Skip to main content
Erschienen in: Current HIV/AIDS Reports 2/2016

Open Access 21.03.2016 | HIV Pathogenesis and Treatment (AL Landay, Section Editor)

NK Cells in HIV Disease

verfasst von: Eileen Scully, Galit Alter

Erschienen in: Current HIV/AIDS Reports | Ausgabe 2/2016

Abstract

Natural killer (NK) cells play a critical role in viral immunity. In the setting of HIV infection, epidemiologic and functional evidence support a role for NK cells in both protection from new infection and in viral control. Specifically, NK cells directly mediate immune pressure leading to virus evolution, and NK cell receptor genotypic profiles, clonal repertoires, and functional capacity have all been implicated in virus containment. In addition, indirect NK cell-mediated antibody-dependent cellular cytotoxicity has been linked to vaccine-induced protective immunity against HIV infection. With recent advances in our understanding of NK cell deficiency, development, memory-like responses, and editing of the adaptive immune system, the opportunities to direct and exploit NK cell antiviral immunity to target HIV have exponentially grown. In this review, we seek to highlight the intersections between discoveries in basic NK cell biology and the challenges of HIV chronic infection, vaccine development, and cure/eradication strategies.
Hinweise
This article is part of the Topical Collection on HIV Pathogenesis and Treatment

Introduction

Natural killer (NK) cells occupy a unique niche in the immune response, bridging the innate and adaptive immune systems. They are capable of recognizing generic signals of stress, transformation, or infection with immediate effector function. NK cells are the critical antiviral effectors of the innate immune system, and natural deficiencies are associated with susceptibility to viral infections [1, 2]. However, there is still a nascent understanding of their pleiotropic functions as direct effectors, but also as editors of adaptive immunity in both infection and vaccination [3]. Recent work has also highlighted the adaptive features of NK cells that can confer a memory-like phenotype in the setting of infection [4]. NK cells have the potential to directly respond to viruses, to develop memory-like responses after initial pathogen encounter or vaccination, and to shape the adaptive immune response (Fig. 1).
As the understanding of NK cell biology grows, there are promising avenues to address several current challenges in HIV: development of an effective vaccine, control of residual inflammation in treated disease, and emerging strategies for eradication or cure. NK cell deficiency syndromes have revealed specific developmental programs that shape the mature NK cell population and can broadly modulate responses [5]. The HIV vaccine literature has identified the critical importance of antibody-dependent cellular cytotoxicicty (ADCC), mediated predominantly by NK cells, via non-neutralizing antibodies as a pathway to protective vaccine responses [6]. Importantly, these ADCC responses are associated with specific immunoglobulin subclasses [7] and glycan modifications that directly impact the efficiency of NK cell responses [8, 9]. This affords multiple avenues for optimization of antibodies to maximize NK cell function for preventative and therapeutic vaccination [10].
The profile of NK cell responsiveness beyond CD16-mediated antibody recognition also points to anti-HIV function. High dimensional characterization of the combinatorial diversity of NK receptor expression offers new insights into the phenotypic features of protective NK cell responses and how they may be directed [11, 12••]. Building on prior work showing antigen specificity in mouse models, recent studies have demonstrated NK cell memory responses to simian immunodeficiency virus (SIV) antigens in nonhuman primates, suggesting a pathway to induce antigen-driven memory in humans [13••]. In line with recent findings in the myeloid compartment delineating the phenomena of innate training and tolerance [14], unique epigenetic signatures in NK cells primed by cytomegalovirus (CMV) infection demonstrate how NK cells can be programmed by an initial pathogen (or potentially vaccine) exposure [15••, 16••]. Taken together, this work suggests infection can shape the NK cell repertoire and alter subsequent responses; a detailed understanding of this functional plasticity will inform efforts to develop preventative and therapeutic vaccines against HIV.
In this review, we discuss the role of NK cells in the immune response to HIV in the context of recent advances in NK cell biology, highlighting opportunities to harness these innate cytolytic effector cells for the rapid containment and clearance of HIV.

NK Cells in HIV Infection

The Role of NK Cells in the Immune Response to HIV Infection

Population-level genetic associations between NK cell receptor expression and HIV-1 outcomes and evolution reveal the impact of NK cells on HIV-1 disease progression. The germline encoded inhibitory receptors including the killer immunoglobulin-like receptors (KIRs) heavily influence NK cell activation which is governed by the integration of activating and inhibitory signals. Interactions between KIRs and their cognate HLA ligands set a threshold for NK activity and have been shown to critically influence the course of viral infection, associating with resolution of hepatitis C infection [17]. In HIV, HLA/KIR combinations have been associated with the pace of disease progression [18, 19] and protection against disease acquisition [20, 21]. The mechanisms conferring this protection may include both NK cell education/licensing through inhibitory receptor ligation [22] and the direct interaction of KIRs with HIV-1-derived peptide motifs presented on HLA molecules. The latter is supported by virus evolution in the presence of specific KIRs; footprints in the HIV genome associate with KIR genotype. Specific viral variant/KIR combinations associate with differences in NK cell viral inhibition in vitro [23] and HLA/KIR combinations confer differences in HIV control [24]. The HLA/KIR interactions directed by specific HIV-derived peptides are further linked to measures of NK cell function in vitro and patterns of viral escape in population studies [25, 26]. These data suggest that NK cell activation threshold, determined in part by the genetic array of inhibitory receptors, and the virus-derived peptides available for presentation on host HLA cooperate to define the protective efficacy of NK cell responses. The latter could be exploited to harness function; computation approaches may be able to predict peptides that modulate NK function based on host HLA/KIR patterns to individualize vaccine design.
These data demonstrate direct interactions between NK cells and HIV peptides; however, it is not known whether these signals govern in vivo effector function and which of the overlapping recognition pathways is sufficient for an effective response. Although counterintuitive, focusing on immune responses that HIV has evolved to evade may point to highly effective means of NK activation. HIV-mediated downregulation of HLA molecules, which shields infected cells from killing by CD8+ T cells [27], theoretically offers the “missing self” trigger for NK cells. However, this is limited by HIV immune evasion; the downregulation of HLA A and B by Nef is coupled to preservation of HLA C and E, maintaining self signals, and preventing NK activation [28]. Likewise, HIV infection directly triggers the upregulation of stress ligands for cytotoxicity receptors including NKG2D [29, 30]. Again, HIV is able to limit expression of these ligands and other coactivating signals such as NTBA-4 through accessory proteins [29, 31]. While the precise contribution of each activation pathway in natural infection is difficult to prove, from the perspective of prevention and cure, each could be leveraged to optimize responses. Small molecule inhibitors disabling immune evasion through viral accessory proteins may enhance native or vaccine-induced immune recognition.
ADCC is a potent means of NK cell control of HIV-1 infection. NK cells express the FcγRIIIA receptor (CD16) that binds the constant (Fc) domain of IgG antibodies. CD16 engagement is a strong activator of NK cell function, and allows antigen-specific recruitment of NK responses. Importantly, ADCC activity was associated with the modest protective efficacy in the RV144 HIV vaccine trial [6] and has been implicated in phenotypes of viral control [32, 33]. Responses characterized by coordinated antibody function, including NK mediated ADCC and NK cytokine secretion/degranulation associated with viral controller phenotypes [34]. ADCC efficiency is linked to specific antibody features including subclass and glycosylation that can be modified to enhance NK cell recruitment and activation. NK cell maturation and education can further enhance HIV-specific ADCC activity [35, 36]. Of note, as with other effector pathways, ADCC is also limited by viral evasion: the viral accessory protein Vpu antagonizes the antiviral factor tetherin, altering the release of virus aggregates and disabling ADCC mediated viral recognition [3741]. A small molecule inhibitor of vpu’s interaction with tetherin has recently been described suggesting an additional target for intervention [42]. An optimized approach would prime NK cells, induce antibodies with glycan and subclass features maximized for ADCC and could potentially be coupled with pharmacologic blockade of viral immune evasion to further enhance antiviral function.
The collective data highlight the importance of NK cells in HIV disease. There are multiple pathways for target elimination, all balanced by mechanisms of viral evasion that define kinetic windows of maximal response and opportunities for enhancement. In addition to these specific interactions, NK cell recognition of generic signals of stress and infection induced early in HIV infection is an important area for exploration. Optimized NK cell function can be achieved through directly augmenting NK cell function with priming and subset expansion or inhibitory receptor blockade, indirectly through optimization of antibodies to recruit NK cells, and potentially also through blockade of viral accessory proteins to limit immune evasion.

The Consequences of HIV Infection for NK Cell Distribution and Function

Chronic HIV-1 infection alters the population distribution and functional capacity of NK cells. Numerous studies have described these changes in the setting of HIV infection and have at times produced diverging and contradictory results. This lack of clarity is likely related to multiple factors: the dependence of NK cell function on the associated HLA genotype has not always been fully considered, many studies are cross sectional in nature and may not have adequately controlled for confounders between groups including CMV serostatus, age, and gender, and finally the diversity of NK cell phenotypes is not fully captured in a limited examination of markers [11, 12••].
NK cells are divided into subsets based on their expression of CD56 and CD16; these subsets are thought to represent stages in NK cell differentiation, although the precise relationship has not been fully defined. Based on immunodeficiency syndromes, both CD56bright and CD56dim populations contribute to antiviral immunity. CD56brightCD16 cells are a minor population in healthy individuals with relatively limited cytotoxic capacity but strong production of cytokines. CD56dim NK cells are the majority of the circulating population with less proliferative potential, increased cytotoxic capacity, and progressive acquisition of KIRs and markers including CD57 during differentiation. A third minor population is the CD56neg NK cells that are CD16+; these cells have been difficult to characterize with their limited expression of lineage markers. NK cells express a broad complement of additional receptors including the predominantly inhibitory KIRs; the natural cytotoxicity receptors (NCRs) including NKp30, NKp44, and NKp46; the C-type lectin receptors including NKG2D, NKG2C, and NKG2A; signaling lymphocyte activation (SLAM) family receptors as well as the Fcγ receptor CD16 [43]. NK cell degranulation/cytotoxicity and cytokine secretion are governed by the overall balance of activating and inhibitory signals, with an important role for education through inhibitory signaling during development [44]. The roles of each subset in HIV-1 infection are still being defined.
During primary HIV infection, initiation of antiretroviral therapy was associated with reciprocal variation in the CD16+CD56dim and CD56high NK cells, with the latter, more immature population, increasing after therapy [45]. Initiation of therapy also changes immunoregulatory markers; modulation of galectin 9 and T cell immunoglobulin and mucin-domain containing molecule-3 (TIM-3) in early infection leads to enhanced NK cell activity, but in chronic HIV the interaction may contribute to NK cell dysfunction [46]. Dysregulated TIM-3 expression on NK cells is linked to impaired CD4 recovery after treatment, suggesting that NK cell dysfunction may contribute to a poor immunologic reconstitution in some individuals [47]. CD56negCD16+ cells with reduced ADCC activity also accumulate during chronic infection, a defect that is significantly restored with viral suppression [48, 49]. Counterbalancing this restoration of NK cell ADCC is the decline in antibody conferred activity after initiation of antiretroviral therapy (ART) [50]. Thus, to fully exploit the potential for NK cell-mediated function in cure/eradication strategies through ADCC, restoration of subset distribution is an essential step, beyond optimizing antibody subclass, glycan structure and titer through vaccination or administration of a monoclonal.
In vitro studies have sought to dissect receptor patterns that lead to NK cell control of HIV infection. When exposed to infected CD4+ T cells, autologous NK cells expressing NKG2A responded with CD107a (a marker of degranulation), IFNγ, and CCL4 at the highest frequencies [51]. Profiling the NKG2A receptor expression in an individual may offer an index of their likelihood to have an effective NK cell response to a therapeutic intervention. In the setting of significant inflammation in a cohort in Uganda with HIV subtype D infection, impaired effector function (IFNγ production) and loss of a highly activated subset of NK cells has been described [52]. Further evidence of dysfunction is suggested by studies in the SIV nonhuman primate model demonstrating anergic NK cell accumulation in lymph nodes in chronic infection [53]. Research to redirect or rescue function will be informed by the more complete understanding of the pathology of HIV. An alternative window to identify highly effective activation pathways may be through the enhanced effector capability seen in NK cells from individuals with spontaneous viral control. One recent example is the recent report of a potential role for NK cells in the long-term suppression of HIV-1 in the VISCONTI cohort of post-treatment controllers (Scott-Algara, et al., Abstract 15,CROI 2015, Seattle, WA).
In support of the concept that NK cells can be directed, methods of bolstering NK cell functional capacity in HIV infection have been described. A therapeutic HIV vaccine that restored CD4+ T cell IL-2 production led to enhanced production of IFNγ by NK cells [54]. Secretion of IFNα by other cells drives NK cell activation and lysis of HIV-infected CD4+ T cells through NKp46- and NKG2D-mediated signaling [55]. The cross-talk between adaptive and NK cell responses is a critical means for optimizing NK cell function. Coordinating the recruitment and activity of multiple cell types, particularly in the setting of an inflamed and anergic immune system, may be the key to promoting more effective NK cell control of HIV.

NK Cell Development

Genetic Determinants of NK Cell Deficiency Syndromes

Naturally occurring NK cell deficiency syndromes offer important insights into the biology of these cytolytic effectors. Deficiency syndromes are broadly divided into classical NK cell deficiency (CNKD) with numeric and functional deficits, and functional NK cell deficiency (FNKD) in which cells are present but poorly functional (reviewed in [5, 56]). Genetic defects associated with both types of syndrome have been identified. In CNKD1, mutations in the transcription factor GATA2 confer deficits in peripheral NK cell numbers and function, with marked susceptibility to viral infections (varicella zoster virus, herpes simplex virus, cytomegalovirus, human papilloma virus) and mycobacterial infections. These patients specifically lack of the CD56bright subset and show globally impaired expression of NKG2D [57]. A distinct syndrome, CNKD2, is linked to a mutation in MCM4, with a converse phenotype of absence of the CD56dim population with clinical findings of recurrent viral infections and EBV-driven lymphoproliferative disorders [58]. The deficit in opposite subsets in these two syndromes is striking given their proposed developmental relationship and the susceptibility to viral infections seen in both patient groups. In a separate patient cohort with marked susceptibility to herpesvirus infections and a form of FNKD, studies identified a mutation in the membrane distal domain of CD16. This mutation abrogated an interaction between CD16 and CD2, preventing activation of spontaneous cytotoxicity responses despite preserved ADCC [59]. The efficiency of ADCC responses is also associated with control of herpes simplex virus (HSV). Higher affinity allotypes of CD16 and IgG are linked to control of HSV (versus recurrent outbreaks) and are associated with enhanced ADCC in vitro [60, 61]. Multiple additional genetic loci have been linked to deficiencies in NK cell number and function in combined immune deficiency syndromes [62].
Taken together, these findings highlight key functional and developmental aspects of NK cell biology. First, both the CD56bright and CD56dim populations have significant roles in control of virus infections. Second, expression of CD16 has dual significance, promoting cytotoxic activity triggered by CD2 ligation as well as mediating ADCC. Third, the efficiency of the ADCC response is associated with successful control of herpes virus, reinforcing the role of ADCC in phenotypes of elite control in HIV. These observations may inform some of the disruption seen with shifts in the CD56 expression with chronic HIV infection, and supports the pursuit of both direct antiviral and ADCC strategies in HIV eradication and cure efforts.

Homeostasis and Maturation Signals

Given the defects seen in chronic HIV infection, understanding key differentiation signals in NK cell biology may point to therapeutic strategies to shape or direct their responses. IL-15 is a master regulator of NK cell maturation, survival, and functional competence. IL-15 preferentially expands cytotoxic NK subsets, with less proliferative stimulus for regulatory T cells than IL-2, shifting the net balance towards activation [63]. Recent human clinical trials in patients with metastatic cancer have demonstrated proliferation and activation of NK cells after administration of recombinant human IL-15 [64••]. In this exploratory study, no objective remissions were achieved; however, there was preliminary evidence of immune activity seen in the clearance of lung lesions in two of the subjects [64••]. This particular approach is relevant to HIV cure strategies; enhancing innate surveillance by NK cells in concert with latent virus reactivation can circumvent limitations of traditional adaptive responses to HIV. Ongoing efforts to optimize delivery methods (e.g., with heterodimeric formats of the cytokine and a receptor subunit [65]) and synergistic combinations of cytokine with antibody or effector CTLs are active areas of investigation (Euler, Jones, and Alter, in preparation). In a humanized mouse model of HIV infection, use of an IL-15 superagonist (IL-15 bound to the soluble IL-15Rα) effectively blocked HIV infection, in an NK cell dependent manner [66]. In parallel to these studies, multiple clinical trials of adoptive therapy with expanded allogeneic NK cells in cancer are assessing the therapeutic potential of ex vivo expansion and reinfusion into patients [67]. Results of these trials will be critical to define the safety of NK cell expansion and administration and may inform which subsets are critical to immune protection. Both of these approaches, in vivo exposure to IL-15 and ex vivo NK cell manipulation and reinfusion, have potential advantages, and further work is needed to define the most promising avenues.
Multiple additional pathways to modulate NK cell maturation and function are under investigation. Immunomodulatory therapies have potential efficacy in NK cells analogous to the effects described for the adaptive immune system; blocking PD-1 on NK cells from multiple myeloma patients enhanced killing of autologous tumor cells [68]. Indirectly, proteasome inhibitors used in cancer chemotherapy also enhance NK cell function, at least in part by downregulation of HLA molecules triggering missing self responses [69]. Due to their highly potent cytotoxic activity, an array of inhibitory receptors tonically repress the function of NK cells [44], offering another target for intervention. Direct blockade of inhibitory KIR receptors has been shown to augment spontaneous NK cell cytotoxicity in animal models [70] and is in exploratory clinical trials of cancer therapy [71].

NK Cell Activation and Specificity

Efforts to modulate NK cells with cytokines and direct therapies point back to a critical question in NK cell biology. While there is considerable variation in NK cell response potential, the precise determinants (e.g., receptor expression and maturation state) of an NK cell response to pathogen challenge are not clear. In contrast to adaptive immune responses that are governed by antigen-specific gene-rearranged receptors, NK cells integrate multiple activating and inhibitory signals to reach a threshold of activation. Importantly, both the KIR genotype of an individual [22] and the multiple unique combinations of receptors that can be expressed [11, 12••] contribute to the NK population diversity of receptor expression and activation potential. However, apart from the genetic determinants, environmental and stochastic factors including infectious exposures such as CMV are also capable of profoundly shaping the response potential of NK cells through alterations in repertoire distribution [72, 73]. Building on differences in surface receptors, work defining transcriptional signatures of NK cells in distinct activation states has provided a context to understand NK cell activation alone and in relationship to innate lymphoid cells and traditional T cells [74]. Moving forward, to truly harness NK cells towards a therapeutic goal, the determinants of NK cell repertoire composition, activation threshold, and role in susceptibility to infection must be defined.

NK Cell Repertoire Diversity

Detailed studies with high dimensional analysis enabled by cytometry by time of flight (CyTOF) has provided novel insights into the diversity and specificity of the NK cell repertoire [11, 12••]. These studies reveal the potential for NK cell combinatorial receptor diversity, with estimates of up to 30,000 unique receptor phenotypes on a single NK cell in an individual subject. Remarkably, most individual receptor combinations contributed to only a minority of the overall NK cell population, with only the less mature NKG2A+/CD94+ NK cells and more mature CD57+/CD16+ population as primary distinct phenotypes [11]. Distinct responses to stimuli, proliferative potential, cytokine production, and cytolytic activity are seen in subsets defined by the expression of CD57 [75]. Diversity of phenotypes in adults is correlated with CD57 expression, suggesting that maturation leads to divergence, and was associated with risk of HIV acquisition. In case control analyses of high-risk women, increased NK cell diversity was associated with an increased probability of HIV-1 infection [12••]. It is not clear whether the NK diversity reflects a “distraction” from the challenging pathogen that increases susceptibility or if it is linked to other immune characteristics such as T cell exhaustion. These data raise questions about preventative and therapeutic vaccination goals; a naïve repertoire may have more flexibility and protective potential than one directed at other pathogens; however, a vaccine-directed, pathogen-specific response should be superior to both. Understanding how clonal NK cell populations can be reshaped and directed will critically inform the potential to harness a range of biological functions of NK cells through vaccine design.

NK Cell Memory

While classically considered as nonspecific innate effector cells, there is growing data to support adaptive, memory-like features of NK cells. These features can be divided into two categories, antigen-specific responses with classical recall characteristics and a more general alteration in effector function conditioned by an infection, vaccination, or inflammatory environment in line with the phenomenon of innate training in the myeloid compartment [14, 76]. Both types of memory responses are potential targets for immune intervention.
The mouse model of hapten-mediated contact hypersensitivity reactions provided the first demonstration of persistent, antigen-specific, and transferrable memory in NK cells [77]. Further work identified the importance of the chemokine receptor CXCR6 in memory development and extended the observations from haptens to influenza virus-like particles and inactivated vaccinia virus; the receptors mediating this recognition have remained undefined [78]. The first report of NK cell memory in the setting of a viral infection demonstrated NK cell-mediated responses to mouse cytomegalovirus (MCMV) [79]. Development of murine CMV (MCMV) memory was linked to interactions between the NK receptor LY49H and the viral glycoprotein m157 [80, 81], and memory cells were broadly distributed in the tissues, in contrast to the liver-resident hapten-directed memory cells [79]. More recently, memory NK cells specific for SIV antigens have been identified in the nonhuman primate model [13••]. NK cells derived from rhesus macaques infected with SIV or a virus combining SIV and HIV (SHIV) showed antigen-specific killing of targets pulsed with HIV protein components, and a persistent response to vaccine antigens was identified in animals who had received an adenovirus 26 (Ad26) vectored SIV vaccine 5 years earlier [13••]. These memory NK cells were elicited by both vaccination and infection, were durable and specific, and were recovered from both the liver and spleen [13••]. These animal model data demonstrate antigen-specific NK cell memory and while direct evidence in humans is lacking, these studies strongly support efforts to identify and harness this activity in human disease. Access to the relevant cell populations, e.g., hepatic or splenic cells, may be critical to identify these responses and should be one focus of study. In parallel, in vitro studies to induce memory-like response and animal models including the humanized mouse offer additional opportunities to explore the biology of these cytolytic effectors.
Antigen-specific memory in NK cells has clear parallels to traditional adaptive immunological memory. Consistent with their unique role bridging the innate and adaptive responses, NK cells are also capable of a memory phenotype that is induced by more generalized signals. Exposure of murine NK cells to cytokines (IL-12, IL-15, and IL-18) in vitro can induce a memory-like phenotype. When transferred into naïve hosts, these cells have a resting phenotype, but produce more IFNγ on cytokine restimulation, a response that remains detectable 3 weeks after cell transfer [82]. Human NK cells have similar properties, with a brief exposure to cytokines conditioning enhanced responsiveness on rechallenge [83]. How this memory-like state is established is a topic of investigation, but epigenetic remodeling of a noncoding sequence at the IFNG gene linked to increased IFN production can be induced by in vitro cytokine priming [84]. Epigenetic programming also underlies the sustained shifts in NK cell profiles that are seen in human CMV (HCMV) infection. HCMV infection drives expansion of a population of CD94-NKG2C NK cells [85], and in the setting of hematopoietic stem cell transplantation, this population has been demonstrated to have a memory-like response to CMV [86]. The interaction between HLA-E and CD94-NKG2C contributes to this expansion [87], but additional pathways to NK memory in HCMV are also operative, as evidenced by the FcεRIγ-deficient adaptive NK cells that expand after activation through CD16 [16••]. HCMV infection is associated with sustained changes in NK cell repertoire, distinct epigenetic profiles [15••, 16••], and altered functional profiles [15••, 16••]. The responses appear to be directed by exposure to the pathogen, in some cases directly through a viral antigen, and in other cases through secondary recognition of specific antibody. In congruence with the findings in HCMV, an NK cell population that lacks FcRγ expression and has enhanced ADCC activity was identified in HIV-infected subjects, with some features shared with the memory-like population induced by CMV and some unique surface receptor characteristics [88]. These findings have compelling links to both the biology of adaptive immune responses and the growing field of research in innate training and tolerance whereby epigenetic programs direct altered secondary responses after an initial exposure [14, 89], and both pathways can be harnessed towards goals of HIV prevention and cure.

NK Cell Editing of Adaptive Immunity

Recent studies have focused attention on a critical role for NK cells in the shaping adaptive immune responses. In MCMV infection, NK cells rapidly eliminate infected targets, limiting the type I interferon response, preserving conventional dendritic cells and CD8 T cell responses [90]. In this infection, NK cells limit exposure of CD4 and CD8 T cells to infected dendritic cells shaping the subsequent adaptive response [91] and importantly, also limit tissue site T cell-mediated pathology [92]. In HIV, NK cell editing of dendritic cells is aberrant in the context of chronic inflammation and elevated IL-10, leading to poorly dendritic cells with limited immunogenicity [93]. Similarly, NK cells dictate immune response characteristics in an indirect fashion in the lymphocytic choriomeningitis virus (LCMV) mouse model. In this system, NK cells have no significant role in elimination of virus-infected targets, but they eliminate activated CD4 T cells either limiting immunopathology, or contributing to exhaustion and inefficient CD8 T cell control in chronic infection [9496]. NK cells have also been shown to shape the induction of antibody; perforin-mediated elimination of T follicular helper (Tfh) cells in the lymph node by NK cells in acute infection was shown to disrupt germinal center formation, limiting immune memory development [97••]. Tfh cells have been identified as the dominant population supporting replication and virus production in viremic HIV-1 infection [98], and are likely a significant contributor to the HIV-1 reservoir.
The context-dependent effects of NK cells on adaptive immunity highlight the need for careful direction of efforts to harness their activity in HIV infection. Specifically, disabling their CD4 suppressive effects after vaccination may promote more breadth of antibody response. In contrast, during acute infection, enhancing NK-mediated elimination of Tfh cells may limit the size of the reservoir that is established. Likewise, during a curative intervention, unleashing NK cell targeting of Tfh cells could again lead to reservoir reduction. Directed recruitment of NK cells to lymph nodes also offers a pathway to increase their efficacy [99]. The impact of NK cells on interventions targeting adaptive responses offers a novel pathway to enhance efforts at prevention and cure [3].

Conclusions

NK cells have emerged as multifunctional effector cells with the potential to control infections and shape adaptive immune responses. NK cells exert immune pressure on HIV and contribute to protective vaccine responses and some phenotypes of immune control. Significantly, there has been little effort to optimize, direct, or specifically target NK cells in therapeutic and preventative interventions. Mounting evidence from basic biological studies of NK cells points to the multiple avenues to enhance their antiviral and immunomodulatory function in HIV. The barrier to eliciting effective immunity to HIV is significant, and the potential to harness this important and potent effector population must be a high-priority component of research efforts in prevention and cure.

Acknowledgments

We would like to thank Avery Normandin for expert assistance in figure design.

Compliance with Ethical Standards

Conflict of Interest

Eileen Scully and Galit Alter declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med. 1989;320:1731–5.CrossRefPubMed Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med. 1989;320:1731–5.CrossRefPubMed
2.
Zurück zum Zitat Fleisher G, Starr S, Koven N, Kamiya H, Douglas SD, Henle W. A non-x-linked syndrome with susceptibility to severe Epstein-Barr virus infections. J Pediatr. 1982;100:727–30.CrossRefPubMed Fleisher G, Starr S, Koven N, Kamiya H, Douglas SD, Henle W. A non-x-linked syndrome with susceptibility to severe Epstein-Barr virus infections. J Pediatr. 1982;100:727–30.CrossRefPubMed
3.
Zurück zum Zitat Rydyznski CE, Waggoner SN. Boosting vaccine efficacy the natural (killer) way. Trends Immunol. 2015;36:536–46.CrossRefPubMed Rydyznski CE, Waggoner SN. Boosting vaccine efficacy the natural (killer) way. Trends Immunol. 2015;36:536–46.CrossRefPubMed
4.
6.
7.
Zurück zum Zitat Chung AW, Ghebremichael M, Robinson H, et al. Polyfunctional Fc-effector profiles mediated by IgG subclass selection distinguish RV144 and VAX003 vaccines. Sci Transl Med. 2014;6:228ra238. Chung AW, Ghebremichael M, Robinson H, et al. Polyfunctional Fc-effector profiles mediated by IgG subclass selection distinguish RV144 and VAX003 vaccines. Sci Transl Med. 2014;6:228ra238.
8.
Zurück zum Zitat Bournazos S, Klein F, Pietzsch J, Seaman MS, Nussenzweig MC, Ravetch JV. Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell. 2014;158:1243–53.CrossRefPubMedPubMedCentral Bournazos S, Klein F, Pietzsch J, Seaman MS, Nussenzweig MC, Ravetch JV. Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell. 2014;158:1243–53.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Chung AW, Crispin M, Pritchard L, et al. Identification of antibody glycosylation structures that predict monoclonal antibody Fc-effector function. AIDS. 2014;28:2523–30.CrossRefPubMedPubMedCentral Chung AW, Crispin M, Pritchard L, et al. Identification of antibody glycosylation structures that predict monoclonal antibody Fc-effector function. AIDS. 2014;28:2523–30.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Ackerman ME, Alter G. Opportunities to exploit non-neutralizing HIV-specific antibody activity. Curr HIV Res. 2013;11:365–77.CrossRefPubMed Ackerman ME, Alter G. Opportunities to exploit non-neutralizing HIV-specific antibody activity. Curr HIV Res. 2013;11:365–77.CrossRefPubMed
11.
Zurück zum Zitat Horowitz A, Strauss-Albee DM, Leipold M, et al. Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci Transl Med. 2013;5:208ra145.PubMedPubMedCentral Horowitz A, Strauss-Albee DM, Leipold M, et al. Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci Transl Med. 2013;5:208ra145.PubMedPubMedCentral
12.••
Zurück zum Zitat Strauss-Albee DM, Fukuyama J, Liang EC, et al. Human NK cell repertoire diversity reflects immune experience and correlates with viral susceptibility. Sci Transl Med. 2015;7:297ra115. Using high dimensional analysis, this study shows the potential diversity of unique NK cell phenotypes determined by different receptor combinations and links this diversity to susceptibility to infection with HIV.CrossRefPubMedPubMedCentral Strauss-Albee DM, Fukuyama J, Liang EC, et al. Human NK cell repertoire diversity reflects immune experience and correlates with viral susceptibility. Sci Transl Med. 2015;7:297ra115. Using high dimensional analysis, this study shows the potential diversity of unique NK cell phenotypes determined by different receptor combinations and links this diversity to susceptibility to infection with HIV.CrossRefPubMedPubMedCentral
13.••
Zurück zum Zitat Reeves RK, Li H, Jost S, et al. Antigen-specific NK cell memory in rhesus macaques. Nat Immunol. 2015;16:927–32. This study demonstrates that antigen specific memory to SIV and SHIV antigens can be generated by infection and vaccination in non-human primates.CrossRefPubMedPubMedCentral Reeves RK, Li H, Jost S, et al. Antigen-specific NK cell memory in rhesus macaques. Nat Immunol. 2015;16:927–32. This study demonstrates that antigen specific memory to SIV and SHIV antigens can be generated by infection and vaccination in non-human primates.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Netea MG, Latz E, Mills KH, O’Neill LA. Innate immune memory: a paradigm shift in understanding host defense. Nat Immunol. 2015;16:675–9.CrossRefPubMed Netea MG, Latz E, Mills KH, O’Neill LA. Innate immune memory: a paradigm shift in understanding host defense. Nat Immunol. 2015;16:675–9.CrossRefPubMed
15.••
Zurück zum Zitat Schlums H, Cichocki F, Tesi B, et al. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity. 2015;42:443–56. This study is paired with the work of Lee et al., and demonstrates that CMV infection can induce epigenetic programs associated with altered functional profiles of NK cells.CrossRefPubMedPubMedCentral Schlums H, Cichocki F, Tesi B, et al. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity. 2015;42:443–56. This study is paired with the work of Lee et al., and demonstrates that CMV infection can induce epigenetic programs associated with altered functional profiles of NK cells.CrossRefPubMedPubMedCentral
16.••
Zurück zum Zitat Lee J, Zhang T, Hwang I, et al. Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity. 2015;42:431–42. Paired with the work of Schlums et al., this study provides evidence for epigenetic changes in NK cells that mediate functional programming and demonstrates the role of antibody in shaping theses responses.CrossRefPubMedPubMedCentral Lee J, Zhang T, Hwang I, et al. Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity. 2015;42:431–42. Paired with the work of Schlums et al., this study provides evidence for epigenetic changes in NK cells that mediate functional programming and demonstrates the role of antibody in shaping theses responses.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Khakoo SI, Thio CL, Martin MP, et al. HLA and NK cell inhibitory receptor genes in resolving hepatitis C virus infection. Science. 2004;305:872–4.CrossRefPubMed Khakoo SI, Thio CL, Martin MP, et al. HLA and NK cell inhibitory receptor genes in resolving hepatitis C virus infection. Science. 2004;305:872–4.CrossRefPubMed
18.
Zurück zum Zitat Martin MP, Gao X, Lee JH, et al. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet. 2002;31:429–34.PubMed Martin MP, Gao X, Lee JH, et al. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet. 2002;31:429–34.PubMed
20.
Zurück zum Zitat Boulet S, Kleyman M, Kim JY, et al. A combined genotype of KIR3DL1 high expressing alleles and HLA-B*57 is associated with a reduced risk of HIV infection. AIDS. 2008;22:1487–91.CrossRefPubMed Boulet S, Kleyman M, Kim JY, et al. A combined genotype of KIR3DL1 high expressing alleles and HLA-B*57 is associated with a reduced risk of HIV infection. AIDS. 2008;22:1487–91.CrossRefPubMed
21.
Zurück zum Zitat Boulet S, Sharafi S, Simic N, et al. Increased proportion of KIR3DS1 homozygotes in HIV-exposed uninfected individuals. AIDS. 2008;22:595–9.CrossRefPubMed Boulet S, Sharafi S, Simic N, et al. Increased proportion of KIR3DS1 homozygotes in HIV-exposed uninfected individuals. AIDS. 2008;22:595–9.CrossRefPubMed
22.
Zurück zum Zitat Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58.CrossRefPubMed Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58.CrossRefPubMed
24.
Zurück zum Zitat Alter G, Martin MP, Teigen N, et al. Differential natural killer cell-mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. J Exp Med. 2007;204:3027–36.CrossRefPubMedPubMedCentral Alter G, Martin MP, Teigen N, et al. Differential natural killer cell-mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. J Exp Med. 2007;204:3027–36.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Holzemer A, Thobakgale CF, Jimenez Cruz CA, et al. Selection of an HLA-C*03:04-Restricted HIV-1 p24 gag sequence variant is associated with viral escape from KIR2DL3+ natural killer cells: data from an observational cohort in South Africa. PLoS Med. 2015;12:e1001900.CrossRefPubMedPubMedCentral Holzemer A, Thobakgale CF, Jimenez Cruz CA, et al. Selection of an HLA-C*03:04-Restricted HIV-1 p24 gag sequence variant is associated with viral escape from KIR2DL3+ natural killer cells: data from an observational cohort in South Africa. PLoS Med. 2015;12:e1001900.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat van Teijlingen NH, Holzemer A, Korner C, et al. Sequence variations in HIV-1 p24 Gag-derived epitopes can alter binding of KIR2DL2 to HLA-C*03:04 and modulate primary natural killer cell function. AIDS. 2014;28:1399–408.CrossRefPubMed van Teijlingen NH, Holzemer A, Korner C, et al. Sequence variations in HIV-1 p24 Gag-derived epitopes can alter binding of KIR2DL2 to HLA-C*03:04 and modulate primary natural killer cell function. AIDS. 2014;28:1399–408.CrossRefPubMed
27.
Zurück zum Zitat Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature. 1998;391:397–401.CrossRefPubMed Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature. 1998;391:397–401.CrossRefPubMed
28.
Zurück zum Zitat Specht A, DeGottardi MQ, Schindler M, Hahn B, Evans DT, Kirchhoff F. Selective downmodulation of HLA-A and -B by Nef alleles from different groups of primate lentiviruses. Virology. 2008;373:229–37.CrossRefPubMed Specht A, DeGottardi MQ, Schindler M, Hahn B, Evans DT, Kirchhoff F. Selective downmodulation of HLA-A and -B by Nef alleles from different groups of primate lentiviruses. Virology. 2008;373:229–37.CrossRefPubMed
29.
Zurück zum Zitat Norman JM, Mashiba M, McNamara LA, et al. The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat Immunol. 2011;12:975–83.CrossRefPubMedPubMedCentral Norman JM, Mashiba M, McNamara LA, et al. The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat Immunol. 2011;12:975–83.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Richard J, Sindhu S, Pham TN, Belzile JP, Cohen EA. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood. 2010;115:1354–63.CrossRefPubMedPubMedCentral Richard J, Sindhu S, Pham TN, Belzile JP, Cohen EA. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood. 2010;115:1354–63.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Shah AH, Sowrirajan B, Davis ZB, et al. Degranulation of natural killer cells following interaction with HIV-1-infected cells is hindered by downmodulation of NTB-A by Vpu. Cell Host Microbe. 2010;8:397–409.CrossRefPubMedPubMedCentral Shah AH, Sowrirajan B, Davis ZB, et al. Degranulation of natural killer cells following interaction with HIV-1-infected cells is hindered by downmodulation of NTB-A by Vpu. Cell Host Microbe. 2010;8:397–409.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Wren LH, Chung AW, Isitman G, et al. Specific antibody-dependent cellular cytotoxicity responses associated with slow progression of HIV infection. Immunology. 2013;138:116–23.CrossRefPubMedPubMedCentral Wren LH, Chung AW, Isitman G, et al. Specific antibody-dependent cellular cytotoxicity responses associated with slow progression of HIV infection. Immunology. 2013;138:116–23.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Lambotte O, Ferrari G, Moog C, et al. Heterogeneous neutralizing antibody and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers. AIDS. 2009;23:897–906.CrossRefPubMedPubMedCentral Lambotte O, Ferrari G, Moog C, et al. Heterogeneous neutralizing antibody and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers. AIDS. 2009;23:897–906.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Ackerman ME, Mikhailova A, Brown EP, et al. Polyfunctional HIV-Specific Antibody Responses Are Associated with Spontaneous HIV Control. PLoS Pathog. 2016;12:e1005315.CrossRefPubMedPubMedCentral Ackerman ME, Mikhailova A, Brown EP, et al. Polyfunctional HIV-Specific Antibody Responses Are Associated with Spontaneous HIV Control. PLoS Pathog. 2016;12:e1005315.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Parsons, M.S., Loh, L., Gooneratne, S., Center, R.J., and Kent, S.J. Role of education and differentiation in determining the potential of natural killer cells to respond to antibody-dependent stimulation. AIDS. 2014;28:2781–6.CrossRef Parsons, M.S., Loh, L., Gooneratne, S., Center, R.J., and Kent, S.J. Role of education and differentiation in determining the potential of natural killer cells to respond to antibody-dependent stimulation. AIDS. 2014;28:2781–6.CrossRef
36.
Zurück zum Zitat Gooneratne SL, Richard J, Lee WS, Finzi A, Kent SJ, Parsons MS. Slaying the Trojan horse: natural killer cells exhibit robust anti-HIV-1 antibody-dependent activation and cytolysis against allogeneic T cells. J Virol. 2015;89:97–109.CrossRefPubMedPubMedCentral Gooneratne SL, Richard J, Lee WS, Finzi A, Kent SJ, Parsons MS. Slaying the Trojan horse: natural killer cells exhibit robust anti-HIV-1 antibody-dependent activation and cytolysis against allogeneic T cells. J Virol. 2015;89:97–109.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Alvarez RA, Hamlin RE, Monroe A, et al. HIV-1 Vpu antagonism of tetherin inhibits antibody-dependent cellular cytotoxic responses by natural killer cells. J Virol. 2014;88:6031–46.CrossRefPubMedPubMedCentral Alvarez RA, Hamlin RE, Monroe A, et al. HIV-1 Vpu antagonism of tetherin inhibits antibody-dependent cellular cytotoxic responses by natural killer cells. J Virol. 2014;88:6031–46.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Arias JF, Heyer LN, von Bredow B, et al. Tetherin antagonism by Vpu protects HIV-infected cells from antibody-dependent cell-mediated cytotoxicity. Proc Natl Acad Sci U S A. 2014;111:6425–30.CrossRefPubMedPubMedCentral Arias JF, Heyer LN, von Bredow B, et al. Tetherin antagonism by Vpu protects HIV-infected cells from antibody-dependent cell-mediated cytotoxicity. Proc Natl Acad Sci U S A. 2014;111:6425–30.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Kramski M, Stratov I, Kent SJ. The role of HIV-specific antibody-dependent cellular cytotoxicity in HIV prevention and the influence of the HIV-1 Vpu protein. AIDS. 2015;29:137–44.CrossRefPubMed Kramski M, Stratov I, Kent SJ. The role of HIV-specific antibody-dependent cellular cytotoxicity in HIV prevention and the influence of the HIV-1 Vpu protein. AIDS. 2015;29:137–44.CrossRefPubMed
40.
Zurück zum Zitat Li SX, Barrett BS, Heilman KJ, et al. Tetherin promotes the innate and adaptive cell-mediated immune response against retrovirus infection in vivo. J Immunol. 2014;193:306–16.CrossRefPubMedPubMedCentral Li SX, Barrett BS, Heilman KJ, et al. Tetherin promotes the innate and adaptive cell-mediated immune response against retrovirus infection in vivo. J Immunol. 2014;193:306–16.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Pham TN, Lukhele S, Hajjar F, Routy JP, Cohen EA. HIV Nef and Vpu protect HIV-infected CD4+ T cells from antibody-mediated cell lysis through down-modulation of CD4 and BST2. Retrovirology. 2014;11:15.CrossRefPubMedPubMedCentral Pham TN, Lukhele S, Hajjar F, Routy JP, Cohen EA. HIV Nef and Vpu protect HIV-infected CD4+ T cells from antibody-mediated cell lysis through down-modulation of CD4 and BST2. Retrovirology. 2014;11:15.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Mi Z, Ding J, Zhang Q, et al. A small molecule compound IMB-LA inhibits HIV-1 infection by preventing viral Vpu from antagonizing the host restriction factor BST-2. Sci Rep. 2015;5:18499.CrossRefPubMedPubMedCentral Mi Z, Ding J, Zhang Q, et al. A small molecule compound IMB-LA inhibits HIV-1 infection by preventing viral Vpu from antagonizing the host restriction factor BST-2. Sci Rep. 2015;5:18499.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol. 2015;15:243–54.CrossRefPubMed Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol. 2015;15:243–54.CrossRefPubMed
45.
Zurück zum Zitat Ripa M, Pogliaghi M, Chiappetta S, et al. Dynamics of adaptive and innate immunity in patients treated during primary human immunodeficiency virus infection: results from Maraviroc in HIV Acute Infection (MAIN) randomized clinical trial. Clin Microbiol Infect. 2015;21(876):e871–874. Ripa M, Pogliaghi M, Chiappetta S, et al. Dynamics of adaptive and innate immunity in patients treated during primary human immunodeficiency virus infection: results from Maraviroc in HIV Acute Infection (MAIN) randomized clinical trial. Clin Microbiol Infect. 2015;21(876):e871–874.
46.
Zurück zum Zitat Jost S, Moreno-Nieves UY, Garcia-Beltran WF, et al. Dysregulated Tim-3 expression on natural killer cells is associated with increased Galectin-9 levels in HIV-1 infection. Retrovirology. 2013;10:74.CrossRefPubMedPubMedCentral Jost S, Moreno-Nieves UY, Garcia-Beltran WF, et al. Dysregulated Tim-3 expression on natural killer cells is associated with increased Galectin-9 levels in HIV-1 infection. Retrovirology. 2013;10:74.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat de Kivit S, Lempsink LJ, Plants J, Martinson J, Keshavarzian A, Landay AL. Modulation of TIM-3 expression on NK and T cell subsets in HIV immunological non-responders. Clin Immunol. 2015;156:28–35.CrossRefPubMed de Kivit S, Lempsink LJ, Plants J, Martinson J, Keshavarzian A, Landay AL. Modulation of TIM-3 expression on NK and T cell subsets in HIV immunological non-responders. Clin Immunol. 2015;156:28–35.CrossRefPubMed
48.
Zurück zum Zitat Mavilio D, Lombardo G, Benjamin J, et al. Characterization of CD56-/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A. 2005;102:2886–91.CrossRefPubMedPubMedCentral Mavilio D, Lombardo G, Benjamin J, et al. Characterization of CD56-/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A. 2005;102:2886–91.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Brunetta E, Hudspeth KL, Mavilio D. Pathologic natural killer cell subset redistribution in HIV-1 infection: new insights in pathophysiology and clinical outcomes. J Leukoc Biol. 2010;88:1119–30.CrossRefPubMed Brunetta E, Hudspeth KL, Mavilio D. Pathologic natural killer cell subset redistribution in HIV-1 infection: new insights in pathophysiology and clinical outcomes. J Leukoc Biol. 2010;88:1119–30.CrossRefPubMed
50.
Zurück zum Zitat Madhavi, V., Ana-Sosa-Batiz, F.E., Jegaskanda, S., Center, R.J., Winnall, W.R., Parsons, M.S., Ananworanich, J., Cooper, D.A., Kelleher, A.D., Hsu, D., et al. Antibody-dependent effector functions against HIV decline in subjects receiving antiretroviral therapy. J Infect Dis. 2015;211:529–38.CrossRef Madhavi, V., Ana-Sosa-Batiz, F.E., Jegaskanda, S., Center, R.J., Winnall, W.R., Parsons, M.S., Ananworanich, J., Cooper, D.A., Kelleher, A.D., Hsu, D., et al. Antibody-dependent effector functions against HIV decline in subjects receiving antiretroviral therapy. J Infect Dis. 2015;211:529–38.CrossRef
51.
Zurück zum Zitat Lisovsky I, Isitman G, Song R, et al. A higher frequency of NKG2A+ than of NKG2A- NK cells responds to autologous HIV-infected CD4 cells irrespective of whether or not they coexpress KIR3DL1. J Virol. 2015;89:9909–19.CrossRefPubMedPubMedCentral Lisovsky I, Isitman G, Song R, et al. A higher frequency of NKG2A+ than of NKG2A- NK cells responds to autologous HIV-infected CD4 cells irrespective of whether or not they coexpress KIR3DL1. J Virol. 2015;89:9909–19.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Naluyima P, Eller MA, Laeyendecker O, et al. Impaired natural killer cell responses are associated with loss of the highly activated NKG2A(+)CD57(+)CD56(dim) subset in HIV-1 subtype D infection in Uganda. AIDS. 2014;28:1273–8.CrossRefPubMedPubMedCentral Naluyima P, Eller MA, Laeyendecker O, et al. Impaired natural killer cell responses are associated with loss of the highly activated NKG2A(+)CD57(+)CD56(dim) subset in HIV-1 subtype D infection in Uganda. AIDS. 2014;28:1273–8.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Schafer JL, Li H, Evans TI, Estes JD, Reeves RK. Accumulation of cytotoxic CD16+ NK cells in simian immunodeficiency virus-infected lymph nodes associated with in situ differentiation and functional anergy. J Virol. 2015;89:6887–94.CrossRefPubMedPubMedCentral Schafer JL, Li H, Evans TI, Estes JD, Reeves RK. Accumulation of cytotoxic CD16+ NK cells in simian immunodeficiency virus-infected lymph nodes associated with in situ differentiation and functional anergy. J Virol. 2015;89:6887–94.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Jost S, Tomezsko PJ, Rands K, et al. CD4+ T-cell help enhances NK cell function following therapeutic HIV-1 vaccination. J Virol. 2014;88:8349–54.CrossRefPubMedPubMedCentral Jost S, Tomezsko PJ, Rands K, et al. CD4+ T-cell help enhances NK cell function following therapeutic HIV-1 vaccination. J Virol. 2014;88:8349–54.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Tomescu C, Mavilio D, Montaner LJ. Lysis of HIV-1-infected autologous CD4+ primary T cells by interferon-alpha-activated NK cells requires NKp46 and NKG2D. AIDS. 2015;29:1767–73.CrossRefPubMed Tomescu C, Mavilio D, Montaner LJ. Lysis of HIV-1-infected autologous CD4+ primary T cells by interferon-alpha-activated NK cells requires NKp46 and NKG2D. AIDS. 2015;29:1767–73.CrossRefPubMed
56.
Zurück zum Zitat Wood SM, Ljunggren HG, Bryceson YT. Insights into NK cell biology from human genetics and disease associations. Cell Mol Life Sci. 2011;68:3479–93.CrossRefPubMed Wood SM, Ljunggren HG, Bryceson YT. Insights into NK cell biology from human genetics and disease associations. Cell Mol Life Sci. 2011;68:3479–93.CrossRefPubMed
57.
Zurück zum Zitat Mace EM, Hsu AP, Monaco-Shawver L, et al. Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56(bright) subset. Blood. 2013;121:2669–77.CrossRefPubMedPubMedCentral Mace EM, Hsu AP, Monaco-Shawver L, et al. Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56(bright) subset. Blood. 2013;121:2669–77.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Gineau L, Cognet C, Kara N, et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J Clin Invest. 2012;122:821–32.CrossRefPubMedPubMedCentral Gineau L, Cognet C, Kara N, et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J Clin Invest. 2012;122:821–32.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Grier JT, Forbes LR, Monaco-Shawver L, et al. Human immunodeficiency-causing mutation defines CD16 in spontaneous NK cell cytotoxicity. J Clin Invest. 2012;122:3769–80.CrossRefPubMedPubMedCentral Grier JT, Forbes LR, Monaco-Shawver L, et al. Human immunodeficiency-causing mutation defines CD16 in spontaneous NK cell cytotoxicity. J Clin Invest. 2012;122:3769–80.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Moraru M, Cisneros E, Gomez-Lozano N, et al. Host genetic factors in susceptibility to herpes simplex type 1 virus infection: contribution of polymorphic genes at the interface of innate and adaptive immunity. J Immunol. 2012;188:4412–20.CrossRefPubMed Moraru M, Cisneros E, Gomez-Lozano N, et al. Host genetic factors in susceptibility to herpes simplex type 1 virus infection: contribution of polymorphic genes at the interface of innate and adaptive immunity. J Immunol. 2012;188:4412–20.CrossRefPubMed
61.
Zurück zum Zitat Moraru M, Black LE, Muntasell A, et al. NK cell and Ig interplay in defense against Herpes Simplex Virus Type 1: epistatic interaction of CD16A and IgG1 allotypes of variable affinities modulates antibody-dependent cellular cytotoxicity and susceptibility to clinical reactivation. J Immunol. 2015;195:1676–84.CrossRefPubMed Moraru M, Black LE, Muntasell A, et al. NK cell and Ig interplay in defense against Herpes Simplex Virus Type 1: epistatic interaction of CD16A and IgG1 allotypes of variable affinities modulates antibody-dependent cellular cytotoxicity and susceptibility to clinical reactivation. J Immunol. 2015;195:1676–84.CrossRefPubMed
62.
Zurück zum Zitat Voss, M., and Bryceson, Y.T. Natural killer cell biology illuminated by primary immunodeficiency syndromes in humans. Clin Immunol. 2015. Voss, M., and Bryceson, Y.T. Natural killer cell biology illuminated by primary immunodeficiency syndromes in humans. Clin Immunol. 2015.
63.
64.••
Zurück zum Zitat Conlon KC, Lugli E, Welles HC, et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J Clin Oncol. 2015;33:74–82. This provides first-in-human demonstration of the safety and effects of in vivo administration of IL-15 on NK cell dynamics.CrossRefPubMedPubMedCentral Conlon KC, Lugli E, Welles HC, et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J Clin Oncol. 2015;33:74–82. This provides first-in-human demonstration of the safety and effects of in vivo administration of IL-15 on NK cell dynamics.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Bergamaschi C, Kulkarni V, Rosati M, et al. Intramuscular delivery of heterodimeric IL-15 DNA in macaques produces systemic levels of bioactive cytokine inducing proliferation of NK and T cells. Gene Ther. 2015;22:76–86.CrossRefPubMedPubMedCentral Bergamaschi C, Kulkarni V, Rosati M, et al. Intramuscular delivery of heterodimeric IL-15 DNA in macaques produces systemic levels of bioactive cytokine inducing proliferation of NK and T cells. Gene Ther. 2015;22:76–86.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Seay K, Church C, Zheng JH, et al. In vivo activation of human NK cells by treatment with an interleukin-15 superagonist potently inhibits acute in vivo HIV-1 infection in humanized mice. J Virol. 2015;89:6264–74.CrossRefPubMedPubMedCentral Seay K, Church C, Zheng JH, et al. In vivo activation of human NK cells by treatment with an interleukin-15 superagonist potently inhibits acute in vivo HIV-1 infection in humanized mice. J Virol. 2015;89:6264–74.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Benson Jr DM, Bakan CE, Mishra A, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116:2286–94.CrossRefPubMedPubMedCentral Benson Jr DM, Bakan CE, Mishra A, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116:2286–94.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Wu X, Shao Y, Tao Y, et al. Proteasome inhibitor lactacystin augments natural killer cell cytotoxicity of myeloma via downregulation of HLA class I. Biochem Biophys Res Commun. 2011;415:187–92.CrossRefPubMed Wu X, Shao Y, Tao Y, et al. Proteasome inhibitor lactacystin augments natural killer cell cytotoxicity of myeloma via downregulation of HLA class I. Biochem Biophys Res Commun. 2011;415:187–92.CrossRefPubMed
70.
Zurück zum Zitat Kohrt HE, Thielens A, Marabelle A, et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood. 2014;123:678–86.CrossRefPubMedPubMedCentral Kohrt HE, Thielens A, Marabelle A, et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood. 2014;123:678–86.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Childs RW, Carlsten M. Therapeutic approaches to enhance natural killer cell cytotoxicity against cancer: the force awakens. Nat Rev Drug Discov. 2015;14:487–98.CrossRefPubMed Childs RW, Carlsten M. Therapeutic approaches to enhance natural killer cell cytotoxicity against cancer: the force awakens. Nat Rev Drug Discov. 2015;14:487–98.CrossRefPubMed
72.
Zurück zum Zitat Beziat V, Liu LL, Malmberg JA, et al. NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs. Blood. 2013;121:2678–88.CrossRefPubMedPubMedCentral Beziat V, Liu LL, Malmberg JA, et al. NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs. Blood. 2013;121:2678–88.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Lopez-Verges S, Milush JM, Schwartz BS, et al. Expansion of a unique CD57(+)NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc Natl Acad Sci U S A. 2011;108:14725–32.CrossRefPubMedPubMedCentral Lopez-Verges S, Milush JM, Schwartz BS, et al. Expansion of a unique CD57(+)NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc Natl Acad Sci U S A. 2011;108:14725–32.CrossRefPubMedPubMedCentral
74.
75.
Zurück zum Zitat Nielsen CM, White MJ, Goodier MR, Riley EM. Functional significance of CD57 expression on human NK cells and relevance to disease. Front Immunol. 2013;4:422.CrossRefPubMedPubMedCentral Nielsen CM, White MJ, Goodier MR, Riley EM. Functional significance of CD57 expression on human NK cells and relevance to disease. Front Immunol. 2013;4:422.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Cerwenka, A., and Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat Rev Immunol. 2016. Cerwenka, A., and Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat Rev Immunol. 2016.
77.
Zurück zum Zitat O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006;7:507–16.CrossRefPubMed O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006;7:507–16.CrossRefPubMed
78.
Zurück zum Zitat Paust S, Gill HS, Wang BZ, et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat Immunol. 2010;11:1127–35.CrossRefPubMedPubMedCentral Paust S, Gill HS, Wang BZ, et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat Immunol. 2010;11:1127–35.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296:1323–6.CrossRefPubMed Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296:1323–6.CrossRefPubMed
81.
Zurück zum Zitat Smith HR, Heusel JW, Mehta IK, et al. Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc Natl Acad Sci U S A. 2002;99:8826–31.CrossRefPubMedPubMedCentral Smith HR, Heusel JW, Mehta IK, et al. Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc Natl Acad Sci U S A. 2002;99:8826–31.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, Yokoyama WM. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci U S A. 2009;106:1915–9.CrossRefPubMedPubMedCentral Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, Yokoyama WM. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci U S A. 2009;106:1915–9.CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Luetke-Eversloh M, Hammer Q, Durek P, et al. Human cytomegalovirus drives epigenetic imprinting of the IFNG locus in NKG2Chi natural killer cells. PLoS Pathog. 2014;10:e1004441.CrossRefPubMedPubMedCentral Luetke-Eversloh M, Hammer Q, Durek P, et al. Human cytomegalovirus drives epigenetic imprinting of the IFNG locus in NKG2Chi natural killer cells. PLoS Pathog. 2014;10:e1004441.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Guma M, Angulo A, Vilches C, Gomez-Lozano N, Malats N, Lopez-Botet M. Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. Blood. 2004;104:3664–71.CrossRefPubMed Guma M, Angulo A, Vilches C, Gomez-Lozano N, Malats N, Lopez-Botet M. Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. Blood. 2004;104:3664–71.CrossRefPubMed
86.
Zurück zum Zitat Foley B, Cooley S, Verneris MR, et al. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J Immunol. 2012;189:5082–8.CrossRefPubMedPubMedCentral Foley B, Cooley S, Verneris MR, et al. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J Immunol. 2012;189:5082–8.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat Rolle A, Pollmann J, Ewen EM, et al. IL-12-producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion. J Clin Invest. 2014;124:5305–16.CrossRefPubMedPubMedCentral Rolle A, Pollmann J, Ewen EM, et al. IL-12-producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion. J Clin Invest. 2014;124:5305–16.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Zhou J, Amran FS, Kramski M, et al. An NK cell population lacking FcRgamma is expanded in chronically infected HIV patients. J Immunol. 2015;194:4688–97.CrossRefPubMed Zhou J, Amran FS, Kramski M, et al. An NK cell population lacking FcRgamma is expanded in chronically infected HIV patients. J Immunol. 2015;194:4688–97.CrossRefPubMed
89.
Zurück zum Zitat Kleinnijenhuis J, Quintin J, Preijers F, et al. BCG-induced trained immunity in NK cells: role for non-specific protection to infection. Clin Immunol. 2014;155:213–9.CrossRefPubMed Kleinnijenhuis J, Quintin J, Preijers F, et al. BCG-induced trained immunity in NK cells: role for non-specific protection to infection. Clin Immunol. 2014;155:213–9.CrossRefPubMed
90.
Zurück zum Zitat Robbins SH, Bessou G, Cornillon A, et al. Natural killer cells promote early CD8 T cell responses against cytomegalovirus. PLoS Pathog. 2007;3:e123.CrossRefPubMedPubMedCentral Robbins SH, Bessou G, Cornillon A, et al. Natural killer cells promote early CD8 T cell responses against cytomegalovirus. PLoS Pathog. 2007;3:e123.CrossRefPubMedPubMedCentral
91.
Zurück zum Zitat Andrews DM, Estcourt MJ, Andoniou CE, et al. Innate immunity defines the capacity of antiviral T cells to limit persistent infection. J Exp Med. 2010;207:1333–43.CrossRefPubMedPubMedCentral Andrews DM, Estcourt MJ, Andoniou CE, et al. Innate immunity defines the capacity of antiviral T cells to limit persistent infection. J Exp Med. 2010;207:1333–43.CrossRefPubMedPubMedCentral
92.
Zurück zum Zitat Schuster IS, Wikstrom ME, Brizard G, et al. TRAIL+ NK cells control CD4+ T cell responses during chronic viral infection to limit autoimmunity. Immunity. 2014;41:646–56.CrossRefPubMed Schuster IS, Wikstrom ME, Brizard G, et al. TRAIL+ NK cells control CD4+ T cell responses during chronic viral infection to limit autoimmunity. Immunity. 2014;41:646–56.CrossRefPubMed
93.
Zurück zum Zitat Alter G, Kavanagh D, Rihn S, et al. IL-10 induces aberrant deletion of dendritic cells by natural killer cells in the context of HIV infection. J Clin Invest. 2010;120:1905–13.CrossRefPubMedPubMedCentral Alter G, Kavanagh D, Rihn S, et al. IL-10 induces aberrant deletion of dendritic cells by natural killer cells in the context of HIV infection. J Clin Invest. 2010;120:1905–13.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Lang PA, Lang KS, Xu HC, et al. Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. Proc Natl Acad Sci U S A. 2012;109:1210–5.CrossRefPubMedPubMedCentral Lang PA, Lang KS, Xu HC, et al. Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. Proc Natl Acad Sci U S A. 2012;109:1210–5.CrossRefPubMedPubMedCentral
95.
Zurück zum Zitat Waggoner SN, Cornberg M, Selin LK, Welsh RM. Natural killer cells act as rheostats modulating antiviral T cells. Nature. 2012;481:394–8.PubMedCentral Waggoner SN, Cornberg M, Selin LK, Welsh RM. Natural killer cells act as rheostats modulating antiviral T cells. Nature. 2012;481:394–8.PubMedCentral
96.
97.••
Zurück zum Zitat Rydyznski C, Daniels KA, Karmele EP, et al. Generation of cellular immune memory and B-cell immunity is impaired by natural killer cells. Nat Commun. 2015;6:6375. This study adds to the data on NK cell editing of T cell responses demonstrating the role of NK cells in shaping the development of immunologic memory through antibody generation.CrossRefPubMedPubMedCentral Rydyznski C, Daniels KA, Karmele EP, et al. Generation of cellular immune memory and B-cell immunity is impaired by natural killer cells. Nat Commun. 2015;6:6375. This study adds to the data on NK cell editing of T cell responses demonstrating the role of NK cells in shaping the development of immunologic memory through antibody generation.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Perreau M, Savoye AL, De Crignis E, et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med. 2013;210:143–56.CrossRefPubMedPubMedCentral Perreau M, Savoye AL, De Crignis E, et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med. 2013;210:143–56.CrossRefPubMedPubMedCentral
99.
Zurück zum Zitat Luteijn R, Sciaranghella G, van Lunzen J, et al. Early viral replication in lymph nodes provides HIV with a means by which to escape NK-cell-mediated control. Eur J Immunol. 2011;41:2729–40.CrossRefPubMed Luteijn R, Sciaranghella G, van Lunzen J, et al. Early viral replication in lymph nodes provides HIV with a means by which to escape NK-cell-mediated control. Eur J Immunol. 2011;41:2729–40.CrossRefPubMed
Metadaten
Titel
NK Cells in HIV Disease
verfasst von
Eileen Scully
Galit Alter
Publikationsdatum
21.03.2016
Verlag
Springer US
Erschienen in
Current HIV/AIDS Reports / Ausgabe 2/2016
Print ISSN: 1548-3568
Elektronische ISSN: 1548-3576
DOI
https://doi.org/10.1007/s11904-016-0310-3

Weitere Artikel der Ausgabe 2/2016

Current HIV/AIDS Reports 2/2016 Zur Ausgabe

HIV Pathogenesis and Treatment (AL Landay, Section Editor)

How to Define the Latent Reservoir: Tools of the Trade

The Science of Prevention (JD Stekler and J Baeten, Section Editors)

HIV Self-Testing: a Review of Current Implementation and Fidelity

The Science of Prevention (JD Stekler and J Baeten, Section Editors)

Current and (Potential) Future Effects of the Affordable Care Act on HIV Prevention

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.