Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
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
2.
Exp Eye Res ; 246: 110015, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39089568

ABSTRACT

Fragile X Syndrome (FXS), the most common inherited form of human intellectual disability, is a monogenic neurodevelopmental disorder caused by a loss-of-function mutation of the FMR1 gene. FMR1 is encoding the Fragile X Messenger Ribonucleo Protein (FMRP) an RNA-binding protein that regulates the translation of synaptic proteins. The absence of FMRP expression has many important consequences on synaptic plasticity and function, leading to the FXS clinical phenotype. Over the last decade, a visual neurosensorial phenotype had been described in the FXS patients as well as in the murine model (Fmr1-/ymice), characterized by retinal deficits associated to retinal perception alterations. However, although the transcriptomic profile in the absence of FMRP has been studied in the cerebral part of the central nervous system (CNS), there are no actual data for the retina which is an extension of the CNS. Herein, we investigate the transcriptomic profile of mRNA from whole retinas of Fmr1-/ymice. Interestingly, we found a specific signature of Fmrp absence on retinal mRNA expression with few common genes compared to other brain studies. Gene Ontology on these retinal specific genes demonstrated an enrichment in retinal development genes as well as in synaptic genes. These alterations could be linked to the reported retinal phenotype of the FXS condition. In conclusion, we describe for the first time, retinal-specific transcriptomic changes in the absence of FMRP.

4.
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
5.
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
6.
J Allergy Clin Immunol ; 148(2): 394-406, 2021 08.
Article in English | MEDLINE | ID: mdl-33508265

ABSTRACT

BACKGROUND: Asthma severity has been linked to exposure to gram-negative bacteria from the environment that are recognized by NOD1 receptor and are present in house dust mite (HDM) extracts. NOD1 polymorphism has been associated with asthma. OBJECTIVE: We sought to evaluate whether either host or HDM-derived microbiota may contribute to NOD1-dependent disease severity. METHODS: A model of HDM-induced experimental asthma was used and the effect of NOD1 deficiency was evaluated. Contribution of host microbiota was evaluated by fecal transplantation. Contribution of HDM-derived microbiota was assessed by 16S ribosomal RNA sequencing, mass spectrometry analysis, and peptidoglycan depletion of the extracts. RESULTS: In this model, loss of the bacterial sensor NOD1 and its adaptor RIPK2 improved asthma features. Such inhibitory effect was not related to dysbiosis caused by NOD1 deficiency, as shown by fecal transplantation of Nod1-deficient microbiota to wild-type germ-free mice. The 16S ribosomal RNA gene sequencing and mass spectrometry analysis of HDM allergen, revealed the presence of some muropeptides from gram-negative bacteria that belong to the Bartonellaceae family. While such HDM-associated muropeptides were found to activate NOD1 signaling in epithelial cells, peptidoglycan-depleted HDM had a decreased ability to instigate asthma in vivo. CONCLUSIONS: These data show that NOD1-dependent sensing of HDM-associated gram-negative bacteria aggravates the severity of experimental asthma, suggesting that inhibiting the NOD1 signaling pathway may be a therapeutic approach to treating asthma.


Subject(s)
Asthma/immunology , Gastrointestinal Microbiome/immunology , Nod1 Signaling Adaptor Protein/immunology , Pyroglyphidae/immunology , Signal Transduction/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/microbiology , Disease Models, Animal , Female , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Signal Transduction/genetics
7.
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
8.
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
9.
Int J Mol Sci ; 23(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008536

ABSTRACT

This study aimed at evaluating the effects of the micro-immunotherapy medicine (MIM) 2LEID, both in vitro and in vivo, on several components of the innate and adaptive immune system. MIM increased the phagocytic activity of macrophages, and it augmented the expression of the activation markers CD69 and HLA-DR in NK cells and monocytes/macrophages, respectively. The effect of MIM was evaluated in a model of respiratory infection induced by influenza A virus administration to immunocompetent mice in which it was able to improve neutrophil recruitment within the lungs (p = 0.1051) and slightly increased the circulating levels of IgM (p = 0.1655). Furthermore, MIM stimulated the proliferation of CD3-primed T lymphocytes and decreased the secretion of the immunosuppressive cytokine IL-10 in CD14+-derived macrophages. Human umbilical vein endothelial cells were finally used to explore the effect of MIM on endothelial cells, in which it slightly increased the expression of immune-related markers such as HLA-I, CD137L, GITRL, PD-L1 and ICAM-1. In conclusion, the present study suggests that MIM might be a promising nonspecific (without antigen specificity) immunostimulant drug in preventing and early treating respiratory infections, but not only exclusively, as it would gently support several facets of the immune system and host defenses.


Subject(s)
Adaptive Immunity/drug effects , Adjuvants, Immunologic/pharmacology , Immunity, Innate/drug effects , Adaptive Immunity/immunology , Animals , Biomarkers/metabolism , Cell Proliferation/physiology , Cells, Cultured , Cytokines/immunology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Humans , Immunity, Innate/immunology , Immunotherapy/methods , Interleukin-10/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lung/drug effects , Lung/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/immunology , Neutrophils/drug effects , Neutrophils/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
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
11.
J Allergy Clin Immunol ; 142(3): 942-958, 2018 09.
Article in English | MEDLINE | ID: mdl-29331644

ABSTRACT

BACKGROUND: IL-33 plays a critical role in regulation of tissue homeostasis, injury, and repair. Whether IL-33 regulates neutrophil recruitment and functions independently of airways hyperresponsiveness (AHR) in the setting of ozone-induced lung injury and inflammation is unclear. OBJECTIVE: We sought to examine the role of the IL-33/ST2 axis in lung inflammation on acute ozone exposure in mice. METHODS: ST2- and Il33-deficient, IL-33 citrine reporter, and C57BL/6 (wild-type) mice underwent a single ozone exposure (1 ppm for 1 hour) in all studies. Cell recruitment in lung tissue and the bronchoalveolar space, inflammatory parameters, epithelial barrier damage, and airway hyperresponsiveness (AHR) were determined. RESULTS: We report that a single ozone exposure causes rapid disruption of the epithelial barrier within 1 hour, followed by a second phase of respiratory barrier injury with increased neutrophil recruitment, reactive oxygen species production, AHR, and IL-33 expression in epithelial and myeloid cells in wild-type mice. In the absence of IL-33 or IL-33 receptor/ST2, epithelial cell injury with protein leak and myeloid cell recruitment and inflammation are further increased, whereas the tight junction proteins E-cadherin and zonula occludens 1 and reactive oxygen species expression in neutrophils and AHR are diminished. ST2 neutralization recapitulated the enhanced ozone-induced neutrophilic inflammation. However, myeloid cell depletion using GR-1 antibody reduced ozone-induced lung inflammation, epithelial cell injury, and protein leak, whereas administration of recombinant mouse IL-33 reduced neutrophil recruitment in Il33-deficient mice. CONCLUSION: Data demonstrate that ozone causes an immediate barrier injury that precedes myeloid cell-mediated inflammatory injury under the control of the IL-33/ST2 axis. Thus IL-33/ST2 signaling is critical for maintenance of intact epithelial barrier and inflammation.


Subject(s)
Air Pollutants/toxicity , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/immunology , Lung Injury/immunology , Oxidants/toxicity , Ozone/toxicity , Animals , Female , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology
12.
J Allergy Clin Immunol ; 139(5): 1650-1666, 2017 May.
Article in English | MEDLINE | ID: mdl-27746240

ABSTRACT

BACKGROUND: Protein kinase C (PKC) θ, a serine/threonine kinase, is involved in TH2 cell activation and proliferation. Type 2 innate lymphoid cells (ILC2s) resemble TH2 cells and produce the TH2 cytokines IL-5 and IL-13 but lack antigen-specific receptors. The mechanism by which PKC-θ drives innate immune cells to instruct TH2 responses in patients with allergic lung inflammation remains unknown. OBJECTIVES: We hypothesized that PKC-θ contributes to ILC2 activation and might be necessary for ILC2s to instruct the TH2 response. METHODS: PRKCQ gene expression was assessed in innate lymphoid cell subsets purified from human PBMCs and mouse lung ILC2s. ILC2 activation and eosinophil recruitment, TH2-related cytokine and chemokine production, lung histopathology, interferon regulatory factor 4 (IRF4) mRNA expression, and nuclear factor of activated T cells (NFAT1) protein expression were determined. Adoptive transfer of ILC2s from wild-type mice was performed in wild-type and PKC-θ-deficient (PKC-θ-/-) mice. RESULTS: Here we report that PKC-θ is expressed in both human and mouse ILC2s. Mice lacking PKC-θ had reduced ILC2 numbers, TH2 cell numbers and activation, airway hyperresponsiveness, and expression of the transcription factors IRF4 and NFAT1. Importantly, adoptive transfer of ILC2s restored eosinophil influx and IL-4, IL-5 and IL-13 production in lung tissue, as well as TH2 cell activation. The pharmacologic PKC-θ inhibitor (Compound 20) administered during allergen challenge reduced ILC2 numbers and activation, as well as airway inflammation and IRF4 and NFAT1 expression. CONCLUSIONS: Therefore our findings identify PKC-θ as a critical factor for ILC2 activation that contributes to TH2 cell differentiation, which is associated with IRF4 and NFAT1 expression in allergic lung inflammation.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Asthma/immunology , Isoenzymes/immunology , Lymphocytes/immunology , Protein Kinase C/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Differentiation , Cytokines/immunology , Dipeptides/pharmacology , Female , Humans , Immunity, Innate , Interferon Regulatory Factors/immunology , Isoenzymes/genetics , Leukocyte Count , Lung/cytology , Lung/immunology , Lung/pathology , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/immunology , Protein Kinase C/genetics , Protein Kinase C-theta , Protein Kinase Inhibitors/pharmacology
13.
Eur J Immunol ; 46(6): 1449-59, 2016 06.
Article in English | MEDLINE | ID: mdl-27000947

ABSTRACT

IL-22 has a detrimental role in skin inflammatory processes, for example in psoriasis. As transcription factor, AhR controls the IL-22 production by several cell types (i.e. Th17 cells). Here, we analyzed the role of Ahr in IL-22 production by immune cells in the inflamed skin, using an imiquimod-induced psoriasis mouse model. Our results indicate that IL-22 is expressed in the ear of imiquimod-treated Ahr(-/-) mice but less than in wild-type mice. We then studied the role of AhR on three cell populations known to produce IL-22 in the skin: γδ T cells, Th17 cells, and ILC3, and a novel IL-22-producing cell type identified in this setting: CD4(-) CD8(-) TCRß(+) T cells. We showed that AhR is required for IL-22 production by Th17, but not by the three other cell types, in the imiquimod-treated ears. Moreover, AhR has a role in the recruitment of γδ T cells, ILC3, and CD4(-) CD8(-) TCRß(+) T cells into the inflamed skin or in their local proliferation. Taken together, AhR has a direct role in IL-22 production by Th17 cells in the mouse ear skin, but not by γδ T cells, CD4(-) CD8(-) TCRß(+) T cells and ILCs.


Subject(s)
Aminoquinolines/adverse effects , Chemotaxis/immunology , Interleukins/biosynthesis , Psoriasis/etiology , Psoriasis/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Proliferation , Chemotaxis/genetics , Disease Models, Animal , Imiquimod , Immunity, Innate/genetics , Immunity, Innate/immunology , Interleukins/genetics , Mice , Mice, Knockout , Psoriasis/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Interleukin-22
14.
Clin Sci (Lond) ; 131(20): 2533-2548, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-29026003

ABSTRACT

T helper (Th)17 immune response participates in allergic lung inflammation and asthma is reduced in the absence of interleukin (IL)-17 in mice. Since IL-17A and IL-17F are induced and bind the shared receptor IL-17RA, we asked whether both IL-17A and IL-17F contribute to house dust mite (HDM) induced asthma. We report that allergic lung inflammation is attenuated in absence of either IL-17A or IL-17F with reduced airway hyperreactivity, eosinophilic inflammation, goblet cell hyperplasia, cytokine and chemokine production as found in absence of IL-17RA. Furthermore, specific antibody neutralization of either IL-17A or IL-17F given during the sensitization phase attenuated allergic lung inflammation and airway hyperreactivity. In vitro activation by HDM of primary dendritic cells revealed a comparable induction of CXCL1 and IL-6 expression and the response to IL-17A and IL-17F relied on IL-17RA signaling via the adaptor protein act1 in fibroblasts. Therefore, HDM-induced allergic respiratory response depends on IL-17RA via act1 signaling and inactivation of either IL-17A or IL-17F is sufficient to attenuate allergic asthma in mice.


Subject(s)
Asthma/drug therapy , Interleukin-17/antagonists & inhibitors , Pyroglyphidae/immunology , Allergens/immunology , Animals , Asthma/immunology , Dendritic Cells/immunology , Disease Models, Animal , Interleukin-17/immunology , Interleukin-6/immunology , Lung/immunology , Mice, Inbred C57BL , Th17 Cells/immunology , Th2 Cells/immunology
16.
Eur J Immunol ; 45(10): 2847-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26147228

ABSTRACT

The pathogenesis of inflammatory skin diseases such as psoriasis involves the release of numerous proinflammatory cytokines, including members of the IL-1 family. Here we report overexpression of IL-1α, IL-1ß, and IL-1 receptor antagonist mRNA, associated to expression of IL-23p19, IL-17A, and IL-22 in skin cells, upon topical application of the TLR7 agonist imiquimod (IMQ) in C57BL/6J mice. IMQ-induced skin inflammation was partially reduced in mice deficient for both IL-1α/IL-1ß or for IL-1 receptor type 1 (IL-1R1), but not in IL-1α- or IL-1ß-deficient mice, demonstrating the redundant activity of IL-1α and IL-1ß for skin inflammation. NLRP3 or apoptosis-associated Speck-like protein containing a Caspase recruitment domain-deficient mice had no significant reduction of skin inflammation in response to IMQ treatment, mainly due to the redundancy of IL-1α. However, IMQ-induced skin inflammation was abolished in the absence of MyD88, the adaptor protein shared by IL-1R and TLR signaling pathways. These results are consistent with the TLR7 dependence of IMQ-induced skin inflammation. Thus, IL-1R1 contributes to the IMQ-induced skin inflammation, and disruption of MyD88 signaling completely abrogates this response.


Subject(s)
Adjuvants, Immunologic/adverse effects , Aminoquinolines/adverse effects , Carrier Proteins/immunology , Drug Eruptions/immunology , Inflammasomes/immunology , Myeloid Differentiation Factor 88/immunology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Animals , Carrier Proteins/genetics , Cytokines/genetics , Cytokines/immunology , Drug Eruptions/genetics , Drug Eruptions/pathology , Imiquimod , Inflammasomes/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/immunology , Skin/pathology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology
18.
J Biol Chem ; 288(26): 18789-802, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23625922

ABSTRACT

Molecules that simultaneously inhibit independent or co-dependent proinflammatory pathways may have advantages over conventional monotherapeutics. OmCI is a bifunctional protein derived from blood-feeding ticks that specifically prevents complement (C)-mediated C5 activation and also sequesters leukotriene B4 (LTB4) within an internal binding pocket. Here, we examined the effect of LTB4 binding on OmCI structure and function and investigated the relative importance of C-mediated C5 activation and LTB4 in a mouse model of immune complex-induced acute lung injury (IC-ALI). We describe two crystal structures of bacterially expressed OmCI: one binding a C16 fatty acid and the other binding LTB4 (C20). We show that the C5 and LTB4 binding activities of the molecule are independent of each other and that OmCI is a potent inhibitor of experimental IC-ALI, equally dependent on both C5 inhibition and LTB4 binding for full activity. The data highlight the importance of LTB4 in IC-ALI and activation of C5 by the complement pathway C5 convertase rather than by non-C proteases. The findings suggest that dual inhibition of C5 and LTB4 may be useful for treatment of human immune complex-dependent diseases.


Subject(s)
Acute Lung Injury/metabolism , Antigen-Antibody Complex/pharmacology , Arthropod Proteins/pharmacology , Carrier Proteins/pharmacology , Lipocalins/pharmacology , Acute Lung Injury/immunology , Acute Lung Injury/therapy , Animals , Arthropod Proteins/metabolism , Carrier Proteins/metabolism , Chromatography, Gas , Complement C5/metabolism , Eicosanoids/metabolism , Fatty Acids/metabolism , Immunoenzyme Techniques , Leukotriene B4/metabolism , Lipocalins/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Sheep , Surface Plasmon Resonance , Thrombin/metabolism
19.
Eur J Immunol ; 43(10): 2683-95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23780878

ABSTRACT

Cerebral malaria is a severe complication of Plasmodium falciparum infection. Although T-cell activation and type II IFN-γ are required for Plasmodium berghei ANKA (PbA)-induced murine experimental cerebral malaria (ECM), the role of type I IFN-α/ß in ECM development remains unclear. Here, we address the role of the IFN-α/ß pathway in ECM devel-opment in response to hepatic or blood-stage PbA infection, using mice deficient for types I or II IFN receptors. While IFN-γR1⁻/⁻ mice were fully resistant, IFNAR1⁻/⁻ mice showed delayed and partial protection to ECM after PbA infection. ECM resistance in IFN-γR1⁻/⁻ mice correlated with unaltered cerebral microcirculation and absence of ischemia, while WT and IFNAR1⁻/⁻ mice developed distinct microvascular pathologies. ECM resistance appeared to be independent of parasitemia. Instead, key mediators of ECM were attenuated in the absence of IFNAR1, including PbA-induced brain sequestration of CXCR3⁺-activated CD8⁺ T cells. This was associated with reduced expression of Granzyme B, IFN-γ, IL-12Rß2, and T-cell-attracting chemokines CXCL9 and CXCL10 in IFNAR1⁻/⁻ mice, more so in the absence of IFN-γR1. Therefore, the type I IFN-α/ß receptor pathway contributes to brain T-cell responses and microvascular pathology, although it is not as essential as IFN-γ for the development of cerebral malaria upon hepatic or blood-stage PbA infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cerebellum/immunology , Interferon Type I/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Plasmodium falciparum/immunology , Animals , CD8-Positive T-Lymphocytes/parasitology , Cell Movement/genetics , Cerebellum/parasitology , Cytotoxicity, Immunologic/genetics , Disease Progression , Humans , Ischemia/genetics , Malaria, Cerebral/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/genetics , Models, Animal , Receptors, CXCR3/metabolism , Receptors, Interferon/genetics , Sporozoites/immunology
SELECTION OF CITATIONS
SEARCH DETAIL