Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 324
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.
Nat Immunol ; 21(9): 1034-1045, 2020 09.
Article in English | MEDLINE | ID: mdl-32661363

ABSTRACT

Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Macrophages/immunology , Microbiota/immunology , Neutrophils/immunology , Skin/immunology , Animals , Cells, Cultured , Chemokine CXCL10/metabolism , Humans , Immunity, Innate , Inflammation , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology , Symbiosis , Wound Healing
3.
Nat Immunol ; 19(2): 130-140, 2018 02.
Article in English | MEDLINE | ID: mdl-29255269

ABSTRACT

Reactive oxygen species (ROS) are generated by virus-infected cells; however, the physiological importance of ROS generated under these conditions is unclear. Here we found that the inflammation and cell death induced by exposure of mice or cells to sources of ROS were not altered in the absence of canonical ROS-sensing pathways or known cell-death pathways. ROS-induced cell-death signaling involved interactions among the cellular ROS sensor and antioxidant factor KEAP1, the phosphatase PGAM5 and the proapoptotic factor AIFM1. Pgam5 -/- mice showed exacerbated lung inflammation and proinflammatory cytokines in an ozone-exposure model. Similarly, challenge with influenza A virus led to increased infiltration of the virus, lymphocytic bronchiolitis and reduced survival of Pgam5 -/- mice. This pathway, which we have called 'oxeiptosis', was a ROS-sensitive, caspase independent, non-inflammatory cell-death pathway and was important for protection against inflammation induced by ROS or ROS-generating agents such as viral pathogens.


Subject(s)
Cell Death/physiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis Inducing Factor/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction/physiology
4.
Immunity ; 54(9): 2024-2041.e8, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34473957

ABSTRACT

Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.


Subject(s)
Adenosine/immunology , Antigens, CD/immunology , Apyrase/immunology , Immune Tolerance/immunology , Macrophages/immunology , Plasma Cells/immunology , Sepsis/immunology , Adenosine/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Cellular Reprogramming/immunology , Macrophages/metabolism , Mice , Plasma Cells/metabolism , Receptor, Adenosine A2A/immunology , Receptor, Adenosine A2A/metabolism , Sepsis/metabolism
5.
Nat Immunol ; 16(8): 859-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26098997

ABSTRACT

The receptor NLRP3 is involved in the formation of the NLRP3 inflammasome that activates caspase-1 and mediates the release of interleukin 1ß (IL-1ß) and IL-18. Whether NLRP3 can shape immunological function independently of inflammasomes is unclear. We found that NLRP3 expression in CD4(+) T cells specifically supported a T helper type 2 (TH2) transcriptional program in a cell-intrinsic manner. NLRP3, but not the inflammasome adaptor ASC or caspase-1, positively regulated a TH2 program. In TH2 cells, NLRP3 bound the Il4 promoter and transactivated it in conjunction with the transcription factor IRF4. Nlrp3-deficient TH2 cells supported melanoma tumor growth in an IL-4-dependent manner and also promoted asthma-like symptoms. Our results demonstrate the ability of NLRP3 to act as a key transcription factor in TH2 differentiation.


Subject(s)
Carrier Proteins/immunology , Cell Differentiation/immunology , Th2 Cells/immunology , Trans-Activators/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Binding/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
6.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006013

ABSTRACT

The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.


Subject(s)
Cytokines/immunology , Gene Expression/immunology , Interleukins/immunology , Rotavirus Infections/immunology , Animals , Caco-2 Cells , Cell Line , Chlorocebus aethiops , Cytokines/genetics , Cytokines/pharmacology , Dogs , Drug Synergism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression/drug effects , HT29 Cells , Humans , Immunoblotting , Interleukins/genetics , Interleukins/pharmacology , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/virology , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Reverse Transcriptase Polymerase Chain Reaction , Rotavirus Infections/genetics , Rotavirus Infections/virology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Vero Cells , Interleukin-22
7.
Nat Immunol ; 15(8): 758-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973819

ABSTRACT

The TH9 subset of helper T cells was initially shown to contribute to the induction of autoimmune and allergic diseases, but subsequent evidence has suggested that these cells also exert antitumor activities. However, the molecular events that account for their effector properties are elusive. Here we found that the transcription factor IRF1 enhanced the effector function of TH9 cells and dictated their anticancer properties. Under TH9-skewing conditions, interleukin 1ß (IL-1ß) induced phosphorylation of the transcription factor STAT1 and subsequent expression of IRF1, which bound to the promoters of Il9 and Il21 and enhanced secretion of the cytokines IL-9 and IL-21 from TH9 cells. Furthermore, IL-1ß-induced TH9 cells exerted potent anticancer functions in an IRF1- and IL-21-dependent manner. Our findings thus identify IRF1 as a target for controlling the function of TH9 cells.


Subject(s)
Interferon Regulatory Factor-1/immunology , Interleukins/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Helper-Inducer/immunology , 3T3 Cells , Animals , Base Sequence , Cell Line , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interferon Regulatory Factor-1/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/antagonists & inhibitors , Interleukin-10/immunology , Interleukin-9/genetics , Interleukin-9/immunology , Interleukin-9/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Phosphorylation/immunology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , RNA Interference , RNA, Small Interfering , STAT1 Transcription Factor/immunology , Sequence Analysis, RNA , T-Lymphocytes, Helper-Inducer/metabolism
8.
Trends Immunol ; 44(12): 1014-1030, 2023 12.
Article in English | MEDLINE | ID: mdl-37951789

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.


Subject(s)
Idiopathic Pulmonary Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/metabolism , Chemokines/metabolism , Macrophages, Alveolar , Cytokines/metabolism , Signal Transduction , Lung
9.
Inflamm Res ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907167

ABSTRACT

BACKGROUND: Senescence is a cellular aging-related process triggered by different stresses and characterized by the secretion of various inflammatory factors referred to as senescence-associated secretory phenotype (SASP), some of which are produced by the NLRP3 inflammasome. Here, we present evidence that the NLRP1 inflammasome is a DNA damage sensor and a key mediator of senescence. METHODS: Senescence was induced in fibroblasts in vitro and in mice. Cellular senescence was assessed by Western blot analysis of several proteins, including p16, p21, p53, and SASP factors, released in the culture media or serum. Inflammasome components, including NLRP1, NLRP3 and GSDMD were knocked out or silenced using siRNAs. RESULTS: In vitro and in vivo results suggest that the NLRP1 inflammasome promotes senescence by regulating the expression of p16, p21, p53, and SASP factors in a Gasdermin D (GSDMD)-dependent manner. Mechanistically, the NLRP1 inflammasome is activated in response to genomic damage detected by the cytosolic DNA sensor cGMP-AMP (cGAMP) synthase (cGAS). CONCLUSION: Our findings show that NLRP1 is a cGAS-dependent DNA damage sensor during senescence and a mediator of SASP release through GSDMD. This study advances the knowledge on the biology of the NLRP1 inflammasome and highlights this pathway as a potential pharmcological target to modulate senescence.

10.
Arch Toxicol ; 97(4): 1001-1015, 2023 04.
Article in English | MEDLINE | ID: mdl-36840754

ABSTRACT

IL-1α is an intracellular danger signal (DAMP) released by macrophages contributing to the development of silica-induced lung inflammation. The exact molecular mechanism orchestrating IL-1α extracellular release from particle-exposed macrophages is still unclear. To delineate this process, murine J774 and bone-marrow derived macrophages were exposed to increasing concentrations (1-40 cm2/ml) of a set of amorphous and crystalline silica particles with different surface chemical features. In particular, these characteristics include the content of nearly free silanols (NFS), a silanol population responsible for silica cytotoxicity recently identified. We first observed de novo stocks of IL-1α in macrophages after silica internalization regardless of particle physico-chemical characteristics and cell stress. IL-1α intracellular production and accumulation were observed by exposing macrophages to biologically-inert or cytotoxic crystalline and amorphous silicas. In contrast, only NFS-rich reactive silica particles triggered IL-1α release into the extracellular milieu from necrotic macrophages. We demonstrate that IL-1α is actively secreted through the formation of gasdermin D (GSDMD) pores in the plasma membrane and not passively released after macrophage plasma membrane lysis. Our findings indicate that the GSDMD pore-dependent secretion of IL-1α stock from macrophages solely depends on cytotoxicity induced by NFS-rich silica. This new regulated process represents a key first event in the mechanism of silica toxicity, suitable to refine the existing adverse outcome pathway (AOP) for predicting the inflammatory activity of silicas.


Subject(s)
Gasdermins , Macrophages , Mice , Animals , Interleukin-1beta/metabolism , Macrophages/metabolism , Necrosis , Silicon Dioxide/chemistry
12.
Allergy ; 77(11): 3320-3336, 2022 11.
Article in English | MEDLINE | ID: mdl-35615773

ABSTRACT

BACKGROUND: Inflammasomes are large protein complexes that assemble in the cytosol in response to danger such as tissue damage or infection. Following activation, inflammasomes trigger cell death and the release of biologically active forms of pro-inflammatory cytokines interleukin (IL)-1ß and IL-18. NOD-like receptor family pyrin domain containing 6 (NLRP6) inflammasome is required for IL-18 secretion by intestinal epithelial cells, macrophages, and T cells, contributing to homeostasis and self-defense against pathogenic microbes. However, the involvement of NLRP6 in type 2 lung inflammation remains elusive. METHODS: Wild-type (WT) and Nlrp6-/- mice were used. Birch pollen extract (BPE)-induced allergic lung inflammation, eosinophil recruitment, Th2-related cytokine and chemokine production, airway hyperresponsiveness, and lung histopathology, Th2 cell differentiation, GATA3, and Th2 cytokines expression, were determined. Nippostrongylus brasiliensis (Nb) infection, worm count in intestine, type 2 innate lymphoid cell (ILC2), and Th2 cells in lungs were evaluated. RESULTS: We demonstrate in Nlrp6-/- mice that a mixed Th2/Th17 immune responses prevailed following birch pollen challenge with increased eosinophils, ILC2, Th2, and Th17 cell induction and reduced IL-18 production. Nippostrongylus brasiliensis infected Nlrp6-/- mice featured enhanced early expulsion of the parasite due to enhanced type 2 immune responses compared to WT hosts. In vitro, NLRP6 repressed Th2 polarization, as shown by increased Th2 cytokines and higher expression of the transcription factor GATA3 in the absence of NLRP6. Exogenous IL-18 administration partially reduced the enhanced airways inflammation in Nlrp6-/- mice. CONCLUSIONS: In summary, our data identify NLRP6 as a negative regulator of type 2 immune responses.


Subject(s)
Immunity, Innate , Pneumonia , Animals , Mice , Cytokines/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Lymphocytes , Mice, Knockout , Nippostrongylus , Pneumonia/metabolism , Th2 Cells
13.
Trends Immunol ; 40(8): 719-734, 2019 08.
Article in English | MEDLINE | ID: mdl-31262653

ABSTRACT

Self-DNA sensing by the immune system has emerged as a key contributing response in the pathogenesis of cancer and autoimmune diseases. Recent studies have established that release of nuclear and mitochondrial DNA can also drive lung inflammatory diseases. Here, we review the latest advances on self-DNA sensing and signaling, the influence of these pathways on lung inflammation, and how these findings contribute to our understanding of basic mechanisms of innate immunity. Within a dozen DNA sensors, the cGAS/STING, inflammasomes and Toll-Like Receptor pathways are central to nucleic acid sensing. We propose a key role for the STING pathway in self-DNA sensing in inflammatory lung conditions, and identify major remaining questions that may further our understanding and potential to control self-DNA sensing and innate immune activation.


Subject(s)
DNA/immunology , Disease Susceptibility , Host-Pathogen Interactions/immunology , Pneumonia/etiology , Pneumonia/metabolism , Animals , Autoimmunity , Biomarkers , Disease Susceptibility/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction
14.
Int J Mol Sci ; 23(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35408954

ABSTRACT

Natterin is a potent pro-inflammatory fish molecule, inducing local and systemic IL-1ß/IL-1R1-dependent neutrophilia mediated by non-canonical NLRP6 and NLRC4 inflammasome activation in mice, independent of NLRP3. In this work, we investigated whether Natterin activates mitochondrial damage, resulting in self-DNA leaks into the cytosol, and whether the DNA sensor cGAS and STING pathway participate in triggering the innate immune response. Employing a peritonitis mouse model, we found that the deficiency of the tlr2/tlr4, myd88 and trif results in decreased neutrophil influx to peritoneal cavities of mice, indicative that in addition to MyD88, TRIF contributes to neutrophilia triggered by TLR4 engagement by Natterin. Next, we demonstrated that gpcr91 deficiency in mice abolished the neutrophil recruitment after Natterin injection, but mice pre-treated with 2-deoxy-d-glucose that blocks glycolysis presented similar infiltration than WT Natterin-injected mice. In addition, we observed that, compared with the WT Natterin-injected mice, DPI and cyclosporin A treated mice had a lower number of neutrophils in the peritoneal exudate. The levels of dsDNA in the supernatant of the peritoneal exudate and processed IL-33 in the supernatant of the peritoneal exudate or cytoplasmic supernatant of the peritoneal cell lysate of WT Natterin-injected mice were several folds higher than those of the control mice. The recruitment of neutrophils to peritoneal cavity 2 h post-Natterin injection was intensely impaired in ifnar KO mice and partially in il-28r KO mice, but not in ifnγr KO mice. Finally, using cgas KO, sting KO, or irf3 KO mice we found that recruitment of neutrophils to peritoneal cavities was virtually abolished in response to Natterin. These findings reveal cytosolic DNA sensors as critical regulators for Natterin-induced neutrophilia.


Subject(s)
Myeloid Differentiation Factor 88 , Toll-Like Receptor 4 , Adaptor Proteins, Vesicular Transport/metabolism , Animals , DNA , Fish Venoms , Membrane Proteins/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Nucleotidyltransferases/metabolism , Pore Forming Cytotoxic Proteins , Signal Transduction , Toll-Like Receptor 4/metabolism
15.
J Cell Mol Med ; 25(10): 4721-4731, 2021 05.
Article in English | MEDLINE | ID: mdl-33734594

ABSTRACT

The aryl hydrocarbon receptor (AHR) controls several inflammatory and metabolic pathways involved in various diseases, including the development of arthritis. Here, we investigated the role of AHR activation in IL-22-dependent acute arthritis using the K/BxN serum transfer model. We observed an overall reduction of cytokine expression in Ahr-deficient mice, along with decreased signs of joint inflammation. Conversely, we report worsened arthritis symptoms in Il-22 deficient mice. Pharmacological stimulation of AHR with the agonist VAG539, as well as injection of recombinant IL-22, given prior arthritogenic triggering, attenuated inflammation and reduced joint destruction. The protective effect of VAG539 was abrogated in Il-22 deficient mice. Finally, conditional Ahr depletion of Rorc-expressing cells was sufficient to attenuate arthritis, thereby uncovering a previously unsuspected role of AHR in type 3 innate lymphoid cells during acute arthritis.


Subject(s)
Arthritis, Experimental/pathology , Basic Helix-Loop-Helix Transcription Factors/physiology , Immunity, Innate/immunology , Inflammation/pathology , Interleukins/physiology , Joints/pathology , Lymphocytes/pathology , Receptors, Aryl Hydrocarbon/physiology , Acute Disease , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Female , Inflammation/etiology , Inflammation/metabolism , Joints/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22
16.
PLoS Pathog ; 15(12): e1008155, 2019 12.
Article in English | MEDLINE | ID: mdl-31856218

ABSTRACT

Cellular response to environmental challenges requires immediate and precise regulation of transcriptional programs. During viral infections, this includes the expression of antiviral genes that are essential to combat the pathogen. Transcribed mRNAs are bound and escorted to the cytoplasm by the cap-binding complex (CBC). We recently identified a protein complex consisting of NCBP1 and NCBP3 that, under physiological conditions, has redundant function to the canonical CBC, consisting of NCBP1 and NCBP2. Here, we provide evidence that NCBP3 is essential to mount a precise and appropriate antiviral response. Ncbp3-deficient cells allow higher virus growth and elicit a reduced antiviral response, a defect happening on post-transcriptional level. Ncbp3-deficient mice suffered from severe lung pathology and increased morbidity after influenza A virus challenge. While NCBP3 appeared to be particularly important during viral infections, it may be more broadly involved to ensure proper protein expression.


Subject(s)
Orthomyxoviridae Infections/immunology , RNA Cap-Binding Proteins/immunology , RNA Cap-Binding Proteins/metabolism , Animals , Influenza A virus/immunology , Mice , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Protein Biosynthesis/physiology
17.
Immunity ; 36(3): 362-73, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22406269

ABSTRACT

Although Th17 cells are known to promote tissue inflammation and autoimmunity, their role during cancer progression remains elusive. Here, we showed that in vitro Th17 cells generated with the cytokines IL-6 and TGF-ß expressed CD39 and CD73 ectonucleotidases, leading to adenosine release and the subsequent suppression of CD4(+) and CD8(+) T cell effector functions. The IL-6-mediated activation of the transcription factor Stat3 and the TGF-ß-driven downregulation of Gfi-1 transcription factor were both essential for the expression of ectonucleotidases during Th17 cell differentiation. Stat3 supported whereas Gfi-1 repressed CD39 and CD73 expression by binding to their promoters. Accordingly, Th17 cells differentiated with IL-1ß, IL-6, and IL-23 but without TGF-ß did not express ectonucleotidases and were not immunosuppressive. Finally, adoptive transfer of Th17 cells induced by TGF-ß and IL-6 promoted tumor growth in a CD39-dependent manner. Thus, ectonucleotidase expression supports the immunosuppressive fate of Th17 cells in cancer.


Subject(s)
5'-Nucleotidase/genetics , Antigens, CD/genetics , Apyrase/genetics , DNA-Binding Proteins/immunology , STAT3 Transcription Factor/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/immunology , Animals , Binding Sites/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Interleukin-6/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , RNA, Small Interfering/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th17 Cells/drug effects , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology
18.
Arterioscler Thromb Vasc Biol ; 40(11): 2619-2631, 2020 11.
Article in English | MEDLINE | ID: mdl-32907372

ABSTRACT

OBJECTIVE: The occurrence of new blood vessel formation in the lungs of asthmatic patients suggests a critical role for airway endothelial cells (ECs) in the disease. IL-33 (Interleukin-33)-a cytokine abundantly expressed in human lung ECs-recently emerged as a key factor in the development of allergic diseases, including asthma. In the present study, we evaluated whether mouse and human ECs exposed to the common Dermatophagoides farinae allergen produce IL-33 and characterized the activated signaling pathways. Approach and Results: Mouse primary lung ECs were exposed in vitro to D farinae extract or rmIL-33 (recombinant murine IL-33). Both D farinae and rmIL-33 induced Il-33 transcription without increasing the IL-33 production and upregulated the expression of its receptor, as well as genes involved in angiogenesis and the regulation of immune responses. In particular, D farinae and rmIL-33 upregulated Fas/Cd95 transcript level, yet without promoting apoptosis. Inhibition of caspases involved in the Fas signaling pathway, increased IL-33 protein level in ECs, suggesting that Fas may decrease IL-33 level through caspase-8-dependent mechanisms. Our data also showed that the NF-κB (nuclear factor-κB), PI3K/Akt, and Wnt/ß-catenin pathways regulate Il-33 transcription in both mouse and human primary ECs. CONCLUSIONS: Herein, we described a new mechanism involved in the control of IL-33 production in lung ECs exposed to allergens.


Subject(s)
Antigens, Dermatophagoides/pharmacology , Endothelial Cells/drug effects , Interleukin-33/pharmacology , Lung/blood supply , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , fas Receptor/metabolism , Animals , Caspase 8/metabolism , Cell Line , Endothelial Cells/enzymology , Endothelial Cells/immunology , Enzyme Activation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Interleukin-33/genetics , Interleukin-33/metabolism , Mice , Up-Regulation , Wnt Signaling Pathway , fas Receptor/genetics
19.
J Neuroinflammation ; 17(1): 268, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32917228

ABSTRACT

BACKGROUND: Interleukin (IL)-33 is expressed in a healthy brain and plays a pivotal role in several neuropathologies, as protective or contributing to the development of cerebral diseases associated with cognitive impairments. However, the role of IL-33 in the brain is poorly understood, raising the question of its involvement in immunoregulatory mechanisms. METHODS: We administered recombinant IL-33 (rmIL-33) by intra-hippocampal injection to C57BL/6 J (WT) and IL-1αß deficient mice. Chronic minocycline administration was performed and cognitive functions were examined trough spatial habituation test. Hippocampal inflammatory responses were investigated by RT-qPCR. The microglia activation was assessed using immunohistological staining and fluorescence-activated cell sorting (FACS). RESULTS: We showed that IL-33 administration in mice led to a spatial memory performance defect associated with an increase of inflammatory markers in the hippocampus while minocycline administration limited the inflammatory response. Quantitative assessment of glial cell activation in situ demonstrated an increase of proximal intersections per radius in each part of the hippocampus. Moreover, rmIL-33 significantly promoted the outgrowth of microglial processes. Fluorescence-activated cell sorting analysis on isolated microglia, revealed overexpression of IL-1ß, 48 h post-rmIL-33 administration. This microglial reactivity was closely related to the onset of cognitive disturbance. Finally, we demonstrated that IL-1αß deficient mice were resistant to cognitive disorders after intra-hippocampal IL-33 injection. CONCLUSION: Thus, hippocampal IL-33 induced an inflammatory state, including IL-1ß overexpression by microglia cells, being causative of the cognitive impairment. These results highlight the pathological role for IL-33 in the central nervous system, independently of a specific neuropathological model.


Subject(s)
Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Inflammation/metabolism , Interleukin-33/pharmacology , Animals , Cognitive Dysfunction/etiology , Hippocampus/drug effects , Inflammation/complications , Mice , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Minocycline/pharmacology , Spatial Memory/drug effects , Spatial Memory/physiology
20.
Cell Commun Signal ; 18(1): 141, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32894139

ABSTRACT

BACKGROUND: Low molecular weight carrageenan (Cg) is a seaweed-derived sulfated polysaccharide widely used as inflammatory stimulus in preclinical studies. However, the molecular mechanisms of Cg-induced inflammation are not fully elucidated. The present study aimed to investigate the molecular basis involved in Cg-induced macrophages activation and cytokines production. METHODS: Primary culture of mouse peritoneal macrophages were stimulated with Kappa Cg. The supernatant and cell lysate were used for ELISA, western blotting, immunofluorescence. Cg-induced mouse colitis was also developed. RESULTS: Here we show that Cg activates peritoneal macrophages to produce pro-inflammatory cytokines such as TNF and IL-1ß. While Cg-induced TNF production/secretion depends on TLR4/MyD88 signaling, the production of pro-IL-1ß relies on TLR4/TRIF/SYK/reactive oxygen species (ROS) signaling pathway. The maturation of pro-IL1ß into IL-1ß is dependent on canonical NLRP3 inflammasome activation via Pannexin-1/P2X7/K+ efflux signaling. In vivo, Cg-induced colitis was reduced in mice in the absence of NLRP3 inflammasome components. CONCLUSIONS: In conclusion, we unravel a critical role of the NLRP3 inflammasome in Cg-induced pro-inflammatory cytokines production and colitis, which is an important discovery on the pro-inflammatory properties of this sulfated polysaccharide for pre-clinical studies. Video abstract Carrageenan (Cg) is one the most used flogistic stimulus in preclinical studies. Nevertheless, the molecular basis of Cg-induced inflammation is not totally elucidated. Herein, Lopes et al. unraveled the molecular basis for Cg-induced macrophages production of biological active IL-1ß. The Cg-stimulated macrophages produces pro-IL-1ß depends on TLR4/TRIF/Syk/ROS, whereas its processing into mature IL-1ß is dependent on the canonical NLRP3 inflammasome.


Subject(s)
Carrageenan/immunology , Cytokines/immunology , Macrophage Activation , Macrophages, Peritoneal/immunology , Animals , Cells, Cultured , Inflammasomes/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL