scholarly journals Involvement of Activating NK Cell Receptors and Their Modulation in Pathogen Immunity

2011 ◽  
Vol 2011 ◽  
pp. 1-11 ◽  
Author(s):  
Francesco Marras ◽  
Federica Bozzano ◽  
Andrea De Maria

Natural Killer (NK) cells are endowed with cell-structure-sensing receptors providing inhibitory protection from self-destruction (inhibitory NK receptors, iNKRs, including killer inhibitory receptors and other molecules) and rapid triggering potential leading to functional cell activation by Toll-like receptors (TLRs), cytokine receptors, and activating NK cell receptors including natural cytotoxicity receptors (NCRs, i.e., NKp46, NKp46, and NKp44). NCR and NKG2D recognize ligands on infected cells which may be endogenous or may directly bind to some structures derived from invading pathogens. In this paper, we address the known direct or indirect interactions between activating receptors and pathogens and their expression during chronic HIV and HCV infections.

2015 ◽  
Vol 89 (19) ◽  
pp. 9909-9919 ◽  
Author(s):  
Irene Lisovsky ◽  
Gamze Isitman ◽  
Rujun Song ◽  
Sandrina DaFonseca ◽  
Alexandra Tremblay-McLean ◽  
...  

ABSTRACTEpidemiological and functional studies implicate NK cells in HIV control. However, there is little information available on which NK cell populations, as defined by the inhibitory NK cell receptors (iNKRs) they express, respond to autologous HIV-infected CD4+(iCD4) T cells. NK cells acquire antiviral functions through education, which requires signals received from iNKRs, such as NKG2A and KIR3DL1 (here, 3DL1), engaging their ligands. NKG2A interacts with HLA-E, and 3DL1 interacts with HLA-A/B antigens expressing the Bw4 epitope. HIV-infected cells downregulate HLA-A/B, which should interrupt negative signaling through 3DL1, leading to NK cell activation, provided there is sufficient engagement of activating NKRs. We examined the functionality of NK cells expressing or not NKG2A and 3DL1 stimulated by HLA-null and autologous iCD4 cells. Flow cytometry was used to gate on each NKG2A+/NKG2A−3DL1+/3DL1−(NKG2A+/−3DL1+/−) population and to measure the frequency of all possible combinations of CD107a expression and gamma interferon (IFN-γ) and CCL4 secretion. The highest frequency of functional NK cells responding to HLA-null cell stimulation was the NKG2A+3DL1+NK cell population. The highest frequencies of functional NK cells responding to autologous iCD4 cells were those expressing NKG2A; coexpression of 3DL1 did not further modulate responsiveness. This was the case for the functional subsets characterized by the sum of all functions tested (total responsiveness), as well as by the trifunctional CD107a+IFN-γ+CCL4+, CD107a+IFN-γ+, total CD107a+, and total IFN-γ+functional subsets. These results indicate that the NKG2A receptor has a role in NK cell-mediated anti-HIV responses.IMPORTANCEHIV-infected CD4 (iCD4) cells activate NK cells, which then control HIV replication. However, little is known regarding which NK cell populations iCD4 cells stimulate to develop antiviral activity. Here, we examine the frequency of NK cell populations, defined by the presence/absence of the NK cell receptors (NKRs) NKG2A and 3DL1, that respond to iCD4 cells. NKG2A and 3DL1 are involved in priming NK cells for antiviral functions upon encountering virus-infected cells. A higher frequency of NKG2A+than NKG2A−NK cells responded to iCD4 cells by developing antiviral functions such as CD107a expression, which correlates with NK cell killing, and secretion of gamma interferon and CCL4. Coexpression of 3DL1 on the NKG2A+and NKG2A−NK cells did not modulate responses to iCD4 cells. Understanding the mechanisms underlying the interaction of NK cells with iCD4 cells that lead to HIV control may contribute to developing strategies that harness NK cells for preventing or controlling HIV infection.


Viruses ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 156
Author(s):  
Jasmina M. Luczo ◽  
Sydney L. Ronzulli ◽  
Stephen M. Tompkins

Natural killer (NK) cells are part of the innate immunity repertoire, and function in the recognition and destruction of tumorigenic and pathogen-infected cells. Engagement of NK cell activating receptors can lead to functional activation of NK cells, resulting in lysis of target cells. NK cell activating receptors specific for non-major histocompatibility complex ligands are NKp46, NKp44, NKp30, NKG2D, and CD16 (also known as FcγRIII). The natural cytotoxicity receptors (NCRs), NKp46, NKp44, and NKp30, have been implicated in functional activation of NK cells following influenza virus infection via binding with influenza virus hemagglutinin (HA). In this review we describe NK cell and influenza A virus biology, and the interactions of influenza A virus HA and other pathogen lectins with NK cell natural cytotoxicity receptors (NCRs). We review concepts which intersect viral immunology, traditional virology and glycobiology to provide insights into the interactions between influenza virus HA and the NCRs. Furthermore, we provide expert opinion on future directions that would provide insights into currently unanswered questions.


2021 ◽  
Vol 22 (13) ◽  
pp. 6670
Author(s):  
Eva Prašnikar ◽  
Andrej Perdih ◽  
Jure Borišek

The innate immune system’s natural killer (NK) cells exert their cytolytic function against a variety of pathological challenges, including tumors and virally infected cells. Their activation depends on net signaling mediated via inhibitory and activating receptors that interact with specific ligands displayed on the surfaces of target cells. The CD94/NKG2C heterodimer is one of the NK activating receptors and performs its function by interacting with the trimeric ligand comprised of the HLA-E/β2m/nonameric peptide complex. Here, simulations of the all-atom multi-microsecond molecular dynamics in five immune complexes provide atomistic insights into the receptor–ligand molecular recognition, as well as the molecular events that facilitate the NK cell activation. We identify NKG2C, the HLA-Eα2 domain, and the nonameric peptide as the key elements involved in the molecular machinery of signal transduction via an intertwined hydrogen bond network. Overall, the study addresses the complex intricacies that are necessary to understand the mechanisms of the innate immune system.


2004 ◽  
Vol 34 (4) ◽  
pp. 961-971 ◽  
Author(s):  
Pascale André ◽  
Roberta Castriconi ◽  
Marion Espéli ◽  
Nicolas Anfossi ◽  
Tiffany Juarez ◽  
...  

Blood ◽  
2002 ◽  
Vol 100 (6) ◽  
pp. 1935-1947 ◽  
Author(s):  
Sherif S. Farag ◽  
Todd A. Fehniger ◽  
Loredana Ruggeri ◽  
Andrea Velardi ◽  
Michael A. Caligiuri

AbstractNatural killer (NK) cells have held great promise for the immunotherapy of cancer for more than 3 decades. However, to date only modest clinical success has been achieved manipulating the NK cell compartment in patients with malignant disease. Progress in the field of NK cell receptors has revolutionized our concept of how NK cells selectively recognize and lyse tumor and virally infected cells while sparing normal cells. Major families of cell surface receptors that inhibit and activate NK cells to lyse target cells have been characterized, including killer cell immunoglobulinlike receptors (KIRs), C-type lectins, and natural cytotoxicity receptors (NCRs). Further, identification of NK receptor ligands and their expression on normal and transformed cells completes the information needed to begin development of rational clinical approaches to manipulating receptor/ligand interactions for clinical benefit. Indeed, clinical data suggest that mismatch of NK receptors and ligands during allogeneic bone marrow transplantation may be used to prevent leukemia relapse. Here, we review how NK cell receptors control natural cytotoxicity and novel approaches to manipulating NK receptor-ligand interactions for the potential benefit of patients with cancer.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1764-1764 ◽  
Author(s):  
Jens Pahl ◽  
Uwe Reusch ◽  
Thorsten Gantke ◽  
Anne Kerber ◽  
Joachim Koch ◽  
...  

Abstract Introduction: AFM13 is an NK-cell engaging CD30/CD16A bispecific tetravalent TandAb antibody currently in phase 2 clinical development in Hodgkin lymphoma (HL) and other CD30+ malignancies. It engages NK-cells through CD16A with high affinity and specificity and confers significantly stronger NK-cell activation compared to other therapeutic antibodies. We have previously shown synergistic efficacy when NK-cell activation by AFM13 is combined with check-point modulation such as anti-PD-1 treatment, which is known to unleash T cell and NK-cell activity. The goal of this study was to identify further candidates for combination treatments and biomarkers that potentially indicate NK-cell responses to AFM13 treatment. Methods: AFM13-mediated NK-cell cytotoxicity and IFN-γ production after 4-hour interaction with HL cell lines was measured by 51Cr release assays and flow cytometry, respectively. Expression of NK-cell receptors, NK-cell proliferation (CFSE dilution) and expansion (absolute cell counts) was analyzed by flow cytometry. Results: The interaction of NK-cells with AFM13-coated tumor cells up-regulated the expression of NK-cell receptors such as CD25, CD69, CD137/4-1BB as well as molecules that may serve as NK-cell check-points when compared with the unrelated NK-cell binding TandAb AFM12 that does not bind to target cells. Importantly, CD16A engagement by AFM13 enhanced the proliferation and expansion potential of NK-cells when subsequently incubated with IL-15 or with particularly low doses of IL-2. NK-cell cytotoxicity and IFN-γ production was substantially increased towards CD30+ tumor cells in the presence of AFM13. Even target cells resistant to naïve and IL-2/IL-15-activated NK-cells were susceptible to AFM13-induced NK-cell cytotoxicity. AFM13 concentrations of as low as 10-2 µg/mL resulted in maximal activity while AFM13 was significantly more potent than native anti-CD30 IgG1 antibody. NK-cell activation by IL-2 or IL-15 had a synergistic effect on AFM13-mediated cytotoxicity. Conclusion: AFM13 specifically enhances the cytotoxic, proliferative and cytokine-producing potential of NK-cells. Our data indicate that the distinctive modulation of NK-cell receptors can be utilized to monitor NK-cell responses during AFM13 therapy and provides candidates for therapeutic combination strategies. Moreover, the combination with low doses of IL-2 or with IL-15 may expand the quantity of tumor-reactive NK-cells after AFM13 treatment and promote NK-cell functionality in the tumor microenvironment in cancer patients. Disclosures Reusch: Affimed: Employment, Patents & Royalties: Patents. Gantke:Affimed GmbH: Employment. Kerber:Affimed: Employment. Koch:Affimed: Employment. Treder:Affimed: Employment. Cerwenka:Affimed: Research Funding.


2020 ◽  
Vol 2020 ◽  
pp. 1-14 ◽  
Author(s):  
Yingying Chen ◽  
Dan Lu ◽  
Alexey Churov ◽  
Rong Fu

Natural killer cells (NK cells) play an important role in innate immunity. NK cells recognize self and nonself depending on the balance of activating receptors and inhibitory receptors. After binding to their ligands, NK cell receptors trigger subsequent signaling conduction and then determine whether NK is activated or inhibited. Furthermore, NK cell response includes cytotoxicity and cytokine release, which is tightly related to the activation of NK cell-activating receptors and the inhibition of inhibitory receptors on the surfaces of NK cells. The expression and function of NK cell surface receptors also alter in virus infection, tumor, and autoimmune diseases and influence the occurrence and development of diseases. So, it is important to understand the mechanism of recognition between NK receptors and their ligands in pathological conditions and the signaling pathways of NK cell receptors. This review mainly summarizes the research progress on NK cell surface receptors and their signal pathways.


2014 ◽  
Vol 32 (4_suppl) ◽  
pp. 95-95
Author(s):  
Christine Pasero ◽  
Gwenaelle Gravis ◽  
Mathilde Guerin ◽  
Palma Rocchi ◽  
Jeanne Thomassin ◽  
...  

95 Background: Immunotherapy is now investigated as a promising alternative treatment for patients (pts) with metastatic prostate cancer (PC). Natural killer (NK) cells are powerful effector cells with antitumoral activity and their role have been explored in solid tumors but not yet in prostate cancer. NK cell cytotoxicity is regulated by a balance between activating and inhibitory receptors. Here, we performed a restrospective study to evaluate the link between NK cells and the time of castration response in newly diagnosed PC patients with metastases. Methods: Newly diagnosed metastatic PC pts were divided according the time of castration response, with an 18-months cutoff value: 18 pts with long castration response (LCR, median = 64.6 months), and 14 pts with short castration response ([SCR] median = 11.2 months), with a median overall survival of 97.7 months and 33.8 months respectively. Circulating NK cells from these patients were studied by flow cytometry to evaluate the expression of activating receptors and the NK cell functionality. Results: We observed thatNK cells from LCR pts express higher levels of the maturation marker CD57 (43.3% vs. 23.3% positive cells, p= 0.002), the receptor CD16 involved in cytotoxicity (29,124 vs. 16,806 MFI, p= 0.02), and the activating receptors NKp46 and NKp30 (17.5 vs. 11.4 RMFI, p= 0.0146 , and 10.9 vs. 6.3 RMFI, p = 0.0128 respectively) than NK cells from SCR pts. This suggests that LCR pts have powerful NK cells. Indeed, NK cells from LCR pts are highly efficient in CD107 functional assay than NK cells from SCR pts (28.9% vs. 19.4%, p =0.002). In vitro blocking experiments show that NKp46 is precisely one of the NK cell receptors involved in the NK-mediated recognition of prostate tumor cells, thus higher expression of NKp46 would help to control PC progression. Conclusions: Together our results show for the first time that efficient NK cells are associated to a long response to castration and prolonged survival in newly diagnosed metastatic PC. NK cell receptors might be useful as predictive biomarkers in metastatic PC, to help in stratification of patients and to design NK cell–based immunotherapeutic strategies for PC.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 61-61
Author(s):  
Qin Tang ◽  
Bartosz Grzywacz ◽  
Hongbo Wang ◽  
Nandini Kataria ◽  
John E. Wagner ◽  
...  

Abstract The natural cytotoxicity receptors (NCRs), NKp30, NKp44 and NKp46, are a recently identified group of receptors that play a central role in NK cell killing of malignant cells. Previous studies show that the NCRs are restricted to the NK cell lineage. However, we noted that a small fraction of freshly isolated umbilical cord blood (UCB) T cells expressed NKp30 (1.7±0.9%, n=8), but not NKp44 (0.4±0.4%, n=8) or NKp46 (0.5±0.4%, n=8). Thus, we investigated whether NCRs could be induced on either UCB or adult peripheral blood (PB) T cells after culture in IL-15 for 14 days. This cytokine was chosen since previous studies show that IL-15 induces other NK cell receptors on both UCB and PB T cells. Surprisingly, UCB, but not PB T cells, acquired NKp30 (37±10% vs 4±2%, p=0.01), NKp44 (41±20% vs. 1±0.6%, p=0.01) and NKp46 (13±7% vs 0.6±0.5%, p=0.01). NCRs were found mainly on CD8+ or CD3+CD56+ UCB T cells. Further studies addressed whether other cytokines could induce NCR expression on UCB T cells. Both IL-2 and IL-15 showed a dose dependent induction of NCRs. In contrast, IL-7 induced only NKp30 on UCB T cells, but in a non-dose dependent manner. IL-4 abrogated NCR expression, even in the presence of IL-15. Considering that the ligands for the NCRs are not yet elucidated, we performed functional studies using agonist mAbs and NK cells served as controls. Surprisingly, while all three NCRs were expressed on IL-15 expanded UCB T cells, only NKp30 was functional as demonstrated by degranulation (CD107a), IFN-γ release, and redirected killing assays (reverse ADCC). Previous studies show that the NKp30, −44 and −46 cooperate to induce NK killing. However, the simultaneous triggering of all 3 NCRs did not increase UCB T cell cytotoxicity. Some NK cell receptors modulate TCR triggering, but this was not the case with NCRs. We did find that another NK cell receptor, NKG2D, enhanced the cytoxic triggering of NKp30, but not other NCRs. To address the lack of function of NKp44 and −46 on UCB T cells, the expression of adapter proteins required for signaling through these receptors were determined. The proximal receptor for NKp44 signaling, DAP12, was absent in UCB T cells. Thus, the lack of DAP12 likely explains the absence of NKp44 signaling. Both of the adapters used by NKp30 and −46 (FcεR1γ and CD3ζ) were detected. As stated above, NKp46 was expressed on significantly fewer cells than NKp30 (13±7% vs 37±10%) and this likely accounts for the lack of NKp46 function. Considering that UCB contains mostly naive T cells, we hypothesized that PB naive T cells may also acquire NCRs. To test this, PB T cell subsets (naive, central memory and effector memory) were FACS sorted and cultured for 14 days in IL-15. NKp30 was induced on a small number of naive, but not memory PB T cells. Unlike UCB T cells, NKp30 on naive PB T cells was non-functional. PB T cells lacked FcεR1γ, while UCB T cells expressed it, suggesting that this adapter protein is critical for NKp30 signaling. Collectively, these results show that UCB T cells are unique in their ability to acquire NCRs. Moreover, only NKp30 is functional. The lack of function of NKp44 and -46 is due to the absence of DAP12 and the low level of NKp46 expression, respectively. Lastly, while some naive PB T cells can acquire NKp30, it is not functional due to the lack of FcεR1γ. Such studies highlight the differences between UCB and PB T cells and challenge the dogma that NCRs are NK cell specific.


Sign in / Sign up

Export Citation Format

Share Document