scholarly journals License to Kill: When iNKT Cells Are Granted the Use of Lethal Cytotoxicity

2020 ◽  
Vol 21 (11) ◽  
pp. 3909
Author(s):  
Angélica Díaz-Basabe ◽  
Francesco Strati ◽  
Federica Facciotti

Invariant Natural Killer T (iNKT) cells are a non-conventional, innate-like, T cell population that recognize lipid antigens presented by the cluster of differentiation (CD)1d molecule. Although iNKT cells are mostly known for mediating several immune responses due to their massive and diverse cytokine release, these cells also work as effectors in various contexts thanks to their cytotoxic potential. In this Review, we focused on iNKT cell cytotoxicity; we provide an overview of iNKT cell subsets, their activation cues, the mechanisms of iNKT cell cytotoxicity, the specific roles and outcomes of this activity in various contexts, and how iNKT killing functions are currently activated in cancer immunotherapies. Finally, we discuss the future perspectives for the better understanding and potential uses of iNKT cell killing functions in tumor immunosurveillance.

mBio ◽  
2016 ◽  
Vol 7 (6) ◽  
Author(s):  
Adeline Barthelemy ◽  
Stoyan Ivanov ◽  
Maya Hassane ◽  
Josette Fontaine ◽  
Béatrice Heurtault ◽  
...  

ABSTRACT Influenza A virus infection can predispose to potentially devastating secondary bacterial infections. Invariant natural killer T (iNKT) cells are unconventional, lipid-reactive T lymphocytes that exert potent immunostimulatory functions. Using a mouse model of postinfluenza invasive secondary pneumococcal infection, we sought to establish whether α-galactosylceramide (α-GalCer [a potent iNKT cell agonist that is currently in clinical development]) could limit bacterial superinfection. Our results highlighted the presence of a critical time window during which α-GalCer treatment can trigger iNKT cell activation and influence resistance to postinfluenza secondary pneumococcal infection. Intranasal treatment with α-GalCer during the acute phase (on day 7) of influenza virus H3N2 and H1N1 infection failed to activate (gamma interferon [IFN-γ] and interleukin-17A [IL-17A]) iNKT cells; this effect was associated with a strongly reduced number of conventional CD103 + dendritic cells in the respiratory tract. In contrast, α-GalCer treatment during the early phase (on day 4) or during the resolution phase (day 14) of influenza was associated with lower pneumococcal outgrowth and dissemination. Less intense viral-bacterial pneumonia and a lower morbidity rate were observed in superinfected mice treated with both α-GalCer (day 14) and the corticosteroid dexamethasone. Our results open the way to alternative (nonantiviral/nonantibiotic) iNKT-cell-based approaches for limiting postinfluenza secondary bacterial infections. IMPORTANCE Despite the application of vaccination programs and antiviral drugs, influenza A virus (IAV) infection is responsible for widespread morbidity and mortality (500,000 deaths/year). Influenza infections can also result in sporadic pandemics that can be devastating: the 1918 pandemic led to the death of 50 million people. Severe bacterial infections are commonly associated with influenza and are significant contributors to the excess morbidity and mortality of influenza. Today’s treatments of secondary bacterial (pneumococcal) infections are still not effective enough, and antibiotic resistance is a major issue. Hence, there is an urgent need for novel therapies. In the present study, we set out to evaluate the efficacy of α-galactosylceramide (α-GalCer)—a potent agonist of invariant NKT cells that is currently in clinical development—in a mouse model of postinfluenza, highly invasive pneumococcal pneumonia. Our data indicate that treatment with α-GalCer reduces susceptibility to superinfections and, when combined with the corticosteroid dexamethasone, reduces viral-bacterial pneumonia.


2021 ◽  
Author(s):  
Priya Khurana ◽  
Chakkapong Burudpakdee ◽  
Stephan A. Grupp ◽  
Ulf H. Beier ◽  
David M. Barrett ◽  
...  

ABSTRACTInvariant natural killer T (iNKT) cells comprise a unique subset of lymphocytes that are primed for activation and possess innate NK-like functional features. Currently, iNKT cell-based immunotherapies remain in early clinical stages, and little is known about the ability of these cells to survive and retain effector functions within the solid tumor microenvironment (TME) long-term. In conventional T cells (TCONV), cellular metabolism is linked to effector functions and their ability to adapt to the nutrient-poor TME. In contrast, the bioenergetic requirements of iNKT cells – particularly those of human iNKT cells – at baseline and upon stimulation are not well understood; neither is how these requirements affect cytokine production or anti-tumor effector functions. We find that unlike TCONV, human iNKT cells are not dependent upon glucose or glutamine for cytokine production and cytotoxicity upon stimulation with anti-CD3 and anti-CD28. Additionally, transcriptional profiling revealed that stimulated human iNKT cells are less glycolytic than TCONV and display higher expression of fatty acid oxidation (FAO) and adenosine monophosphate-activated protein kinase (AMPK) pathway genes. Furthermore, stimulated iNKT cells displayed higher mitochondrial mass and membrane potential relative to TCONV. Real-time Seahorse metabolic flux analysis revealed that stimulated human iNKT cells utilize fatty acids as substrates for oxidation more than stimulated TCONV. Together, our data suggest that human iNKT cells possess different bioenergetic requirements from TCONV and display a more memory-like metabolic program relative to effector TCONV. Importantly, iNKT cell-based immunotherapeutic strategies could co-opt such unique features of iNKT cells to improve their efficacy and longevity of anti-tumor responses.


2019 ◽  
Vol 3 (5) ◽  
pp. 813-824 ◽  
Author(s):  
Rupali Das ◽  
Peng Guan ◽  
Susan J. Wiener ◽  
Nishant P. Patel ◽  
Trevor G. Gohl ◽  
...  

Abstract Invariant natural killer T (iNKT) cells comprise a unique lineage of CD1d-restricted lipid-reactive T lymphocytes that potently kill tumor cells and exhibit robust immunostimulatory functions. Optimal tumor-directed iNKT cell responses often require expression of the antigen-presenting molecule CD1d on tumors; however, many tumor cells downregulate CD1d and thus evade iNKT cell recognition. We generated a soluble bispecific fusion protein designed to direct iNKT cells to the site of B-cell cancers in a tumor antigen-specific but CD1d-independent manner. This fusion protein is composed of a human CD1d molecule joined to a single chain antibody FV fragment specific for CD19, an antigen widely expressed on B-cell cancers. The CD1d-CD19 fusion protein binds specifically to CD19-expressing, but not CD19-negative cells. Once loaded with the iNKT cell lipid agonist α-galactosyl ceramide (αGC), the CD1d-CD19 fusion induces robust in vitro activation of and cytokine production by human iNKT cells. iNKT cells stimulated by the αGC-loaded CD1d-CD19 fusion also strongly transactivate T-, B-, and NK-cell responses and promote dendritic cell maturation. Importantly, the αGC-loaded fusion induces robust lysis of CD19+CD1d− Epstein-Barr virus immortalized human B-lymphoblastoid cell lines that are otherwise resistant to iNKT cell killing. Consistent with these findings; administration of the αGC-loaded fusion protein controlled the growth of CD19+CD1d− tumors in vivo, suggesting that it can “link” iNKT cells and CD19+CD1d− targets in a therapeutically beneficial manner. Taken together, these preclinical studies demonstrate that this B cell–directed fusion protein can be used to effectively induce iNKT cell antitumor responses in vitro and in vivo.


2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Roman Covarrubias ◽  
Amy S Major

Invariant Natural Killer T (iNKT) cells are specialized lymphocytes that when activated can regulate chronic inflammatory conditions and atherosclerotic processes. The activation of iNKT cells occurs when glycolipid antigens bind the MHC class-I like molecule CD1d present on antigen presenting cells (APCs). The pathways by which glycolipid antigens target CD1d for presentation and activation of iNKT cells remain unclear, yet the expression of surface receptors associated with lipid homeostasis, such as the LDL receptor (LDLr), have been implicated in the modulation of iNKT cell activation. The LDLr has been shown to modulate this process by binding apoE-containing lipoproteins, which can carry antigenic glycolipids for iNKT cell activation. The LDL receptor-related protein (LRP), a transmembrane receptor from the LDL receptor family of proteins, shares structural homology with LDLr and can bind a number of ligands including apoE-containing lipoproteins. We hypothesized that LRP can play an active role in glycolipid antigen presentation and subsequent activation of iNKT cells. Here, we demonstrate that LRP is preferentially expressed at high levels on F4/80 + macrophages, when compared to other APCs. We also show that a specialized subset of macrophages expressing CD169, known for their ability to present glycolipid antigen to iNKT cells, have increased levels of LRP when compared to CD169 - macrophages. Using mice with a targeted deletion of LRP in macrophages, we observed decreased activation of iNKT cells in vitro (24, 48 hours) and normal IFN-gamma but blunted IL-4 response in vivo. Further flow cytometric analysis showed normal surface expression of CD1d in LRP-cKO macrophages as well as normal uptake of fluorescently labeled glycolipid in vitro . Additionally, analysis of the iNKT cell compartment in LRP-cKO mice revealed intact numbers and percentages of iNKT cells and no homeostatic disruption as evidenced by absence of programmed death-1 and LY-49. Collectively, these data suggest that macrophage LRP contributes to early iNKT cell activation by enhancing early IL-4 responses.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5104-5104
Author(s):  
Weihua Song ◽  
Hans J.J. van der Vliet ◽  
Yu-Tzu Tai ◽  
Ruojie Wang ◽  
Rao Prabhala ◽  
...  

Abstract Invariant NKT (iNKT) cells are important immunoregulatory cells that recognize glycolipid antigens with CD1d restriction and contribute to antitumor immune responses through the production of IFN-γ and IL-2. However, in progressive multiple myeloma (MM), the iNKT cell population is decreased along with its capacity to produce IFN-γ. Thus, a novel strategy for the immunotherapy of MM entails the enhancement of iNKT cell functions. In this study, we established iNKT cell lines from MM patients via enrichment with Vα24+ and subsequently with Vβ11+ cells, followed by several rounds of stimulation with α-GalCer-pulsed DCs. These techniques resulted in highly purified iNKT cell lines (>97%). To evaluate potential in vivo interaction between iNKT cells and myeloma cells, we evaluated the CD1d expression on primary myeloma cells as well as MM cell lines. Gene expression profiling revealed compared to normal plasma cells, majority of primary MM cells (11 out of 15) expressed higher levels of CD1d; in contrast, all 6 MM cell lines tested had no expression. Flow cytometric analysis further confirmed the expression of CD1d on primary MM cells and lack of its expression on 12 different MM cell lines. A CD1d-transfected MM1S cell line (MM1S-CD1d) was therefore established for the functional study. To determine whether CD1d-expressing primary MM cells have the antigen presenting capacity, iNKT cell lines from healthy donors (n=2) and MM patients (n=2) were cocultured with 5 cases of CD1d positive primary MM cells with or without α-GalCer. Monitored by the CD25 expression, we demonstrated primary MM cells presented α-GalCer and also endogenous antigen(s) to activate iNKT cells. We have further evaluated the functional profile of expanded iNKT cell lines from MM patients (n=5). Upon stimulation with α-GalCer-pulsed MM.1S-CD1d cells, iNKT cells produced high levels of Th1-type cytokines (IFN-γ and IL-2) compared to low level Th2-type cytokine production (IL-4). Our results thus demonstrate that iNKT cell lines from MM patients were functionally restored by expansion with α-GalCer-pulsed DCs in vitro. To further augment iNKT cells function, we evaluated effects of lenalidomide on iNKT cell lines, an immunomodulatory drug which has been demonstrated to enhance T cell costimulation and NK cell activity. Lenalidomide did not directly stimulate iNKT cells in the presence or absence of α-GalCel. Importantly, upon CD1d-restricted activation by α-GalCer-loaded MM1S-CD1d cells, lenalidomide significantly enhanced the Th1-type immune responses of iNKT cell lines from both healthy donors and MM patients. Compared to those of controls, a significant increase of IFN- γ (healthy donor, p< 0.001, n=7; MM patients, p<0.05, n=3) and IL-2 (MM patients, p<0.0015, n=3) occurred. Meanwhile, lenalidomide had no significant effect on the production of IL-4 by iNKT cell lines (healthy donor, p>0.05, n=7; MM patients, p>0.05, n=3). Taken together, our results provide preclinical feasibility and support a rationale to evaluate efficacy of adoptive transfer of iNKT cells in MM. Moreover, it provides a clinical basis for use of lenalidomide to enhance iNKT cell mediated immunotherapy in myeloma.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 346-346
Author(s):  
Marie T Rubio ◽  
Lucia Teixeira ◽  
Pierre Milpied ◽  
Emmanuel Bachy ◽  
Felipe Suarez ◽  
...  

Abstract Abstract 346 Introduction: Invariant Natural Killer T (iNKT) cells express a highly restricted T cell receptor (TCR) repertoire composed of a single invariant Va14Ja18 chain in mice and a Va24Ja18 chain in humans. In contrast to their conventional counterpart, iNKT cells recognize CD1d-bound glycolipids rather than peptides. iNKT cells have elicited a lively interest in the last years because of their implication in several immune responses including experimental graft-versus-host-disease (GVHD). In this study, we addressed the question whether human iNKT cells could be associated with the outcome of allogeneic haematopoietic stem cell transplantation (HSCT). Materials and methods: Forty-seven patients allografted for diverse hematological malignancies in our institution entered prospectively the study. We sequentially analyzed the reconstitution of peripheral blood CD1d tetramer + iNKT, CD56+ NK, and CD4+, CD8+, CD4+CD25high T cells for 90 days after allogeneic HSCT by flow cytometry. Results were correlated to the clinical evolution of allogeneic HSCT. Results: We observed two groups of patients according to their iNKT/106 T cell ratios on days 30, 60 and 90 after HSCT. Patients with at least one point above the normal ratio of 1000 iNKT/106 T cells were considered in the iNKT high group (n=17), while those with a poor reconstitution, defined by all points below that threshold, were analyzed in the iNKT low group (n=30). iNKT reconstitution was the only significant difference in terms of immune reconstitution between the two groups since reconstitution of T CD4+, CD8+, or CD4+CD25high and NK cells was similar at all time points in both groups. Chimerism, analyzed by PCR amplification of short tandem repeat markers, showed that in all cases iNKT displayed a 100% donor origin. Pre-transplant characteristics of patients were similar between the two groups except for the conditioning regimen and the source of stem cells. Patients in the iNKT high group had more often received a reduced-intensity conditioning (80.5% versus 46.7%, p=0.004) and peripheral blood stem cells (88.5% versus 53.6%, p=0.015). Occurrence and severity of acute GVHD was significantly reduced in the iNKT high group (23.5 % grade I-II and 0% grade III-IV) compared to the iNKT low group (66.7 % grade I-II and 26.6% grade III-IV), (p<0.001), while development of chronic GVHD was not significantly affected by the iNKT reconstitution (29.4 % in the iNKT high group versus 48.2% in the iNKT low group), (p=0.57). This lead to a significantly reduced toxicity related mortality (TRM) in the iNKT high group (8.2% versus 36.9% in the iNKT low group at 2 years), (p=0.028) without an increased risk of relapse (17.6% in the iNKT high group versus 16.7% in the iNKT low group) at 2 years, (p=0.877). Median follow up was 28.7 months. Overall survival was improved in the iNKT high group (86.3% versus 58.0% in the iNKT low group, at 2 years), (p=0.056) in a landmark analysis at day 90. These results prompted us to further analyze whether iNKT cell frequency could be a predictive marker of acute GVHD. In a subgroup analysis performed on patients with availabel data on day 14 post-HSCT (n=23), the risk of developing acute GVHD was significantly higher in patients with a iNKT/106 T cell ratio below 500 (92 % of aGVHD) in comparison to those with a ratio above 500 (40 % of aGVHD), (p=0.0028). Conclusion: Our data suggest that donor-derived iNKT cell reconstitution after allogeneic HSCT may represent a predictive factor of acute GVHD and a prognostic factor of transplant outcome. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3659-3659
Author(s):  
Chuanhui Xu ◽  
Riemer de Vries ◽  
Lydia Visser ◽  
Arjan Diepstra ◽  
Stephan D Gadola ◽  
...  

Abstract Abstract 3659 Poster Board III-595 Introduction Hodgkin lymphoma (HL) is a B-cell neoplasm characterized by a minority of neoplastic cells, the so-called Hodgkin and Reed-Sternberg (HRS) cells. The HRS cells are located within an extensive infiltrate of reactive cells, including T cells, B cells, plasma cells, stromal cells, eosinophils and macrophages. CD1 molecules are non-classical MHC I-like molecules that present lipid antigens to T cells, triggering a specific immune response. Of the five CD1 isoforms (CD1a, CD1b, CD1c, CD1d and CD1e) expressed in human tissue, only CD1c and CD1d are expressed in B cells. The T cell receptors (TCRs) of T cells that recognize CD1c are indistinguishable from those that recognize MHC class I or II complexes. In contrast, CD1d presents lipid antigens to invariant Natural Killer T cells (iNKT cells), which are characterized by the expression of a semi-invariant Vα24Jα18 chain plus a limited set of β chains. CD4− and CD4+ iNKT cells were demonstrated as two distinct functional subsets in terms of cytokine production and cytotoxic activation. Immunoregulatory CD1 restricted T cells have been implicated in the pathogenesis of many cancers, but its role in HL is still unknown. In this study, we analyzed the expression of CD1c, CD1d and presence of iNKT cells in HL. Methods Expression of CD1c and CD1d was determined by immunocytochemistry in four HL cell lines KMH2, L1236, L428 and U-HO1. CD1c and CD1d expression in both HRS cells and reactive cells was studied in 46 HL patient tissues by immunohistochemistry. Cell suspensions of ten HL cases were studied for the presence of iNKT cells using monoclonal antibodies against human iNKT cell (clone 6B11) and TCR Vβ11 (clone C21) by flow cytometry. The percentage of CD4+ iNKT cells was determined in the gated iNKT cell population. Results Cytoplasmic CD1d expression was found in four HL cell lines and membranous CD1d expression was found in KMH2 and L1236. CD1c expression was negative in all four HL cell lines. CD1d expression was detected in HRS cells in 46% (21/46) of the HL cases. No correlation was observed between CD1d expression and presence of EBV in HRS cells. In contrast, CD1c expression was negative in HRS cells in all HL cases. Both CD1c and CD1d were detected in reactive cells in all HL cases albeit at varying frequencies. The mean percentage of CD1d restricted iNKT cells was 4% (range 0.8-8%) in HL cell suspensions irrespective of CD1d expression status in HRS cells. In reactive lymph node (RLN) the mean percentage of iNKT cells was also 4% (range 0.4-7%). Approximately half of the iNKT cells were CD4+ in both HL and RLN cell suspensions. Conclusion Expression of CD1d was found in four HL cell lines and in HRS cells of ∼50% of the HL cases. In HL cell suspensions, about 4% (range 0.8-8%) of the reactive background cells were iNKT cells. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 210 (6) ◽  
pp. 1079-1086 ◽  
Author(s):  
Zuoan Yi ◽  
Laura L. Stunz ◽  
Gail A. Bishop

TCR signaling is a prerequisite for early stage development of invariant natural killer T (iNKT) cells, whereas IL-15 signaling is required for expansion and maturation at later stages. In this study, we show that TNF receptor associated factor 3 (TRAF3) plays a critical role in the transition between these two distinct signaling pathways and developmental stages. TRAF3-deficient iNKT cells in CD4CreTRAF3flox/flox (T-TRAF3−/−) mice exhibit defective up-regulation of T-bet and CD122, two critical molecules for IL-15 signaling, and as a consequence, IL-15–mediated iNKT cell proliferation and survival are impaired. Consistently, development of iNKT cells in T-TRAF3−/− mice shows a major defect at developmental stages 2 and 3, but not stages 0 and 1. We further demonstrated that defective T-bet up-regulation occurring during the stage 1 to stage 2 transition results from reduced TCR signaling in TRAF3−/− iNKT cells. In addition, mature TRAF3−/− iNKT cells displayed defective cytokine responses upon TCR stimulation. Collectively, our results reveal that by modulating the relative strength of TCR signaling, TRAF3 is an important regulator of iNKT cell development and functions.


2009 ◽  
Vol 206 (6) ◽  
pp. 1365-1378 ◽  
Author(s):  
Jean-Marc Doisne ◽  
Laurent Bartholin ◽  
Kai-Ping Yan ◽  
Céline N. Garcia ◽  
Nadia Duarte ◽  
...  

Invariant natural killer T (iNKT) cells constitute a distinct subset of T lymphocytes exhibiting important immune-regulatory functions. Although various steps of their differentiation have been well characterized, the factors controlling their development remain poorly documented. Here, we show that TGF-β controls the differentiation program of iNKT cells. We demonstrate that TGF-β signaling carefully and specifically orchestrates several steps of iNKT cell development. In vivo, this multifaceted role of TGF-β involves the concerted action of different pathways of TGF-β signaling. Whereas the Tif-1γ branch controls lineage expansion, the Smad4 branch maintains the maturation stage that is initially repressed by a Tif-1γ/Smad4-independent branch. Thus, these three different branches of TGF-β signaling function in concert as complementary effectors, allowing TGF-β to fine tune the iNKT cell differentiation program.


2019 ◽  
Vol 70 (9) ◽  
pp. 1865-1874 ◽  
Author(s):  
Naomi F Walker ◽  
Charles Opondo ◽  
Graeme Meintjes ◽  
Nishtha Jhilmeet ◽  
Jon S Friedland ◽  
...  

Abstract Background Tuberculosis (TB) is the leading cause of mortality and morbidity in people living with human immunodeficiency virus (HIV) infection (PLWH). PLWH with TB disease are at risk of the paradoxical TB-associated immune reconstitution inflammatory syndrome (TB-IRIS) when they commence antiretroviral therapy. However, the pathophysiology is incompletely understood and specific therapy is lacking. We investigated the hypothesis that invariant natural killer T (iNKT) cells contribute to innate immune dysfunction associated with TB-IRIS. Methods In a cross-sectional study of 101 PLWH and HIV-uninfected South African patients with active TB and controls, iNKT cells were enumerated using α-galactosylceramide-loaded CD1d tetramers and subsequently functionally characterized by flow cytometry. In a second study of 49 people with HIV type 1 (HIV-1) and active TB commencing antiretroviral therapy, iNKT cells in TB-IRIS patients and non-IRIS controls were compared longitudinally. Results Circulating iNKT cells were reduced in HIV-1 infection, most significantly the CD4+ subset, which was inversely associated with HIV-1 viral load. iNKT cells in HIV-associated TB had increased surface CD107a expression, indicating cytotoxic degranulation. Relatively increased iNKT cell frequency in patients with HIV-1 infection and active TB was associated with development of TB-IRIS following antiretroviral therapy initiation. iNKT cells in TB-IRIS were CD4+CD8– subset depleted and degranulated around the time of TB-IRIS onset. Conclusions Reduced iNKT cell CD4+ subsets as a result of HIV-1 infection may skew iNKT cell functionality toward cytotoxicity. Increased CD4– cytotoxic iNKT cells may contribute to immunopathology in TB-IRIS.


Sign in / Sign up

Export Citation Format

Share Document