Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 745939, 2021.
Article in English | MEDLINE | ID: mdl-34616407

ABSTRACT

Natural Killer (NK) cells play a key role in cancer immunosurveillance. However, NK cells from cancer patients display an altered phenotype and impaired effector functions. In addition, evidence of a regulatory role for NK cells is emerging in diverse models of viral infection, transplantation, and autoimmunity. Here, we analyzed clear cell renal cell carcinoma (ccRCC) datasets from The Cancer Genome Atlas (TCGA) and observed that a higher expression of NK cell signature genes is associated with reduced survival. Analysis of fresh tumor samples from ccRCC patients unraveled the presence of a high frequency of tumor-infiltrating PD-L1+ NK cells, suggesting that these NK cells might exhibit immunoregulatory functions. In vitro, PD-L1 expression was induced on NK cells from healthy donors (HD) upon direct tumor cell recognition through NKG2D and was further up-regulated by monocyte-derived IL-18. Moreover, in vitro generated PD-L1hi NK cells displayed an activated phenotype and enhanced effector functions compared to PD-L1- NK cells, but simultaneously, they directly inhibited CD8+ T cell proliferation in a PD-L1-dependent manner. Our results suggest that tumors might drive the development of PD-L1-expressing NK cells that acquire immunoregulatory functions in humans. Hence, rational manipulation of these regulatory cells emerges as a possibility that may lead to improved anti-tumor immunity in cancer patients.


Subject(s)
B7-H1 Antigen/biosynthesis , CD8-Positive T-Lymphocytes/cytology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Killer Cells, Natural/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Datasets as Topic , Disease-Free Survival , Gene Expression , Humans , Interferon-gamma/pharmacology , Interleukin-18/pharmacology , K562 Cells , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Monitoring, Immunologic , Monocytes/metabolism , Recombinant Proteins/pharmacology , Up-Regulation
2.
J Leukoc Biol ; 109(1): 185-194, 2021 01.
Article in English | MEDLINE | ID: mdl-33095941

ABSTRACT

After recognition, NK cells can kill susceptible target cells through perforin-dependent mechanisms or by inducing death receptor-mediated apoptosis, and they can also secrete cytokines that are pivotal for immunomodulation. Despite the critical role as effector cells against tumors and virus-infected cells, NK cells have been implicated in the regulation of T cell-mediated responses in different models of autoimmunity, transplantation, and viral infections. Here, we review the mechanisms described for NK cell-mediated inhibition of adaptive immune responses, with spotlight on the emerging evidence of their regulatory role that shapes antitumor immune responses.


Subject(s)
Adaptive Immunity/immunology , Cytotoxicity, Immunologic/immunology , Infections/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Humans
3.
Methods Mol Biol ; 2097: 125-136, 2020.
Article in English | MEDLINE | ID: mdl-31776924

ABSTRACT

Natural killer (NK) cells can kill virus-infected cells and tumor cells without prior sensitization and secrete numerous cytokines and chemokines that modulate the activity of different cells of the immune system. The recognition of target cells is mediated by germ line-encoded receptors, and the activity of NK cells can be further regulated by soluble factors such as cytokines and Toll-like receptor ligands. Thus, NK cells display an exciting potential as a powerful immunotherapeutic tool against malignant diseases, and different strategies are being tested aiming to overcome tumor-induced NK cell suppression and restore NK-cell mediated antitumor activity. This section describes different flow cytometry-based protocols to study NK cell effector functions, which can be used to evaluate the immunomodulatory ability of different therapeutic compounds.


Subject(s)
Immunomodulation/immunology , Killer Cells, Natural/immunology , Cell Degranulation , Cell Membrane/metabolism , Cell Separation , Humans , Killer Cells, Natural/physiology , Phenotype , Staining and Labeling
4.
Medicina (B Aires) ; 79(Spec 6/1): 564-569, 2019.
Article in English | MEDLINE | ID: mdl-31864227

ABSTRACT

In recent times, our understanding of the role of the immune system in different physiopathological situations has increased markedly. A new set of cells, generically known as innate lymphoid cells (ILC), has been discovered in the lymphoid compartment. Five ILC subsets can be recognized according to phenotypic and functional similarities with different subpopulations of T lymphocytes. Unlike T and B lymphocytes, ILC do not express antigen receptors nor undergo selection and clonal expansion upon activation. Instead, they respond rapidly to cytokines and danger signals in infected or inflamed tissues, producing cytokines that direct the immune response toward a type suitable for controlling the initial insult. In addition, ILC establish a crosstalk with other cells of the microenvironment that contributes to the maintenance and restoration of tissue homeostasis. Although many evidences on ILC were obtained from animal models, solid data confirm their existence in humans and their role in various inflammatory disorders. In this article, we address new knowledge on ILC, particularly on their role in the homeostasis of the immune system and in various inflammatory pathologies, in order to present new actors regulating immunity and immunopathology and affecting human health.


En tiempos recientes, nuestra comprensión del rol del sistema inmune en diferentes situaciones fisiopatológicas ha aumentado notablemente. En el compartimiento linfoide se ha descubierto un conjunto de células denominadas células linfoides innatas o innate lymphoid cells (ILC). Las ILC incluyen cinco grupos, clasificados según su similitud fenotípica y funcional con diferentes subpoblaciones de linfocitos T. A diferencia de los linfocitos T y B, las ILC no expresan receptores de antígeno ni sufren selección y expansión clonal cuando se activan. En cambio, responden rápidamente frente a citoquinas y señales de peligro en tejidos infectados o inflamados produciendo citoquinas que dirigen la respuesta inmune hacia un tipo adecuado para controlar la noxa original. Además, las ILC establecen un diálogo cruzado con otras células del microambiente que contribuye al mantenimiento y la restauración de la homeostasis tisular. Si bien muchas evidencias acerca de las ILC fueron obtenidas en modelos animales, existen datos sólidos que confirman su existencia en seres humanos y su papel en diversos trastornos inflamatorios. En este artículo, abordamos los nuevos conocimientos acerca de las ILC, y su rol en la homeostasis del sistema inmune y en diversas patologías inflamatorias, con el fin de presentar nuevos actores que regulan la inmunidad y la inmunopatología, lo que repercute en la salud humana.


Subject(s)
Homeostasis/immunology , Immunity, Innate/immunology , Inflammation/immunology , Intestinal Diseases/immunology , Lung Diseases/immunology , Lymphocytes/immunology , Skin Diseases/immunology , Homeostasis/physiology , Humans , Inflammation/physiopathology , Intestinal Diseases/physiopathology , Lung Diseases/physiopathology , Skin Diseases/physiopathology
5.
J Immunol ; 200(3): 1008-1015, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29282306

ABSTRACT

NK cells play important roles during immunosurveillance against tumors and viruses as they trigger cytotoxicity against susceptible cells and secrete proinflammatory cytokines such as IFN-γ. In addition, upon activation, macrophages can become proinflammatory (M1) or anti-inflammatory (M2) cells. Although the consequences of the cross-talk between M1 and NK cells are known, the outcome of the cross-talk between M2 and NK cells remains ill-defined. Therefore, in the current work, we investigated the outcome and the underlying mechanisms of the interaction between resting or stimulated human NK cells with M1 or M2. We observed a lower percentage of activated NK cells that produced less IFN-γ upon coculture with M2. Also, CD56dim NK cells cocultured with M2 displayed lower degranulation and cytotoxic activity than NK cells cocultured with M1. Soluble TGF-ß and M2-driven upregulation of CD85j (ILT-2) on NK cells accounted for the diminished IFN-γ production by CD56bright NK cells, whereas M2-driven upregulation of CD85j on NK cells accounted for the generation of hyporesponsive CD56dim NK cells with limited degranulation and cytotoxic capacity. Accordingly, M2 expressed higher amounts of HLA-G, the main ligand for CD85j, than M1. Hyporesponsiveness to degranulation in NK cells was not restored at least for several hours upon removal of M2. Therefore, alternatively activated macrophages restrain NK cell activation and effector functions through different mechanisms, leading to NK cells that display diminished IFN-γ production and at least a transiently impaired degranulation ability. These results unravel an inhibitory circuit of possible relevance in pathological situations.


Subject(s)
Cell Communication/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Transforming Growth Factor beta/metabolism , Antigens, CD/immunology , CD56 Antigen/metabolism , Cells, Cultured , Coculture Techniques , HLA-G Antigens/metabolism , Humans , Interferon-gamma/immunology , Leukocyte Immunoglobulin-like Receptor B1/immunology , Macrophages/immunology , Transforming Growth Factor beta/immunology
6.
Front Immunol ; 8: 1959, 2017.
Article in English | MEDLINE | ID: mdl-29403472

ABSTRACT

Interleukin (IL)-23 is a member of the IL-12 family of cytokines that, as the other members of this family, is secreted by monocytes, macrophages, and dendritic cells (DC) upon recognition of bacterial, viral, and fungal components. IL-23 is critical during immunity against acute infections, and it is also involved in the development of autoimmune diseases. Although immunoregulatory effects of IL-23 on mouse natural killer (NK) cells have been described, the effect of IL-23 on human NK cells remains ill-defined. In this study, we observed that monocytes stimulated with LPS secreted IL-23 and that blockade of this cytokine during monocyte and NK cell coculture led to a diminished production of IFN-γ by NK cells. Accordingly, rIL-23-induced NK cell activation and stimulated IFN-γ production by CD56bright NK cells. This effect involved MEK1/MEK2, JNK, PI3K, mammalian target of rapamycin, and NF-κB, but not STAT-1, STAT-3, nor p38 MAPK pathways. Moreover, while NK cell-mediated cytotoxicity remained unaltered, antibody-dependent cellular cytotoxicity (ADCC) was enhanced after IL-23 stimulation. In addition, IL-23 displayed a synergistic effect with IL-18 for IFN-γ production by both CD56bright and CD56dim NK cells, and this effect was due to a priming effect of IL-23 for IL-18 responsiveness. Furthermore, NK cells pre-stimulated with IL-18 promoted an increase in CD86 expression and IL-12 secretion by DC treated with LPS, and IL-23 potentiated these effects. Moreover, IL-23-driven enhancement of NK cell "helper" function was dependent on NK cell-derived IFN-γ. Therefore, our results suggest that IL-23 may trigger NK cell-mediated "helper" effects on adaptive immunity, shaping T cell responses during different pathological situations through the regulation of DC maturation.

7.
J Immunol ; 197(3): 953-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27342842

ABSTRACT

Despite the classical function of NK cells in the elimination of tumor and of virus-infected cells, evidence for a regulatory role for NK cells has been emerging in different models of autoimmunity, transplantation, and viral infections. However, this role has not been fully explored in the context of a growing tumor. In this article, we show that NK cells can limit spontaneous cross-priming of tumor Ag-specific CD8(+) T cells, leading to reduced memory responses. After challenge with MC57 cells transduced to express the model Ag SIY (MC57.SIY), NK cell-depleted mice exhibited a significantly higher frequency of SIY-specific CD8(+) T cells, with enhanced IFN-γ production and cytotoxic capability. Depletion of NK cells resulted in a CD8(+) T cell population skewed toward an effector memory T phenotype that was associated with enhanced recall responses and delayed tumor growth after a secondary tumor challenge with B16.SIY cells. Dendritic cells (DCs) from NK cell-depleted tumor-bearing mice exhibited a more mature phenotype. Interestingly, tumor-infiltrating and tumor-draining lymph node NK cells displayed an upregulated expression of the inhibitory molecule programmed death ligand 1 that, through interaction with programmed death-1 expressed on DCs, limited DC activation, explaining their reduced ability to induce tumor-specific CD8(+) T cell priming. Our results suggest that NK cells can, in certain contexts, have an inhibitory effect on antitumor immunity, a finding with implications for immunotherapy in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms, Experimental/immunology , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology
8.
J Immunol ; 195(5): 2141-8, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26232426

ABSTRACT

Cross-talk between mature dendritic cells (mDC) and NK cells through the cell surface receptors NKp30 and DNAM-1 leads to their reciprocal activation. However, the impact of regulatory dendritic cells (regDC) on NK cell function remains unknown. As regDC constrain the immune response in different physiological and pathological conditions, the aim of this work was to investigate the functional outcome of the interaction between regDC and NK cells and the associated underlying mechanisms. RegDC generated from monocyte-derived DC treated either with LPS and dexamethasone, vitamin D3, or vitamin D3 and dexamethasone instructed NK cells to secrete lower amounts of IFN-γ than NK cells exposed to mDC. Although regDC triggered upregulation of the activation markers CD69 and CD25 on NK cells, they did not induce upregulation of CD56 as mDC, and silenced IFN-γ secretion through mechanisms involving insufficient secretion of IL-18, but not IL-12 or IL-15 and/or induction of NK cell apoptosis. Blocking experiments demonstrated that regDC curb IFN-γ secretion by NK cells through a dominant suppressive mechanism involving IL-10, NK cell inhibitory receptors, and, unexpectedly, engagement of the activating receptor NKp46. Our findings unveil a previously unrecognized cross-talk through which regDC shape NK cell function toward an alternative activated phenotype unable to secrete IFN-γ, highlighting the plasticity of NK cells in response to tolerogenic stimuli. In addition, our findings contribute to identify a novel inhibitory role for NKp46 in the control of NK cell function, and have broad implications in the resolution of inflammatory responses and evasion of antitumor responses.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/immunology , Dendritic Cells/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Cell Communication/drug effects , Cell Communication/immunology , Cells, Cultured , Cholecalciferol/immunology , Cholecalciferol/pharmacology , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dexamethasone/immunology , Dexamethasone/pharmacology , Flow Cytometry , Glucocorticoids/immunology , Glucocorticoids/pharmacology , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/biosynthesis , Interleukin-10/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/immunology , Vitamins/immunology , Vitamins/pharmacology
9.
Eur J Immunol ; 45(1): 192-202, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25308526

ABSTRACT

IL-27, a member of the IL-12 family of cytokines, is produced by APCs, and displays pro- and anti-inflammatory effects. How IL-27 affects human NK cells still remains unknown. In this study, we observed that mature DCs secreted IL-27 and that blockade of IL-27R (CD130) reduced the amount of IFN-γ produced by NK cells during their coculture, showing the importance of IL-27 during DC-NK-cell crosstalk. Accordingly, human rIL-27 stimulated IFN-γ secretion by NK cells in a STAT1-dependent manner, induced upregulation of CD25 and CD69 on NK cells, and displayed a synergistic effect with IL-18. Preincubation experiments demonstrated that IL-27 primed NK cells for IL-18-induced IFN-γ secretion, which was associated with an IL-27-driven upregulation of T-bet expression. Also, IL-27 triggered NKp46-dependent NK-cell-mediated cytotoxicity against Raji, T-47D, and HCT116 cells, and IL-18 enhanced this cytotoxic response. Such NK-cell-mediated cytotoxicity involved upregulation of perforin, granule exocytosis, and TRAIL-mediated cytotoxicity but not Fas-FasL interaction. Moreover, IL-27 also potentiated Ab-dependent cell-mediated cytotoxicity against mAb-coated target cells. Taken together, IL-27 stimulates NK-cell effector functions, which might be relevant in different physiological and pathological situations.


Subject(s)
Dendritic Cells/immunology , Interleukin-18/pharmacology , Interleukins/pharmacology , Killer Cells, Natural/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Cell Proliferation/drug effects , Cell Survival/immunology , Coculture Techniques , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Regulation , HCT116 Cells , Humans , Interleukin-18/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Primary Cell Culture , Recombinant Proteins/pharmacology , Signal Transduction
10.
Immunity ; 41(5): 830-42, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517615

ABSTRACT

Spontaneous T cell responses against tumors occur frequently and have prognostic value in patients. The mechanism of innate immune sensing of immunogenic tumors leading to adaptive T cell responses remains undefined, although type I interferons (IFNs) are implicated in this process. We found that spontaneous CD8(+) T cell priming against tumors was defective in mice lacking stimulator of interferon genes complex (STING), but not other innate signaling pathways, suggesting involvement of a cytosolic DNA sensing pathway. In vitro, IFN-? production and dendritic cell activation were triggered by tumor-cell-derived DNA, via cyclic-GMP-AMP synthase (cGAS), STING, and interferon regulatory factor 3 (IRF3). In the tumor microenvironment in vivo, tumor cell DNA was detected within host antigen-presenting cells, which correlated with STING pathway activation and IFN-? production. Our results demonstrate that a major mechanism for innate immune sensing of cancer occurs via the host STING pathway, with major implications for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Membrane Proteins/immunology , Adaptive Immunity , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/immunology , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon-beta/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Nucleotidyltransferases , Receptors, Antigen, T-Cell/immunology , Receptors, Purinergic P2X7/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Tumor Microenvironment/immunology
11.
J Immunol ; 193(8): 4254-60, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25217157

ABSTRACT

Endogenous type I IFN production after innate immune recognition of tumor cells is critical for generating natural adaptive immune responses against tumors in vivo. We recently have reported that targeting low doses of IFN-ß to the tumor microenvironment using tumor-specific mAbs can facilitate antitumor immunity, which could be augmented further with PD-L1/PD-1 blockade. However, sustained high doses of type I IFNs in the tumor microenvironment, which are potently therapeutic alone, may function through distinct mechanisms. In the current report, we demonstrate that high-dose intratumoral type I IFNs indeed exerted a profound therapeutic effect in the murine B16 model, which unexpectedly did not increase T cell responses. Moreover, bone marrow chimeras revealed a role for type I IFN signaling on nonhematopoietic cells, and most of the therapeutic effect was retained in mice deficient in T, B, and NK cells. Rather, the tumor vasculature was ablated with high-dose intratumoral IFN-ß, and conditional deletion of IFN-α/ßR in Tie2-positive vascular endothelial cells eliminated most of the antitumor activity. Therefore, the major component of the antitumor activity of sustained high doses of type I IFNs occurs through a direct antiangiogenic effect. Our data help resolve conditions under which distinct antitumor mechanisms of type I IFNs are operational in vivo.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Interferon-beta/administration & dosage , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Receptor, Interferon alpha-beta/genetics , Animals , Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Killer Cells, Natural/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects
12.
Trends Immunol ; 34(2): 67-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23122052

ABSTRACT

Unexpectedly, many cancers appear to induce a spontaneous adaptive T cell response. The presence of a T cell infiltrate has been linked to favorable clinical outcome in multiple cancer types. However, the innate immune pathways that bridge to an adaptive immune response under sterile conditions are poorly understood. Recent data have indicated that tumors can induce type I interferon (IFN) production by host antigen-presenting cells (APCs), which is required for a spontaneous T cell response in vivo. The innate immune sensing pathways that trigger type I IFN production are being elucidated. Host type I IFNs are also required for optimal therapeutic efficacy with radiation. This recently uncovered role for host type I IFNs for antitumor immunity has important fundamental and clinical implications.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Neoplasms/immunology , Animals , Humans , Mice , Signal Transduction
13.
Cancer Immunol Immunother ; 61(8): 1343-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22722449

ABSTRACT

A subset of patients with a variety of cancers shows evidence of a natural adaptive immune response against their tumor, as evidenced by spontaneous T-cell infiltration, circulating anti-tumor T cells, or antibody responses. Evidence has indicated that such natural immune responses have positive prognostic import in early stage disease and may be predictive of clinical response to immunotherapeutics in advanced disease. However, these observations raise a new critical fundamental question-what innate immune signals might be generated in the context of non-pathogen-induced cancers that drive productive antigen presentation toward induction of an adaptive immune response? Gene expression profiling in melanoma revealed that tumors having high expression of T-cell markers also show evidence of a type I IFN transcriptional signature. Mechanistic experiments in mice have revealed that a spontaneous CD8(+) T-cell response against transplantable tumors depends on host type I IFN signaling, through a mechanism dependent upon CD8α(+) dendritic cells (DCs). The requirement for type I IFN production by host DCs has suggested a subset of innate immune sensing receptors and signaling pathways that might be involved with initiating this process. Elucidating further these innate immune mechanisms should provide new insights into cancer immunotherapy.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Neoplasms/immunology , Signal Transduction/immunology , Animals , Congresses as Topic , Dendritic Cells/immunology , Humans , Mice
14.
J Leukoc Biol ; 91(2): 321-31, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22124136

ABSTRACT

HDACi are being used as a novel, therapeutic approach for leukemias and other hematological malignancies. However, their effect on immune cells remains ill-defined, as HDACi may impair immune surveillance. In this work, we demonstrate that TSA, VPA, and NaB inhibited IFN-γ production by CD56(dim) and CD56(bright) NK cells and NK cell-mediated cytotoxicity against K562 target cells. HDACi promoted minor NK cell apoptosis but inhibited nuclear mobilization of NF-κB p50, which was accompanied by a robust down-regulation of NKG2D and NKp46 on resting NK cells and of NKG2D, NKp44, NKp46, and CD25 on cytokine-activated NK cells. Decreased CD25 expression promoted a weakened IFN-γ secretion upon restimulation of NK cells with IL-2, whereas reduced expression of NKG2D and NKp46 was accompanied by an impaired NKG2D- and NKp46-dependent cytotoxicity. Moreover, NK cells from normal mice treated in vivo with TSA displayed a diminished expression of NK1.1, NKG2D, and NKp46 and secreted reduced amounts of IFN-γ upon ex vivo stimulation with cytokines. Thus, our preclinical results indicate that HDACi exert deleterious effects on NK cell function, which may weaken immune surveillance and facilitate relapse of the malignant disease in HDACi-treated patients.


Subject(s)
Antineoplastic Agents/pharmacology , Butyrates/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Immunologic Surveillance/drug effects , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cells, Cultured/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , Histone Deacetylase Inhibitors/adverse effects , Humans , Interferon-gamma/metabolism , Interleukins/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/genetics , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/genetics , Vorinostat
15.
J Exp Med ; 208(10): 2005-16, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21930765

ABSTRACT

Despite lack of tumor control in many models, spontaneous T cell priming occurs frequently in response to a growing tumor. However, the innate immune mechanisms that promote natural antitumor T cell responses are undefined. In human metastatic melanoma, there was a correlation between a type I interferon (IFN) transcriptional profile and T cell markers in metastatic tumor tissue. In mice, IFN-ß was produced by CD11c(+) cells after tumor implantation, and tumor-induced T cell priming was defective in mice lacking IFN-α/ßR or Stat1. IFN signaling was required in the hematopoietic compartment at the level of host antigen-presenting cells, and selectively for intratumoral accumulation of CD8α(+) dendritic cells, which were demonstrated to be essential using Batf3(-/-) mice. Thus, host type I IFNs are critical for the innate immune recognition of a growing tumor through signaling on CD8α(+) DCs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Lymphocyte Subsets/immunology , Melanoma/immunology , Signal Transduction/immunology , Animals , Cell Line, Tumor , Humans , Immunity, Innate/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Metastasis/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology
16.
Medicina (B Aires) ; 71(4): 357-60, 2011.
Article in English | MEDLINE | ID: mdl-21893449

ABSTRACT

The MHC class I chain-related protein A (MICA) is an inducible molecule almost not expressed by normal cells but strongly up-regulated in tumor cells. MICA-expressing cells are recognized by natural killer (NK) cells, CD8+ abTCR and gdTCR T lymphocytes through the NKG2D receptor. Engagement of NKG2D by MICA triggers IFN-g secretion and cytotoxicity against malignant cells. Although most solid tumors express MICA and this molecule is a target during immune surveillance against tumors, it has been observed that high grade tumors from different histotypes express low amounts of cell surface MICA due to a metalloprotease-induced shedding. Also, melanomas develop after a complex process of neotransformation of normal melanocytes. However, the expression of MICA in premalignant stages (primary human quiescent melanocytic nevi) remains unknown. Here, we assessed expression of MICA by flow cytometry using cell suspensions from 15 primary nevi isolated from 11 patients. When collected material was abundant, cell lysates were prepared and MICA expression was also analyzed by Western blot. We observed that MICA was undetectable in the 15 primary nevi (intradermic, junction, mixed, lentigo and congenital samples) as well as in normal skin, benign lesions (seborrheic keratosis), premalignant lesions (actinic keratosis) and benign basocellular cancer. Conversely, a primary recently diagnosed melanoma showed intense cell surface MICA. We conclude that the onset of MICA expression is a tightly regulated process that occurs after melanocytes trespass the stage of malignant transformation. Thus, analysis of MICA expression in tissue sections of skin samples may constitute a useful marker to differentiate between benign and malignant nevi.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Nevus/metabolism , Precancerous Conditions/metabolism , Skin Neoplasms/metabolism , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Nevus/immunology , Nevus/pathology , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology
17.
Mol Immunol ; 48(15-16): 1940-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21689853

ABSTRACT

The inflammatory response is a self-limiting process which involves the sequential activation of signaling pathways leading to the production of both pro- and anti-inflammatory mediators. Galectin-1 (Gal-1), an endogenous lectin found in peripheral lymphoid organs and inflammatory sites, elicits a broad spectrum of biological functions predominantly by acting as a potent anti-inflammatory factor and as a suppressive agent for T-cell responses. However, the molecular pathways underlying Gal-1 expression and function remain poorly understood. Here we identified a regulatory loop linking Gal-1 expression and function to NF-κB activation. NF-κB-activating stimuli increased Gal-1 expression on T cells, an effect which could be selectively prevented by inhibitors of NF-κB signaling. Accordingly, transient transfection of the p65 subunit of NF-κB was sufficient to induce high Gal-1 expression. Using in silico studies and chromatin immunoprecipitation analysis we have identified a functional NF-κB binding site within the first intron of the LGALS1 gene. In addition, our results show that exogenous Gal-1 can attenuate NF-κB activation, as shown by inhibition of IκB-α degradation induced by pro-inflammatory stimuli, higher cytoplasmic retention of p65, lower NF-κB DNA binding activity and impaired transcriptional activation of target genes. The present study suggest a novel regulatory loop by which NF-κB induces expression of Gal-1, which in turn may lead to negative control of NF-κB signaling.


Subject(s)
Galectin 1/biosynthesis , Gene Expression Regulation/immunology , NF-kappa B/metabolism , Signal Transduction/immunology , Binding Sites , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Feedback, Physiological/physiology , Galectin 1/genetics , Galectin 1/immunology , Gene Expression , Humans , Microscopy, Confocal , NF-kappa B/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transfection
18.
EMBO Rep ; 10(8): 908-15, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19498463

ABSTRACT

Natural killer (NK) cells trigger cytotoxicity and interferon (IFN)-gamma secretion on engagement of the natural-killer group (NKG)2D receptor or members of the natural cytotoxicity receptor (NCR) family, such as NKp46, by ligands expressed on tumour cells. However, it remains unknown whether T cells can regulate NK cell-mediated anti-tumour responses. Here, we investigated the early events occurring during T cell-tumour cell interactions, and their impact on NK cell functions. We observed that on co-culture with some melanomas, activated CD4(+) T cells promoted degranulation, and NKG2D- and NKp46-dependent IFN-gamma secretion by NK cells, probably owing to the capture of NKG2D and NKp46 ligands from the tumour-cell surface (trogocytosis). This effect was observed in CD4(+), CD8(+) and resting T cells, which showed substantial amounts of cell surface major histocompatibility complex class I chain-related protein A on co-culture with tumour cells. Our findings identify a new, so far, unrecognized mechanism by which effector T cells support NK cell function through the capture of specific tumour ligands with profound implications at the crossroad of innate and adaptive immunity.


Subject(s)
Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Neoplasms/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Microscopy, Confocal , Mitogens/pharmacology , Phytohemagglutinins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
19.
J Immunol ; 180(7): 4606-14, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354183

ABSTRACT

Most tumors grow in immunocompetent hosts despite expressing NKG2D ligands (NKG2DLs) such as the MHC class I chain-related genes A and B (MICA/B). However, their participation in tumor cell evasion is still not completely understood. Here we demonstrate that several human melanomas (cell lines and freshly isolated metastases) do not express MICA on the cell surface but have intracellular deposits of this NKG2DL. Susceptibility to NK cell-mediated cytotoxicity correlated with the ratio of NKG2DLs to HLA class I molecules but not with the amounts of MICA on the cell surface of tumor cells. Transfection-mediated overexpression of MICA restored cell surface expression and resulted in an increased in vitro cytotoxicity and IFN-gamma secretion by human NK cells. In xenografted nude mice, these melanomas exhibited a delayed growth and extensive in vivo apoptosis. Retardation of tumor growth was due to NK cell-mediated antitumor activity against MICA-transfected tumors, given that this effect was not observed in NK cell-depleted mice. Also, mouse NK cells killed MICA-overexpressing melanomas in vitro. A mechanistic analysis revealed the retention of MICA in the endoplasmic reticulum, an effect that was associated with accumulation of endoH-sensitive (immature) forms of MICA, retrograde transport to the cytoplasm, and degradation by the proteasome. Our study identifies a novel strategy developed by melanoma cells to evade NK cell-mediated immune surveillance based on the intracellular sequestration of immature forms of MICA in the endoplasmic reticulum. Furthermore, this tumor immune escape strategy can be overcome by gene therapy approaches aimed at overexpressing MICA on tumor cells.


Subject(s)
Cytotoxicity, Immunologic/immunology , Histocompatibility Antigens Class I/immunology , Intercellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/immunology , Melanoma/immunology , Cell Line, Tumor , Cell Proliferation , GPI-Linked Proteins , Humans , Melanoma/pathology , Melanoma/ultrastructure , Microscopy, Immunoelectron , Sensitivity and Specificity
20.
J Immunol ; 179(6): 3472-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17804388

ABSTRACT

NK cells express different TLRs, such as TLR3, TLR7, and TLR9, but little is known about their role in NK cell stimulation. In this study, we used specific agonists (poly(I:C), loxoribine, and synthetic oligonucleotides containing unmethylated CpG sequences to stimulate human NK cells without or with suboptimal doses of IL-12, IL-15, or IFN-alpha, and investigated the secretion of IFN-gamma, cytotoxicity, and expression of the activating receptor NKG2D. Poly(I:C) and loxoribine, in conjunction with IL-12, but not IL-15, triggered secretion of IFN-gamma. Inhibition of IFN-gamma secretion by chloroquine suggested that internalization of the TLR agonists was necessary. Also, secretion of IFN-gamma was dependent on MEK1/ERK, p38 MAPK, p70(S6) kinase, and NF-kappaB, but not on calcineurin. IFN-alpha induced a similar effect, but promoted lesser IFN-gamma secretion. However, cytotoxicity (51Cr release assays) against MHC class I-chain related A (MICA)- and MICA+ tumor targets remained unchanged, as well as the expression of the NKG2D receptor. Excitingly, IFN-gamma secretion was significantly increased when NK cells were stimulated with poly(I:C) or loxoribine and IL-12, and NKG2D engagement was induced by coculture with MICA+ tumor cells in a PI3K-dependent manner. We conclude that resting NK cells secrete high levels of IFN-gamma in response to agonists of TLR3 or TLR7 and IL-12, and this effect can be further enhanced by costimulation through NKG2D. Hence, integration of the signaling cascades that involve TLR3, TLR7, IL-12, and NKG2D emerges as a critical step to promote IFN-gamma-dependent NK cell-mediated effector functions, which could be a strategy to promote Th1-biased immune responses in pathological situations such as cancer.


Subject(s)
Cytotoxicity, Immunologic , Interferon-gamma/metabolism , Interleukin-12/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cells, Cultured , Clone Cells , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Dose-Response Relationship, Immunologic , Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Interferon-alpha/physiology , Interleukin-12/pharmacology , Killer Cells, Natural/metabolism , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/physiology , Receptors, Natural Killer Cell , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...