer, T.; Brumhard, S.; Bonaguro, L.; et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020, 182, 1419–1440.e23. [Google Scholar] [CrossRef]
  129. Chen, H.; Liu, W.; Wang, Y.; Liu, D.; Zhao, L.; Yu, J. SARS-CoV-2 activates lung epithelial cell proinflammatory signaling and leads to immune dysregulation in COVID-19 patients. EBioMedicine 2021, 70, 103500. [Google Scholar] [CrossRef]
  130. Dewan, M.Z.; Takada, M.; Terunuma, H.; Deng, X.; Ahmed, S.; Yamamoto, N.; Toi, M. Natural killer activity of peripheral-blood mononuclear cells in breast cancer patients. Biomed. Pharmacother. 2009, 63, 703–706. [Google Scholar] [CrossRef]
  131. Zhang, L.; Zhu, F.; Xie, L.; Wang, C.; Wang, J.; Chen, R.; Jia, P.; Guan, H.Q.; Peng, L.; Chen, Y.; et al. Clinical characteristics of COVID-19-infected cancer patients: A retrospective case study in three hospitals within Wuhan, China. Ann. Oncol. 2020, 31, 894–901. [Google Scholar] [CrossRef]
  132. Hussain, A.; Mahawar, K.; Xia, Z.; Yang, W.; El-Hasani, S. Obesity and mortality of COVID-19. Meta-analysis. Obes. Res. Clin. Pract. 2020, 14, 295–300. [Google Scholar] [CrossRef] [PubMed]
  133. Hamer, M.; Kivimäki, M.; Gale, C.R.; Batty, G.D. Lifestyle risk factors, inflammatory mechanisms, and COVID-19 hospitalization: A community-based cohort study of 387,109 adults in UK. Brain Behav. Immun. 2020, 87, 184–187. [Google Scholar] [CrossRef] [PubMed]
  134. Callaway, E. Omicron likely to weaken COVID vaccine protection. Nature 2021, 600, 367–368. [Google Scholar] [CrossRef]
  135. Lisco, A.; Hsu, A.P.; Dimitrova, D.; Proctor, D.M.; Mace, E.M.; Ye, P.; Anderson, M.V.; Hicks, S.N.; Grivas, C.; Hammoud, D.A.; et al. Treatment of Relapsing HPV Diseases by Restored Function of Natural Killer Cells. N. Engl. J. Med. 2021, 385, 921–929. [Google Scholar] [CrossRef] [PubMed]
  136. Terunuma, H.; Deng, X.; Nishino, N.; Watanabe, K. NK cell-based autologous immune enhancement therapy (AIET) for cancer. J. Stem Cells Regen. Med. 2013, 9, 9–13. [Google Scholar] [CrossRef] [PubMed]
  137. Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
  138. Christodoulou, I.; Rahnama, R.; Ravich, J.W.; Seo, J.; Zolov, S.N.; Marple, A.N.; Markovitz, D.M.; Bonifant, C.L. Glycoprotein Targeted CAR-NK Cells for the Treatment of SARS-CoV-2 Infection. Front. Immunol. 2021, 12, 763460. [Google Scholar] [CrossRef]
  139. Ma, M.T.; Badeti, S.; Chen, C.H.; Kim, J.; Choudhary, A.; Honnen, B.; Reichman, C.; Calianese, D.; Pinter, A.; Jiang, Q.; et al. CAR-NK Cells Effectively Target SARS-CoV-2-Spike-Expressing Cell Lines In Vitro. Front. Immunol. 2021, 12, 652223. [Google Scholar] [CrossRef]
  140. García-García, I.; Guerra-García, P.; Ferreras, C.; Borobia, A.M.; Carcas, A.J.; Queiruga-Parada, J.; Vicario, J.L.; Mirones, I.; Solano, C.; Eguizabal, C.; et al. A phase I/II dose-escalation multi-center study to evaluate the safety of infusion of natural killer cells or memory T cells as adoptive therapy in coronavirus pneumonia and/or lymphopenia: RELEASE study protocol. Trials 2021, 22, 674. [Google Scholar] [CrossRef]
  141. Casper, C.; Groysman, L.; Malhotra, V.; Whitman, E.; Herb, S.; Rave, E.; Lew, A.; Goman, C.; Sagawa, Z.; Thakar, M.; et al. Early report of a phase I/II study of human placental hematopoietic stem cell derived natural killer cells (CYNK-001) for the treatment of adults with COVID-19 (NCT04365101). Cancer Res. 2021, 81 (Suppl. 13), CT201. [Google Scholar] [CrossRef]
  142. Nyberg, T.; Ferguson, N.M.; Nash, S.G.; Webster, H.H.; Flaxman, S.; Andrews, N.; Hinsley, W.; Bernal, J.L.; Kall, M.; Bhatt, S.; et al. Comparative analysis of the risks of hospitalisation and death associated with SARS-CoV-2 omicron (B.1.1.529) and delta (B.1.617.2) variants in England: A cohort study. Lancet 2022, 399, 1303–1312. [Google Scholar] [CrossRef]
  143. Bartsch, Y.C.; Tong, X.; Kang, J.; Avendaño, M.J.; Serrano, E.F.; García-Salum, T.; Pardo-Roa, C.; Riquelme, A.; Cai, Y.; Renzi, I.; et al. Omicron variant Spike-specific antibody binding and Fc activity is preserved in recipients of mRNA or inactivated COVID-19 vaccines. Sci. Transl. Med. 2022, eabn9243. [Google Scholar] [CrossRef] [PubMed]
  144. Singanayagam, A.; Hakki, S.; Dunning, J.; Madon, K.J.; Crone, M.A.; Koycheva, A.; Derqui-Fernandez, N.; Barnett, J.L.; Whitfield, M.G.; Varro, R.; et al. Community transmission and viral load kinetics of the SARS-CoV-2 delta (B.1.617.2) variant in vaccinated and unvaccinated individuals in the UK: A prospective, longitudinal, cohort study. Lancet Infect. Dis. 2022, 22, 183–195. [Google Scholar] [CrossRef]
  145. Hertanto, D.M.; Sutanto, H.; Wiratama, B.S.; Wungu, C.D.K. Modulating the host immune response to fight against COVID-19: Where are we in 2021? Virulence 2021, 12, 1732–1736. [Google Scholar] [CrossRef]
  146. Cleary, S.J.; Pitchford, S.C.; Amison, R.T.; Carrington, R.; Robaina Cabrera, C.L.; Magnen, M.; Looney, M.R.; Gray, E.; Page, C.P. Animal models of mechanisms of SARS-CoV-2 infection and COVID-19 pathology. Br. J. Pharmacol. 2020, 177, 4851–4865. [Google Scholar] [CrossRef] [PubMed]
  147. Cleary, S.J.; Magnen, M.; Looney, M.R.; Page, C.P. Update on animal models for COVID-19 research. Br. J. Pharmacol. 2020, 177, 5679–5681. [Google Scholar] [CrossRef]
  148. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of human NK cells in healthy individual and COVID-19 patient. (A): Healthy human NK cells express the surface molecules CD56 and CD16; the inhibitory receptors MHC-I-specific killer cell immunoglobulin-like receptors (KIRs) and lectin-like CD94–NKG2A heterodimers; the activating receptors of NKG2D; natural cytotoxic receptors NKp30, NKp44, and NKp46; the cytokine receptors of IL-2, IL-12, IL-15, and IL-18; the chemokine receptors CXCR1 and CX3CR1; the effector molecules of cytokines IL-2, IFN-γ, and TNF-α; and cytolytic granules containing perforin and granzyme [45,46,49,52]. (B): In the peripheral blood of COVID-19 patients, dysregulation of NK cells has been observed, the cells exhibiting depletion and dysfunction. They show increased expression levels of CD38, HLA-DR, CD69, and Ksp37, and they exhibit a skewing to exhaustion, with increased expression levels of inhibitory receptors, such as NKG2A, Tim-3, CD244, PD-1, and CD39. Activating receptors with decreased expression levels include NKG2D, Siglec-7, DNAM-1, NKp46, and CD16. As a consequence, NK cells secrete reduced amounts of cytokines, such as IL-2, IFN-γ, and TNF-α, and show reduced degranulation, such as CD107a expression, whereas the production of chemokine MIP-β is increased. Moreover, NK cells show increased expression levels of Ki-67, CD98, CXCR6, CD103, Aiolos, caspase-3, and CD95, which are related to cell cycling, metabolism, tissue residency, hematopoietic development, and apoptosis, whereas the expression level of the homing and extravasation marker CD49d is decreased. In addition, the counts of NKG2C- and CD57-expressing adaptive NK cells are increased [26,27,28,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90] (arrows indicate the surface receptors and intracellular molecules showing a change in expression level).
Figure 1. Schematic representation of human NK cells in healthy individual and COVID-19 patient. (A): Healthy human NK cells express the surface molecules CD56 and CD16; the inhibitory receptors MHC-I-specific killer cell immunoglobulin-like receptors (KIRs) and lectin-like CD94–NKG2A heterodimers; the activating receptors of NKG2D; natural cytotoxic receptors NKp30, NKp44, and NKp46; the cytokine receptors of IL-2, IL-12, IL-15, and IL-18; the chemokine receptors CXCR1 and CX3CR1; the effector molecules of cytokines IL-2, IFN-γ, and TNF-α; and cytolytic granules containing perforin and granzyme [45,46,49,52]. (B): In the peripheral blood of COVID-19 patients, dysregulation of NK cells has been observed, the cells exhibiting depletion and dysfunction. They show increased expression levels of CD38, HLA-DR, CD69, and Ksp37, and they exhibit a skewing to exhaustion, with increased expression levels of inhibitory receptors, such as NKG2A, Tim-3, CD244, PD-1, and CD39. Activating receptors with decreased expression levels include NKG2D, Siglec-7, DNAM-1, NKp46, and CD16. As a consequence, NK cells secrete reduced amounts of cytokines, such as IL-2, IFN-γ, and TNF-α, and show reduced degranulation, such as CD107a expression, whereas the production of chemokine MIP-β is increased. Moreover, NK cells show increased expression levels of Ki-67, CD98, CXCR6, CD103, Aiolos, caspase-3, and CD95, which are related to cell cycling, metabolism, tissue residency, hematopoietic development, and apoptosis, whereas the expression level of the homing and extravasation marker CD49d is decreased. In addition, the counts of NKG2C- and CD57-expressing adaptive NK cells are increased [26,27,28,33,34,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90] (arrows indicate the surface receptors and intracellular molecules showing a change in expression level).
Biomedicines 10 01002 g001
Figure 2. Schematic representation of immune responses to SARS-CoV-2 infection. Upon SARS-CoV-2 infection, local innate immune responses are initiated, that is, tissue-resident NK cells, dendritic cells (DCs), and alveolar macrophages respond rapidly in virus-infected tissues. NK cells can function as innate cytotoxic effectors as well as regulators modulating adaptive immunity to eliminate the virus. Functional NK cells exhibit balanced activation, that is, higher expression levels of activating receptors and lower expression levels of inhibitory receptors upon recognition of the virus in infected tissues. As a consequence, sufficient numbers of activated NK cells exert increased degranulation and cytokine secretion, initiate responses to eliminate SARS-CoV-2-infected cells in the early stage, and crosstalk with DCs to facilitate adaptive immune responses, as follows. DCs recognize the degraded SARS-CoV-2 antigen, activated NK cells secrete an array of cytokines, such as IFN-γ, that promote DC maturation with the establishment of a cytokine milieu, such as the release of IL-12, and DCs present the SARS-CoV-2-derived peptides to CD4 T cells and CD8 T cells through TCR–MHC interactions. Once exposed to the antigen, Th0 CD4 T cells polarize primarily towards Th1, leading to the release of cytokines, which facilitate antigen presentation to cytotoxic CD8 T cells (CTLs). CTLs detect SARS-CoV-2-infected cells and release cytotoxic granules, including granzyme B and perforin, to eliminate virus-infected cells. On the other hand, Th2 CD4 T cells facilitate antigen-recognized B cells to trigger humoral-mediated immune responses and antibody secretion involved in SARS-CoV-2 elimination.
Figure 2. Schematic representation of immune responses to SARS-CoV-2 infection. Upon SARS-CoV-2 infection, local innate immune responses are initiated, that is, tissue-resident NK cells, dendritic cells (DCs), and alveolar macrophages respond rapidly in virus-infected tissues. NK cells can function as innate cytotoxic effectors as well as regulators modulating adaptive immunity to eliminate the virus. Functional NK cells exhibit balanced activation, that is, higher expression levels of activating receptors and lower expression levels of inhibitory receptors upon recognition of the virus in infected tissues. As a consequence, sufficient numbers of activated NK cells exert increased degranulation and cytokine secretion, initiate responses to eliminate SARS-CoV-2-infected cells in the early stage, and crosstalk with DCs to facilitate adaptive immune responses, as follows. DCs recognize the degraded SARS-CoV-2 antigen, activated NK cells secrete an array of cytokines, such as IFN-γ, that promote DC maturation with the establishment of a cytokine milieu, such as the release of IL-12, and DCs present the SARS-CoV-2-derived peptides to CD4 T cells and CD8 T cells through TCR–MHC interactions. Once exposed to the antigen, Th0 CD4 T cells polarize primarily towards Th1, leading to the release of cytokines, which facilitate antigen presentation to cytotoxic CD8 T cells (CTLs). CTLs detect SARS-CoV-2-infected cells and release cytotoxic granules, including granzyme B and perforin, to eliminate virus-infected cells. On the other hand, Th2 CD4 T cells facilitate antigen-recognized B cells to trigger humoral-mediated immune responses and antibody secretion involved in SARS-CoV-2 elimination.
Biomedicines 10 01002 g002
Figure 3. Schematic representation of dysregulated immune responses in COVID-19. NK cells are dysregulated in COVID-19, exhibiting depletion and dysfunction. The dysfunctional NK cells exhibit lower expression levels of activating receptors and higher expression levels of inhibitory receptors. As a consequence, NK cells show reduced degranulation and cytokine secretion. They fail to directly eliminate SARS-CoV-2-infected cells, and to indirectly, because of their impaired crosstalk with DCs, modulate adaptive immune responses. They fail to provide adequate CTL- and antibody-mediated SARS-CoV-2 elimination, allowing the virus to persistently replicate and spread, leading to further recruitment of infiltrating monocytes and granulocytes with the intent to initiate viral clearance. Together, these failures result in excessive and prolonged stimulation of the immune system, with the progressive accumulation of virus-infected cells and inflammatory myeloid cells to the sites of viral infections [17,18,19,20]; this in turn leads to the production of chemokines and cytokines and further recruitment of inflammatory cells, resulting in the dysregulated release of pro-inflammatory cytokines at elevated levels, contributing to over-inflammation and tissue injury [15,16,125]. (Arrows indicate the reduced degranulation and secretion of cytokines and antibodies).
Figure 3. Schematic representation of dysregulated immune responses in COVID-19. NK cells are dysregulated in COVID-19, exhibiting depletion and dysfunction. The dysfunctional NK cells exhibit lower expression levels of activating receptors and higher expression levels of inhibitory receptors. As a consequence, NK cells show reduced degranulation and cytokine secretion. They fail to directly eliminate SARS-CoV-2-infected cells, and to indirectly, because of their impaired crosstalk with DCs, modulate adaptive immune responses. They fail to provide adequate CTL- and antibody-mediated SARS-CoV-2 elimination, allowing the virus to persistently replicate and spread, leading to further recruitment of infiltrating monocytes and granulocytes with the intent to initiate viral clearance. Together, these failures result in excessive and prolonged stimulation of the immune system, with the progressive accumulation of virus-infected cells and inflammatory myeloid cells to the sites of viral infections [17,18,19,20]; this in turn leads to the production of chemokines and cytokines and further recruitment of inflammatory cells, resulting in the dysregulated release of pro-inflammatory cytokines at elevated levels, contributing to over-inflammation and tissue injury [15,16,125]. (Arrows indicate the reduced degranulation and secretion of cytokines and antibodies).
Biomedicines 10 01002 g003
Table 1. Ongoing clinical trials of NK cell therapy for COVID-19.
Table 1. Ongoing clinical trials of NK cell therapy for COVID-19.
No.NK CellSubjectPhaseTrial IdentifierStatusLocation
1UndisclosedNot clearINCT04280224RecruitingChina
2Allogeneic NKG2D-ACE2 CAR-NK cells enriched from umbilical cord blood and engineered geneticallyOnset of illness <14 daysI/IINCT04324996RecruitingChina
3Induced pluripotent stem cell (iPSC)-derived NK cells Hospitalized patientsINCT04363346CompletedMinnesota, USA
4Human placental CD34+-derived and culture-expanded NK cellsModerate diseaseI/IINCT04365101Active, not recruitingNew Jersey, USA
5Allogeneic NK cells from donors who have recovered from COVID-19Onset of symptoms <10 daysI/IINCT04578210RecruitingSpain
6UndisclosedUndisclosedINCT04634370Not yet recruitingBrazil
7Allogeneic NK cells derived from CD34+ hematopoietic stem cellsHospitalized patientsINCT04900454RecruitingSeattle, USA
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Deng, X.; Terunuma, H.; Nieda, M. Exploring the Utility of NK Cells in COVID-19. Biomedicines 2022, 10, 1002. https://doi.org/10.3390/biomedicines10051002

AMA Style

Deng X, Terunuma H, Nieda M. Exploring the Utility of NK Cells in COVID-19. Biomedicines. 2022; 10(5):1002. https://doi.org/10.3390/biomedicines10051002

Chicago/Turabian Style

Deng, Xuewen, Hiroshi Terunuma, and Mie Nieda. 2022. "Exploring the Utility of NK Cells in COVID-19" Biomedicines 10, no. 5: 1002. https://doi.org/10.3390/biomedicines10051002

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop