Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Nat Immunol ; 23(2): 262-274, 2022 02.
Article in English | MEDLINE | ID: mdl-35102345

ABSTRACT

Tumors poorly infiltrated by T cells are more resistant to immunogenic chemotherapies and checkpoint inhibition than highly infiltrated tumors. Using murine models, we found that CCR6+ type 3 innate lymphoid cells (ILC3s) can trigger an increase in the number of T cells infiltrating a tumor. Shortly after administration of cisplatin chemotherapy, production of the chemokine CCL20 and proinflammatory cytokine IL-1ß at the tumor site led to the recruitment and activation of ILC3s. Within the tumor, ILC3 production of the chemokine CXCL10 was responsible for the recruitment of CD4+ and CD8+ T lymphocytes to the tumor. ILC3-dependent infiltration of T cells was essential for antitumor immune responses and increased the efficacy of checkpoint inhibition. Thus, we reveal an essential role of CCL20 and IL-1ß, which promote ILC3-dependent antitumor immunity and enhance tumor sensitivity to immunotherapy.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/immunology , Female , Humans , Immune Checkpoint Inhibitors/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31031005

ABSTRACT

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Subject(s)
Cellular Microenvironment/immunology , Dendritic Cells/immunology , Immunity, Innate , Mitochondria/immunology , Reactive Oxygen Species/immunology , Unfolded Protein Response/immunology , Animals , Cellular Microenvironment/genetics , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Dendritic Cells/pathology , Hexokinase/genetics , Hexokinase/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
3.
Nat Immunol ; 22(3): 322-335, 2021 03.
Article in English | MEDLINE | ID: mdl-33531712

ABSTRACT

Immune system dysfunction is paramount in coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-associated invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in cohorts totaling 208 patients with various stages of disease. MAIT cell frequency is strongly reduced in blood. They display a strong activated and cytotoxic phenotype that is more pronounced in lungs. Blood MAIT cell alterations positively correlate with the activation of other innate cells, proinflammatory cytokines, notably interleukin (IL)-18, and with the severity and mortality of severe acute respiratory syndrome coronavirus 2 infection. We also identified a monocyte/macrophage interferon (IFN)-α-IL-18 cytokine shift and the ability of infected macrophages to induce the cytotoxicity of MAIT cells in an MR1-dependent manner. Together, our results suggest that altered MAIT cell functions due to IFN-α-IL-18 imbalance contribute to disease severity, and their therapeutic manipulation may prevent deleterious inflammation in COVID-19 aggravation.


Subject(s)
COVID-19/immunology , Interferon-alpha/immunology , Interleukin-18/immunology , Macrophages/immunology , Monocytes/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , Aged , Aged, 80 and over , Animals , Bronchoalveolar Lavage , Case-Control Studies , Chlorocebus aethiops , Cohort Studies , Female , France , Humans , Immunophenotyping , Interleukin-10/immunology , Interleukin-15/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Male , Middle Aged , RNA-Seq , SARS-CoV-2 , Severity of Illness Index , Single-Cell Analysis , Vero Cells , Young Adult
4.
Immunity ; 55(12): 2211-2216, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516812

ABSTRACT

CD1 molecules and the MHC-related protein 1 (MR1) present lipid and small molecule antigens, respectively, for T cell surveillance. The biology of these molecules, the antigens they present, and the T cells that respond to them were recently discussed during the 12th International CD1-MR1 Meeting held in Gothenburg, Sweden.


Subject(s)
Antigens, CD1 , Histocompatibility Antigens Class I , Minor Histocompatibility Antigens , Antigens, CD1/metabolism , T-Lymphocytes , Antigens , Antigen Presentation
6.
EMBO J ; 41(12): e108306, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35506364

