28 research outputs found

    Etude des mécanismes régulateurs des cellules NK : rôle de la molécule PD-1 et de la prostaglandine E2 (PGE2)

    Get PDF
    Natural Killer (NK) cells are effectors of the innate immune system, and play a crucial role in virus and cancer immunesurveillance. To escape NK-cell mediated elimination of infected or transformed cells, viruses and tumors have developed multiple strategies to interfere with NK-cell functions. In the present study, we investigated the role of two regulatory molecules, Programmed Death-1 (PD-1) and Prostaglandine E2 (PGE2), in controlling NK cell activation and effector functions. PD-1 is a key immune checkpoint receptor expressed by activated T and B lymphocytes. Upon interaction with its cognate ligands, PD-1 inhibits lymphocyte proliferation and functions. During cancer or chronic viral infections, PD-1 expression is associated with functional exhaustion of effector T cells. Blockade of PD-1 signaling restores T-cell functions, and represents a promising therapeutic tool. We fortuitously observed unusual expression of PD-1 on a subset of CD56dim NK cells in some patients with persistent viral infection (HHV8, HIV or HCV). We show that PD-1 expression on NK cells characterizes a subpopulation of recently activated cells (CD69+, CD25+, Nkp44+) that are sensitive to apoptosis (Annexin V+) and not senescent (CD57-). NKp46 expression was also markedly decreased on PD-1+ NK cells. In vitro functional experiments showed that PD-1+ NK cells had impaired cytotoxic capacity (CD107a degranulation) and reduced IFN-γ production compared to their PD-1- counterpart, suggesting that they might represent functionally exhausted NK cells. Interestingly, exogenous IL-2 and IL-15 could restore PD-1+ NK cell effector functions. While strong non-specific stimulation by PMA/ionomycin transiently induced PD-1 on control NK cells, only activation through NKp46 or NKp30 receptors in the presence of IL-15 could reproducibly induce stable PD-1 expression. To investigate the effect of PD-1 expression on NK cells in the absence of any confounding factor related to the underlying disease, we generated NK cells stably expressing PD-1 after lentiviral transduction. Compared to NK cells transduced with the control vector, PD-1+ cells showed a constitutively decreased CD107a degranulation, thus confirming our findings in NK cells from infected patients. In the second part of this study, we investigated the immunosuppressive role of PGE2 in the control of IL-15-mediated NK cell activation. Our results suggest that PGE2 acts through EP2 and EP4 receptors to inhibit IL-15 induced NKG2D and IL-15Rγ expression on NK cells. These findings allow a better comprehension of PGE2/IL-15 antagonism in the regulation of NK cell responses. In conclusion, our results indicate that PD-1 expression on NK cells could represent a supplementary mechanism of immune evasion strategy, and allow introducing the concept of exhausted NK cells, similar to exhausted PD-1+ T cells. Our results also demonstrate that PGE2 exerts a negative feedback on IL-15-mediated effects on NK cells. Blocking PGE2 or its receptors could be of interest in IL-15 tumor immunotherapy to potentiate IL-15-induced cytotoxic functions of NK cells.Les cellules Natural Killer (NK) sont des effecteurs de l’immunité innée et constituent de véritables sentinelles dans l’immuno-surveillance contre les virus et les processus tumoraux. Pour échapper à la reconnaissance NK, les virus ainsi que les cellules tumorales utilisent de nombreux subterfuges. Au cours de ce travail, nous nous sommes intéressés à deux facteurs régulateurs de la fonction des cellules NK, la molécule PD-1 et la prostaglandine E2 (PGE2). Programmed death 1 (PD-1) est une molécule régulatrice exprimée sur les lymphocytes T et B activés. L’engagement de PD-1 par ses ligands inhibe leurs fonctions effectrices et prolifératives. Au cours de certaines infections virales chroniques ou de tumeurs, l’expression de PD-1 est associée à l'épuisement fonctionnel des lymphocytes T effecteurs. Le blocage de l’axe PD-1/PD-1 ligands restaure les fonctions effectrices des lymphocytes et représente ainsi une approche thérapeutique prometteuse. Nous avons mis en évidence de manière fortuite une expression inhabituelle du récepteur PD-1 sur les cellules NK au cours de certaines infections virales chroniques (HHV8, VIH ou VHC). L’expression de PD-1 sur les cellules NK caractérise une population récemment activée (CD69++, CD25 + et Nkp44 +), susceptible à l’apoptose (Annexin V+), exprimant une moindre quantité du récepteur de cytotoxicité naturelle NKp46. L’analyse fonctionnelle montre que les cellules NK PD-1+ ont des capacités de cytotoxicité (dégranulation CD107a) et de production de cytokines (IFNγ) réduites en comparaison avec leurs homologues PD-1-. De façon intéressante, l’IL-2 et l’IL-15 peuvent restaurer les fonctions effectrices des cellules NK PD-1+. Contrairement aux cellules T dont l’expression de PD-1 est induite par divers stimuli, seule la stimulation des récepteurs NKp46 et NKp30, en synergie avec l’action de l’IL-15 ou l’IL-2, induit in vitro de façon reproductible l’expression de PD-1 sur les cellules NK de témoins sains. Pour pouvoir disposer d’un modèle in vitro de cellules NK PD-1+, nous avons généré des cellules exprimant PD-1 de manière stable par transduction lentivirale de la lignée NKL. En comparaison avec les cellules transduites par le vecteur vide, les cellules NKL PD-1+ ont des capacités cytotoxiques réduites, confirmant nos résultats chez les patients. Nous avons aussi étudié les mécanismes par lesquels la PGE2, une autre molécule immuno-modularice, régule les fonctions des cellules NK. Nos résultats suggèrent que la PGE2 agit à travers ses récepteurs EP2 et EP4 pour inhiber l’expression de NKG2D et de l’IL-15Rγ induite par l’IL-15 sur les cellules NK. Ce travail doit nous permettre de mieux comprendre comment la PGE2 s’oppose aux effets activateurs de l’IL-15 sur les cellules NK, et représente un mécanisme de rétrocontrôle de l’inflammation. En conclusion, nos résultats montrent que l’expression de PD-1 sur les cellules NK représente un mécanisme supplémentaire d’échappement viral à la réponse immune. La suite de l’étude sur la PGE2 devrait nous permettre d’évaluer l’intérêt de l’utilisation d’antagonistes spécifiques dans l’immunothérapie anti-tumorale par l’IL-15. Le lien entre la PGE2 et l’expression de PD-1 sur les cellules NK est en cours d’investigation

    Regulation of NK cells immune response : role of PD-1 and PGE2

    No full text
    Les cellules Natural Killer (NK) sont des effecteurs de l’immunité innée et constituent de véritables sentinelles dans l’immuno-surveillance contre les virus et les processus tumoraux. Pour échapper à la reconnaissance NK, les virus ainsi que les cellules tumorales utilisent de nombreux subterfuges. Au cours de ce travail, nous nous sommes intéressés à deux facteurs régulateurs de la fonction des cellules NK, la molécule PD-1 et la prostaglandine E2 (PGE2). Programmed death 1 (PD-1) est une molécule régulatrice exprimée sur les lymphocytes T et B activés. L’engagement de PD-1 par ses ligands inhibe leurs fonctions effectrices et prolifératives. Au cours de certaines infections virales chroniques ou de tumeurs, l’expression de PD-1 est associée à l'épuisement fonctionnel des lymphocytes T effecteurs. Le blocage de l’axe PD-1/PD-1 ligands restaure les fonctions effectrices des lymphocytes et représente ainsi une approche thérapeutique prometteuse. Nous avons mis en évidence de manière fortuite une expression inhabituelle du récepteur PD-1 sur les cellules NK au cours de certaines infections virales chroniques (HHV8, VIH ou VHC). L’expression de PD-1 sur les cellules NK caractérise une population récemment activée (CD69++, CD25 + et Nkp44 +), susceptible à l’apoptose (Annexin V+), exprimant une moindre quantité du récepteur de cytotoxicité naturelle NKp46. L’analyse fonctionnelle montre que les cellules NK PD-1+ ont des capacités de cytotoxicité (dégranulation CD107a) et de production de cytokines (IFNγ) réduites en comparaison avec leurs homologues PD-1-. De façon intéressante, l’IL-2 et l’IL-15 peuvent restaurer les fonctions effectrices des cellules NK PD-1+. Contrairement aux cellules T dont l’expression de PD-1 est induite par divers stimuli, seule la stimulation des récepteurs NKp46 et NKp30, en synergie avec l’action de l’IL-15 ou l’IL-2, induit in vitro de façon reproductible l’expression de PD-1 sur les cellules NK de témoins sains. Pour pouvoir disposer d’un modèle in vitro de cellules NK PD-1+, nous avons généré des cellules exprimant PD-1 de manière stable par transduction lentivirale de la lignée NKL. En comparaison avec les cellules transduites par le vecteur vide, les cellules NKL PD-1+ ont des capacités cytotoxiques réduites, confirmant nos résultats chez les patients. Nous avons aussi étudié les mécanismes par lesquels la PGE2, une autre molécule immuno-modularice, régule les fonctions des cellules NK. Nos résultats suggèrent que la PGE2 agit à travers ses récepteurs EP2 et EP4 pour inhiber l’expression de NKG2D et de l’IL-15Rγ induite par l’IL-15 sur les cellules NK. Ce travail doit nous permettre de mieux comprendre comment la PGE2 s’oppose aux effets activateurs de l’IL-15 sur les cellules NK, et représente un mécanisme de rétrocontrôle de l’inflammation. En conclusion, nos résultats montrent que l’expression de PD-1 sur les cellules NK représente un mécanisme supplémentaire d’échappement viral à la réponse immune. La suite de l’étude sur la PGE2 devrait nous permettre d’évaluer l’intérêt de l’utilisation d’antagonistes spécifiques dans l’immunothérapie anti-tumorale par l’IL-15. Le lien entre la PGE2 et l’expression de PD-1 sur les cellules NK est en cours d’investigation.Natural Killer (NK) cells are effectors of the innate immune system, and play a crucial role in virus and cancer immunesurveillance. To escape NK-cell mediated elimination of infected or transformed cells, viruses and tumors have developed multiple strategies to interfere with NK-cell functions. In the present study, we investigated the role of two regulatory molecules, Programmed Death-1 (PD-1) and Prostaglandine E2 (PGE2), in controlling NK cell activation and effector functions. PD-1 is a key immune checkpoint receptor expressed by activated T and B lymphocytes. Upon interaction with its cognate ligands, PD-1 inhibits lymphocyte proliferation and functions. During cancer or chronic viral infections, PD-1 expression is associated with functional exhaustion of effector T cells. Blockade of PD-1 signaling restores T-cell functions, and represents a promising therapeutic tool. We fortuitously observed unusual expression of PD-1 on a subset of CD56dim NK cells in some patients with persistent viral infection (HHV8, HIV or HCV). We show that PD-1 expression on NK cells characterizes a subpopulation of recently activated cells (CD69+, CD25+, Nkp44+) that are sensitive to apoptosis (Annexin V+) and not senescent (CD57-). NKp46 expression was also markedly decreased on PD-1+ NK cells. In vitro functional experiments showed that PD-1+ NK cells had impaired cytotoxic capacity (CD107a degranulation) and reduced IFN-γ production compared to their PD-1- counterpart, suggesting that they might represent functionally exhausted NK cells. Interestingly, exogenous IL-2 and IL-15 could restore PD-1+ NK cell effector functions. While strong non-specific stimulation by PMA/ionomycin transiently induced PD-1 on control NK cells, only activation through NKp46 or NKp30 receptors in the presence of IL-15 could reproducibly induce stable PD-1 expression. To investigate the effect of PD-1 expression on NK cells in the absence of any confounding factor related to the underlying disease, we generated NK cells stably expressing PD-1 after lentiviral transduction. Compared to NK cells transduced with the control vector, PD-1+ cells showed a constitutively decreased CD107a degranulation, thus confirming our findings in NK cells from infected patients. In the second part of this study, we investigated the immunosuppressive role of PGE2 in the control of IL-15-mediated NK cell activation. Our results suggest that PGE2 acts through EP2 and EP4 receptors to inhibit IL-15 induced NKG2D and IL-15Rγ expression on NK cells. These findings allow a better comprehension of PGE2/IL-15 antagonism in the regulation of NK cell responses. In conclusion, our results indicate that PD-1 expression on NK cells could represent a supplementary mechanism of immune evasion strategy, and allow introducing the concept of exhausted NK cells, similar to exhausted PD-1+ T cells. Our results also demonstrate that PGE2 exerts a negative feedback on IL-15-mediated effects on NK cells. Blocking PGE2 or its receptors could be of interest in IL-15 tumor immunotherapy to potentiate IL-15-induced cytotoxic functions of NK cells

    Control of NK Cell Activation by Immune Checkpoint Molecules

    No full text
    The development of cancer and chronic infections is facilitated by many subversion mechanisms, among which enhanced expression of immune checkpoints molecules, such as programmed death-1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), on exhausted T cells. Recently, immune checkpoint inhibitors have shown remarkable efficiency in the treatment of a number of cancers. However, expression of immune checkpoints on natural killer (NK) cells and its functional consequences on NK cell effector functions are much less explored. In this review, we focus on the current knowledge on expression of various immune checkpoints in NK cells, how it can alter NK cell-mediated cytotoxicity and cytokine production. Dissecting the role of these inhibitory mechanisms in NK cells is critical for the full understanding of the mode of action of immunotherapies using checkpoint inhibitors in the treatment of cancers and chronic infections

    High effector-memory CD8+ T-cell levels correlate with high PML risk in natalizumab-treated patients

    No full text
    International audienceBackground: Progressive multifocal leukoencephalopathy (PML) is a severe complication of natalizumab (NTZ) treatment in multiple sclerosis (MS) patients. Based on the analysis of cryopreserved cells, several reports have showed that CD62L+ CD4+ T-cells percentage drops before PML onset.Objective: To analyze CD62L and CD45RA expression on fresh-blood CD4+ and CD8+ T-cells from NTZ-treated patients, according to their estimated PML risk.Methods: We prospectively enrolled 74 MS patients, including 62 NTZ-treated, and stratified them into low, intermediate and high PML risk groups. Circulating naïve and memory T-cell subsets were analyzed by flow cytometry.Results: We found no correlation between the percentage of CD62L+ CD4+ T-cells and PML risk. In contrast, the repartition of CD8+ T-cells subpopulations was altered in the high risk group: both the percentage and absolute count of CD8+ CD62L- CD45RA- effector memory T- cells (TEM) was significantly higher compared to patients at lower risk despite similar CD3+ and CD8+ T-cell counts. One high-risk patient with elevated CD8+ TEM and CD62L+ CD4+ T-cell levels developed PML six months after sampling.Conclusion: Our results suggest that CD8+ TEM cells should be evaluated in larger studies as a potential surrogate marker of PML risk in NTZ-treated patients

    Abnormal repression of SHP-1, SHP-2 and SOCS-1 transcription sustains the activation of the JAK/STAT3 pathway and the progression of the disease in multiple myeloma.

    No full text
    Sustained activation of JAK/STAT3 signaling pathway is classically described in Multiple Myeloma (MM). One explanation could be the silencing of the JAK/STAT suppressor genes, through the hypermethylation of SHP-1 and SOCS-1, previously demonstrated in MM cell lines or in whole bone marrow aspirates. The link between such suppressor gene silencing and the degree of bone marrow invasion or the treatment response has not been evaluated in depth. Using real-time RT-PCR, we studied the expression profile of three JAK/STAT suppressor genes: SHP-1, SHP-2 and SOCS-1 in plasma cells freshly isolated from the bone marrows of MM patients and healthy controls. Our data demonstrated an abnormal repression of such genes in malignant plasma cells and revealed a significant correlation between such defects and the sustained activation of the JAK/STAT3 pathway during MM. The repressed expression of SHP-1 and SHP-2 correlated significantly with a high initial degree of bone marrow infiltration but was, unexpectedly, associated with a better response to the induction therapy. Collectively, our data provide new evidences that substantiate the contribution of JAK/STAT suppressor genes in the pathogenesis of MM. They also highlight the possibility that the decreased gene expression of SHP-1 and SHP-2 could be of interest as a new predictive factor of a favorable treatment response, and suggest new potential mechanisms of action of the therapeutic molecules. Whether such defect helps the progression of the disease from monoclonal gammopathy of unknown significance to MM remains, however, to be determined

    PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma

    No full text
    International audienceProgrammed Death-1 (PD-1), an inhibitory receptor expressed by activated lymphocytes, is involved in regulating T-and B-cell responses. PD-1 and its ligands are exploited by a variety of cancers to facilitate tumor escape through PD-1-mediated functional exhaustion of effector T cells. Here, we report that PD-1 is upregulated on Natural Killer (NK) cells from patients with Kaposi sarcoma (KS). PD-1 was expressed in a sub-population of activated, mature CD56(dim)CD16(pos) NK cells with otherwise normal expression of NK surface receptors. PD-1(pos) NK cells from KS patients were hyporesponsive ex vivo following direct triggering of NKp30, NKp46 or CD16 activating receptors, or short stimulation with NK cell targets. PD-1(pos) NK cells failed to degranulate and release IFN gamma, but exogenous IL-2 or IL-15 restored this defect. That PD-1 contributed to NK cell functional impairment and was not simply a marker of dysfunctional NK cells was confirmed in PD-1-transduced NKL cells. In vitro, PD-1 was induced at the surface of healthy control NK cells upon prolonged contact with cells expressing activating ligands, i. e. a condition mimicking persistent stimulation by tumor cells. Thus, PD-1 appears to plays a critical role in mediating NK cell exhaustion. The existence of this negative checkpoint fine-tuning NK activation highlights the possibility that manipulation of the PD-1 pathway may be a strategy for circumventing tumor escape not only from the T cell-, but also the NK-cell mediated immune surveillance

    Human Apoptotic Cells, Generated by Extracorporeal Photopheresis, Modulate Allogeneic Immune Response

    No full text
    International audienceThe induction of specific and sustainable tolerance is a challenging issue in organ transplantation. The discovery of the immunosuppressive properties of apoptotic cells in animal models has paved the way for their use in human transplantation. In this work, we aimed to define a stable, reproducible, and clinically compatible production procedure of human apoptotic cells (Apo-cells). Using a clinically approved extracorporeal photopheresis technique, we have produced and characterized phenotypically and functionally human apoptotic cells. These Apo-cells have immunosuppressive properties proved in vitro and in vivo in NOD/SCID/γC mice by their capacity to modulate an allogeneic response following both a direct and an indirect antigen presentation. These results brought the rationale for the use of Apo-cells in tolerance induction protocol for organ transplantation

    <i>SHP-1</i> and <i>SHP-2</i> gene expression inversely correlate with the level of bone marrow infiltration.

    No full text
    <p>(A) Correlation between bone marrow infiltration and the relative gene expression of <i>SHP-1</i>, <i>SHP-2</i> and <i>SOCS-1</i> in the sorted bone marrow plasma cells. Spearman rank correlation (r) and p values are indicated. (B) The degree of bone marrow infiltration is compared in patients exhibiting normal or decreased expression of <i>SHP-1</i>, <i>SHP-2</i> and <i>SOCS-1</i> genes. P values are indicated.</p
    corecore