ABSTRACT

Influenza virus infection causes considerable morbidity and mortality, but current therapies have limited efficacy. We hypothesized that investigating the metabolic signaling during infection may help to design innovative antiviral approaches. Using bronchoalveolar lavages of infected mice, we here demonstrate that influenza virus induces a major reprogramming of lung metabolism. We focused on mitochondria-derived succinate that accumulated both in the respiratory fluids of virus-challenged mice and of patients with influenza pneumonia. Notably, succinate displays a potent antiviral activity in vitro as it inhibits the multiplication of influenza A/H1N1 and A/H3N2 strains and strongly decreases virus-triggered metabolic perturbations and inflammatory responses. Moreover, mice receiving succinate intranasally showed reduced viral loads in lungs and increased survival compared to control animals. The antiviral mechanism involves a succinate-dependent posttranslational modification, that is, succinylation, of the viral nucleoprotein at the highly conserved K87 residue. Succinylation of viral nucleoprotein altered its electrostatic interactions with viral RNA and further impaired the trafficking of viral ribonucleoprotein complexes. The finding that succinate efficiently disrupts the influenza replication cycle opens up new avenues for improved treatment of influenza pneumonia.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Orthomyxoviridae Infections , Pneumonia , Animals , Antiviral Agents/pharmacology , Humans , Influenza A Virus, H3N2 Subtype/metabolism , Mice , Nucleocapsid Proteins , Nucleoproteins/metabolism , Succinic Acid/metabolism , Succinic Acid/pharmacology , Succinic Acid/therapeutic use , Virus Replication
7.
Trends Immunol ; 43(7): 503-512, 2022 07.
Article in English | MEDLINE | ID: mdl-35654639

ABSTRACT

Invariant natural killer T (iNKT) cells are increasingly regarded as disease biomarkers and immunotherapeutic targets. However, a greater understanding of their biology is necessary to effectively target these cells in the clinic. The discovery of iNKT1/2/17 cell effector subsets was a milestone in our understanding of iNKT cell development and function. Recent transcriptomic studies have uncovered an even greater heterogeneity and challenge our understanding of iNKT cell ontogeny and effector differentiation. We propose a refined model whereby iNKT cells differentiate through a dynamic and continuous instructive process that requires the accumulation and integration of various signals within the thymus or peripheral tissues. Within this framework, we question the existence of true iNKT2 cells and discuss the parallels between mouse and human iNKT cells.


Subject(s)
Natural Killer T-Cells , Animals , Cell Differentiation , Humans , Mice
8.
PLoS Pathog ; 18(7): e1010305, 2022 07.
Article in English | MEDLINE | ID: mdl-35849616

ABSTRACT

Multiple regulated neutrophil cell death programs contribute to host defense against infections. However, despite expressing all necessary inflammasome components, neutrophils are thought to be generally defective in Caspase-1-dependent pyroptosis. By screening different bacterial species, we found that several Pseudomonas aeruginosa (P. aeruginosa) strains trigger Caspase-1-dependent pyroptosis in human and murine neutrophils. Notably, deletion of Exotoxins U or S in P. aeruginosa enhanced neutrophil death to Caspase-1-dependent pyroptosis, suggesting that these exotoxins interfere with this pathway. Mechanistically, P. aeruginosa Flagellin activates the NLRC4 inflammasome, which supports Caspase-1-driven interleukin (IL)-1ß secretion and Gasdermin D (GSDMD)-dependent neutrophil pyroptosis. Furthermore, P. aeruginosa-induced GSDMD activation triggers Calcium-dependent and Peptidyl Arginine Deaminase-4-driven histone citrullination and translocation of neutrophil DNA into the cell cytosol without inducing extracellular Neutrophil Extracellular Traps. Finally, we show that neutrophil Caspase-1 contributes to IL-1ß production and susceptibility to pyroptosis-inducing P. aeruginosa strains in vivo. Overall, we demonstrate that neutrophils are not universally resistant for Caspase-1-dependent pyroptosis.


Subject(s)
Inflammasomes , Pyroptosis , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 1/metabolism , Exotoxins/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/microbiology , Pseudomonas aeruginosa/metabolism
9.
PLoS Biol ; 17(3): e3000169, 2019 03.
Article in English | MEDLINE | ID: mdl-30822302

ABSTRACT

CD1d-restricted invariant natural killer T (iNKT) cells represent a heterogeneous population of lipid-reactive T cells that are involved in many immune responses, mediated through T-cell receptor (TCR)-dependent and/or independent activation. Although numerous microbial lipid antigens (Ags) have been identified, several lines of evidence have suggested the existence of relevant Ags of endogenous origin. However, the identification of their precise nature as well as the molecular mechanisms involved in their generation are still highly controversial and ill defined. Here, we identified two mammalian gangliosides-namely monosialoganglioside GM3 and disialoganglioside GD3-as endogenous activators for mouse iNKT cells. These glycosphingolipids are found in Toll-like receptor-stimulated dendritic cells (DC) as several species varying in their N-acyl fatty chain composition. Interestingly, their ability to activate iNKT cells is highly dependent on the ceramide backbone structure. Thus, both synthetic GM3 and GD3 comprising a d18:1-C24:1 ceramide backbone were able to activate iNKT cells in a CD1d-dependent manner. GM3 and GD3 are not directly recognized by the iNKT TCR and required the Ag presenting cell intracellular machinery to reveal their antigenicity. We propose a new concept in which iNKT cells can rapidly respond to pre-existing self-molecules after stress-induced structural changes in CD1d-expressing cells. Moreover, these gangliosides conferred partial protection in the context of bacterial infection. Thus, this report identified new biologically relevant lipid self-Ags for iNKT cells.


Subject(s)
Ceramides/metabolism , Gangliosides/metabolism , Natural Killer T-Cells/metabolism , Toll-Like Receptor 9/metabolism , Animals , Antigens, CD1d/metabolism , Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , G(M3) Ganglioside/metabolism , Glycosphingolipids/metabolism , Male , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
10.
PLoS Pathog ; 14(10): e1007360, 2018 10.
Article in English | MEDLINE | ID: mdl-30372491

ABSTRACT

Secondary bacterial infections contribute to the excess morbidity and mortality of influenza A virus (IAV) infection. Disruption of lung integrity and impaired antibacterial immunity during IAV infection participate in colonization and dissemination of the bacteria out of the lungs. One key feature of IAV infection is the profound alteration of lung myeloid cells, characterized by the recruitment of deleterious inflammatory monocytes. We herein report that IAV infection causes a transient decrease of lung conventional dendritic cells (cDCs) (both cDC1 and cDC2) peaking at day 7 post-infection. While triggering emergency monopoiesis, IAV transiently altered the differentiation of cDCs in the bone marrow, the cDC1-biaised pre-DCs being particularly affected. The impaired cDC differentiation during IAV infection was independent of type I interferons (IFNs), IFN-γ, TNFα and IL-6 and was not due to an intrinsic dysfunction of cDC precursors. The alteration of cDC differentiation was associated with a drop of local and systemic production of Fms-like tyrosine kinase 3 ligand (Flt3-L), a critical cDC differentiation factor. Overexpression of Flt3-L during IAV infection boosted the cDC progenitors' production in the BM, replenished cDCs in the lungs, decreased inflammatory monocytes' infiltration and lowered lung damages. This was associated with partial protection against secondary pneumococcal infection, as reflected by reduced bacterial dissemination and prolonged survival. These findings highlight the impact of distal viral infection on cDC genesis in the BM and suggest that Flt3-L may have potential applications in the control of secondary infections.


Subject(s)
Dendritic Cells/immunology , Influenza A virus/immunology , Lung/immunology , Membrane Proteins/immunology , Orthomyxoviridae Infections/virology , Pneumococcal Infections/immunology , Superinfection/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/microbiology , Dendritic Cells/virology , Lung/microbiology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Pneumococcal Infections/microbiology , Pneumococcal Infections/virology , Receptor, Interferon alpha-beta/physiology , Streptococcus pneumoniae/immunology
12.
J Immunol ; 197(8): 3225-3232, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27605012

ABSTRACT

Exogenous activation of invariant NKT (iNKT) cells by the superagonist α-galactosylceramide (α-GalCer) can protect against cancer, autoimmune diseases, and infections. In the current study, we investigated the effect of α-GalCer against Bacillus anthracis infection, the agent of anthrax. Using an experimental model of s.c. B. anthracis infection (an encapsulated nontoxigenic strain), we show that concomitant administration of α-GalCer delayed B. anthracis systemic dissemination and prolonged mouse survival. Depletion of subcapsular sinus CD169-positive macrophages by clodronate-containing liposome was associated with a lack of iNKT cell activation in the draining lymph nodes (dLNs) and prevented the protective effect of α-GalCer on bacterial dissemination out of the dLNs. Production of IFN-γ triggered chemokine (C-C motif) ligand 3 synthesis and recruitment of neutrophils in the dLNs, leading to the restraint of B. anthracis dissemination. Our data highlight a novel immunological pathway leading to the control of B. anthracis infection, a finding that might lead to improved therapeutics based on iNKT cells.


Subject(s)
Anthrax/immunology , Anthrax/microbiology , Bacillus anthracis/immunology , Natural Killer T-Cells/immunology , Animals , Anthrax/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
PLoS Pathog ; 10(10): e1004300, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25299581

ABSTRACT

Non-conventional T lymphocytes constitute a special arm of the immune system and act as sentinels against pathogens at mucosal surfaces. These non-conventional T cells (including mucosal-associated invariant T [MAIT] cells, gamma delta [γδ] T cells, and natural killer T [NKT] cells) display several innate cell-like features and are rapidly activated by the recognition of conserved, stress-induced, self, and microbial ligands. Here, we review the role of non-conventional T cells during respiratory infections, with a particular focus on the encapsulated extracellular pathogen Streptococcus pneumoniae, the leading cause of bacterial pneumonia worldwide. We consider whether MAIT cells, γδ T cells, and NKT cells might offer opportunities for preventing and/or treating human pneumococcus infections.


Subject(s)
Natural Killer T-Cells/immunology , Pneumococcal Infections/immunology , Respiratory Tract Infections/immunology , Streptococcus pneumoniae/isolation & purification , T-Lymphocyte Subsets/immunology , Animals , Humans , Mucous Membrane/immunology , Respiratory Tract Infections/etiology , Streptococcus pneumoniae/immunology
16.
Proc Natl Acad Sci U S A ; 110(36): 14711-6, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23964122

ABSTRACT

CD73 inhibits antitumor immunity through the activation of adenosine receptors expressed on multiple immune subsets. CD73 also enhances tumor metastasis, although the nature of the immune subsets and adenosine receptor subtypes involved in this process are largely unknown. In this study, we revealed that A2A/A2B receptor antagonists were effective in reducing the metastasis of tumors expressing CD73 endogenously (4T1.2 breast tumors) and when CD73 was ectopically expressed (B16F10 melanoma). A2A(-/-) mice were strongly protected against tumor metastasis, indicating that host A2A receptors enhanced tumor metastasis. A2A blockade enhanced natural killer (NK) cell maturation and cytotoxic function in vitro, reduced metastasis in a perforin-dependent manner, and enhanced NK cell expression of granzyme B in vivo, strongly suggesting that the antimetastatic effect of A2A blockade was due to enhanced NK cell function. Interestingly, A2B blockade had no effect on NK cell cytotoxicity, indicating that an NK cell-independent mechanism also contributed to the increased metastasis of CD73(+) tumors. Our results thus revealed that CD73 promotes tumor metastasis through multiple mechanisms, including suppression of NK cell function. Furthermore, our data strongly suggest that A2A or A2B antagonists may be useful for the treatment of metastatic disease. Overall, our study has potential therapeutic implications given that A2A/A2B receptor antagonists have already entered clinical trials in other therapeutic settings.


Subject(s)
5'-Nucleotidase/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Receptor, Adenosine A2A/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Granzymes/immunology , Granzymes/metabolism , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Pyrimidines/pharmacology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/immunology , Receptor, Adenosine A2B/metabolism , Triazoles/pharmacology , Xanthines/pharmacology
17.
Antimicrob Agents Chemother ; 59(10): 6064-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195519

ABSTRACT

Prophylactic intranasal administration of the Toll-like receptor 5 (TLR5) agonist flagellin protects mice against respiratory pathogenic bacteria. We hypothesized that TLR5-mediated stimulation of lung immunity might improve the therapeutic index of antibiotics for the treatment of Streptococcus pneumoniae respiratory infections in mice. Intranasal administration of flagellin was combined with either oral administration of amoxicillin or intraperitoneal injection of trimethoprim-sulfamethoxazole to treat S. pneumoniae-infected animals. Compared with standalone treatments, the combination of antibiotic and flagellin resulted in a lower bacterial load in the lungs and greater protection against S. pneumoniae dissemination and was associated with an early increase in neutrophil infiltration in the airways. The antibiotic-flagellin combination treatment was, however, not associated with any exacerbation of inflammation. Moreover, combination treatment was more efficacious than standalone antibiotic treatments in the context of post-influenza virus pneumococcal infection. Lastly, TLR5 signaling was shown to be mandatory for the efficacy of the combined antibacterial therapy. This report is the first to show that combining antibiotic treatment with the stimulation of mucosal innate immunity is a potent antibacterial strategy against pneumonia.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Flagellin/therapeutic use , Pneumococcal Infections/drug therapy , Toll-Like Receptor 5/agonists , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Amoxicillin/therapeutic use , Animals , Female , Immunity, Innate/drug effects , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity
18.
Eur J Immunol ; 44(7): 2111-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687687

ABSTRACT

The NLRP3 inflammasome plays a crucial role in the innate immune response to pathogens and exogenous or endogenous danger signals. Its activity must be precisely and tightly regulated to generate tailored immune responses. However, the immune cell subsets and cytokines controlling NLRP3 inflammasome activity are still poorly understood. Here, we have shown a link between NKT-cell-mediated TNF-α and NLRP3 inflammasome activity. The NLRP3 inflammasome in APCs was critical to potentiate NKT-cell-mediated immune responses, since C57BL/6 NLRP3 inflammasome-deficient mice exhibited reduced responsiveness to α-galactosylceramide. Importantly, NKT cells were found to act as regulators of NLRP3 inflammasome signaling, as NKT-cell-derived TNF-α was required for optimal IL-1ß and IL-18 production by myeloid cells in response to α-galactosylceramide, by acting on the NLRP3 inflammasome priming step. Thus, NKT cells play a role in the positive regulation of NLRP3 inflammasome priming by mediating the production of TNF-α, thus demonstrating another means by which NKT cells control early inflammation.


Subject(s)
Carrier Proteins/physiology , Inflammation/etiology , Natural Killer T-Cells/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigen-Presenting Cells/physiology , Cytokines/biosynthesis , Galactosylceramides/pharmacology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein
19.
J Virol ; 87(12): 6911-24, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596287

ABSTRACT

Interleukin-22 (IL-22) has redundant, protective, or pathogenic functions during autoimmune, inflammatory, and infectious diseases. Here, we addressed the potential role of IL-22 in host defense and pathogenesis during lethal and sublethal respiratory H3N2 influenza A virus (IAV) infection. We show that IL-22, as well as factors associated with its production, are expressed in the lung tissue during the early phases of IAV infection. Our data indicate that retinoic acid receptor-related orphan receptor-γt (RORγt)-positive αß and γδ T cells, as well as innate lymphoid cells, expressed enhanced Il22 transcripts as early as 2 days postinfection. During lethal or sublethal IAV infections, endogenous IL-22 played no role in the control of IAV replication and in the development of the IAV-specific CD8(+) T cell response. During lethal infection, where wild-type (WT) mice succumbed to severe pneumonia, the lack of IL-22 did not accelerate or delay IAV-associated pathogenesis and animal death. In stark contrast, during sublethal IAV infection, IL-22-deficient animals had enhanced lung injuries and showed a lower airway epithelial integrity relative to WT littermates. Of importance, the protective effect of endogenous IL-22 in pulmonary damages was associated with a more controlled secondary bacterial infection. Indeed, after challenge with Streptococcus pneumoniae, IAV-experienced Il22(-/-) animals were more susceptible than WT controls in terms of survival rate and bacterial burden in the lungs. Together, IL-22 plays no major role during lethal influenza but is beneficial during sublethal H3N2 IAV infection, where it limits lung inflammation and subsequent bacterial superinfections.


Subject(s)
Bacterial Infections/immunology , Coinfection/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Interleukins/immunology , Orthomyxoviridae Infections/immunology , Pneumonia/immunology , Animals , Bacterial Infections/microbiology , Coinfection/microbiology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Pneumonia/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Streptococcus pneumoniae/pathogenicity , Interleukin-22
20.
Blood ; 120(15): 3019-29, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22932803

ABSTRACT

Immunomodulators are effective in controlling hematologic malignancy by initiating or reactivating host antitumor immunity to otherwise poorly immunogenic and immune suppressive cancers. We aimed to boost antitumor immunity in B-cell lymphoma by developing a tumor cell vaccine incorporating α-galactosylceramide (α-GalCer) that targets the immune adjuvant properties of NKT cells. In the Eµ-myc transgenic mouse model, single therapeutic vaccination of irradiated, α-GalCer-loaded autologous tumor cells was sufficient to significantly inhibit growth of established tumors and prolong survival. Vaccine-induced antilymphoma immunity required NKT cells, NK cells, and CD8 T cells, and early IL-12-dependent production of IFN-γ. CD4 T cells, gamma/delta T cells, and IL-18 were not critical. Vaccine treatment induced a large systemic spike of IFN-γ and transient peripheral expansion of both NKT cells and NK cells, the major sources of IFN-γ. Furthermore, this vaccine approach was assessed in several other hematopoietic tumor models and was also therapeutically effective against AML-ETO9a acute myeloid leukemia. Replacing α-GalCer with ß-mannosylceramide resulted in prolonged protection against Eµ-myc lymphoma. Overall, our results demonstrate a potent immune adjuvant effect of NKT cell ligands in therapeutic anticancer vaccination against oncogene-driven lymphomas, and this work supports clinical investigation of NKT cell-based immunotherapy in patients with hematologic malignancies.


Subject(s)
Cancer Vaccines/therapeutic use , Galactosylceramides/administration & dosage , Genes, myc/genetics , Immunotherapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Natural Killer T-Cells/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Genes, T-Cell Receptor delta/physiology , Humans , Interferon-gamma/metabolism , Interleukin-12/physiology , Interleukin-18/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphoma, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL