Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(6): 626-35, 2016 06.
Article in English | MEDLINE | ID: mdl-27111143

ABSTRACT

Innate lymphoid cells (ILCs) are critical mediators of mucosal immunity, and group 1 ILCs (ILC1 cells) and group 3 ILCs (ILC3 cells) have been shown to be functionally plastic. Here we found that group 2 ILCs (ILC2 cells) also exhibited phenotypic plasticity in response to infectious or noxious agents, characterized by substantially lower expression of the transcription factor GATA-3 and a concomitant switch to being ILC1 cells that produced interferon-γ (IFN-γ). Interleukin 12 (IL-12) and IL-18 regulated this conversion, and during viral infection, ILC2 cells clustered within inflamed areas and acquired an ILC1-like phenotype. Mechanistically, these ILC1 cells augmented virus-induced inflammation in a manner dependent on the transcription factor T-bet. Notably, IL-12 converted human ILC2 cells into ILC1 cells, and the frequency of ILC1 cells in patients with chronic obstructive pulmonary disease (COPD) correlated with disease severity and susceptibility to exacerbations. Thus, functional plasticity of ILC2 cells exacerbates anti-viral immunity, which may have adverse consequences in respiratory diseases such as COPD.


Subject(s)
Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Influenza A virus/immunology , Lung/immunology , Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Aged , Animals , Cell Differentiation , Cell Plasticity/immunology , Cells, Cultured , Cytokines/metabolism , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , Smoking/adverse effects , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
2.
Nat Immunol ; 17(2): 196-203, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26692173

ABSTRACT

Canonically, immunoglobulin E (IgE) mediates allergic immune responses by triggering mast cells and basophils to release histamine and type 2 helper cytokines. Here we found that in human systemic lupus erythematosus (SLE), IgE antibodies specific for double-stranded DNA (dsDNA) activated plasmacytoid dendritic cells (pDCs), a type of cell of the immune system linked to viral defense, which led to the secretion of substantial amounts of interferon-α (IFN-α). The concentration of dsDNA-specific IgE found in patient serum correlated with disease severity and greatly potentiated pDC function by triggering phagocytosis via the high-affinity FcɛRI receptor for IgE, followed by Toll-like receptor 9 (TLR9)-mediated sensing of DNA in phagosomes. Our findings expand the known pathogenic mechanisms of IgE-mediated inflammation beyond those found in allergy and demonstrate that IgE can trigger interferon responses capable of exacerbating self-destructive autoimmune responses.


Subject(s)
Autoantibodies/immunology , Autoimmunity , Immunoglobulin E/immunology , Interferons/metabolism , Antibodies, Antinuclear/immunology , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines/blood , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunoglobulin G/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Male , Phagocytosis/immunology , Phagosomes/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Toll-Like Receptor 9/metabolism
3.
Stem Cells ; 42(3): 230-250, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38183264

ABSTRACT

Chronic inflammation and dysregulated repair mechanisms after epithelial damage have been implicated in chronic obstructive pulmonary disease (COPD). However, the lack of ex vivo-models that accurately reflect multicellular lung tissue hinders our understanding of epithelial-mesenchymal interactions in COPD. Through a combination of transcriptomic and proteomic approaches applied to a sophisticated in vitro iPSC-alveolosphere with fibroblasts model, epithelial-mesenchymal crosstalk was explored in COPD and following SARS-CoV-2 infection. These experiments profiled dynamic changes at single-cell level of the SARS-CoV-2-infected alveolar niche that unveiled the complexity of aberrant inflammatory responses, mitochondrial dysfunction, and cell death in COPD, which provides deeper insights into the accentuated tissue damage/inflammation/remodeling observed in patients with SARS-CoV-2 infection. Importantly, this 3D system allowed for the evaluation of ACE2-neutralizing antibodies and confirmed the potency of this therapy to prevent SARS-CoV-2 infection in the alveolar niche. Thus, iPSC-alveolosphere cultured with fibroblasts provides a promising model to investigate disease-specific mechanisms and to develop novel therapeutics.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Pulmonary Disease, Chronic Obstructive , Humans , SARS-CoV-2 , Proteomics , Immunotherapy , Inflammation
4.
Immunity ; 42(3): 566-79, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25786179

ABSTRACT

Cigarette smoking is a major risk factor for chronic obstructive pulmonary disease and is presumed to be central to the altered responsiveness to recurrent infection in these patients. We examined the effects of smoke priming underlying the exacerbated response to viral infection in mice. Lack of interleukin-33 (IL-33) signaling conferred complete protection during exacerbation and prevented enhanced inflammation and exaggerated weight loss. Mechanistically, smoke was required to upregulate epithelial-derived IL-33 and simultaneously alter the distribution of the IL-33 receptor ST2. Specifically, smoke decreased ST2 expression on group 2 innate lymphoid cells (ILC2s) while elevating ST2 expression on macrophages and natural killer (NK) cells, thus altering IL-33 responsiveness within the lung. Consequently, upon infection and release, increased local IL-33 significantly amplified type I proinflammatory responses via synergistic modulation of macrophage and NK cell function. Therefore, in COPD, smoke alters the lung microenvironment to facilitate an alternative IL-33-dependent exaggerated proinflammatory response to infection, exacerbating disease.


Subject(s)
Immunity, Innate/drug effects , Interleukins/immunology , Orthomyxoviridae Infections/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Interleukin/immunology , Smoke/adverse effects , Animals , Female , Gene Expression Regulation , Humans , Influenza A virus/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/deficiency , Interleukins/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/pathology , Macrophages/immunology , Macrophages/pathology , Mice, Transgenic , Orthomyxoviridae Infections/etiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , Nicotiana/chemistry , Weight Loss
5.
Eur Respir J ; 62(3)2023 09.
Article in English | MEDLINE | ID: mdl-37442582

ABSTRACT

BACKGROUND: Epithelial damage, repair and remodelling are critical features of chronic airway diseases including chronic obstructive pulmonary disease (COPD). Interleukin (IL)-33 released from damaged airway epithelia causes inflammation via its receptor, serum stimulation-2 (ST2). Oxidation of IL-33 to a non-ST2-binding form (IL-33ox) is thought to limit its activity. We investigated whether IL-33ox has functional activities that are independent of ST2 in the airway epithelium. METHODS: In vitro epithelial damage assays and three-dimensional, air-liquid interface (ALI) cell culture models of healthy and COPD epithelia were used to elucidate the functional role of IL-33ox. Transcriptomic changes occurring in healthy ALI cultures treated with IL-33ox and COPD ALI cultures treated with an IL-33-neutralising antibody were assessed with bulk and single-cell RNA sequencing analysis. RESULTS: We demonstrate that IL-33ox forms a complex with receptor for advanced glycation end products (RAGE) and epidermal growth factor receptor (EGFR) expressed on airway epithelium. Activation of this alternative, ST2-independent pathway impaired epithelial wound closure and induced airway epithelial remodelling in vitro. IL-33ox increased the proportion of mucus-producing cells and reduced epithelial defence functions, mimicking pathogenic traits of COPD. Neutralisation of the IL-33ox pathway reversed these deleterious traits in COPD epithelia. Gene signatures defining the pathogenic effects of IL-33ox were enriched in airway epithelia from patients with severe COPD. CONCLUSIONS: Our study reveals for the first time that IL-33, RAGE and EGFR act together in an ST2-independent pathway in the airway epithelium and govern abnormal epithelial remodelling and muco-obstructive features in COPD.


Subject(s)
Interleukin-33 , Pulmonary Disease, Chronic Obstructive , Humans , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33/genetics , Interleukin-33/metabolism , Oxidation-Reduction , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Receptor for Advanced Glycation End Products/metabolism
7.
N Engl J Med ; 380(14): 1336-1346, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30943337

ABSTRACT

BACKGROUND: Hypereosinophilic syndrome is a group of diseases defined by marked eosinophilia in blood or tissue and eosinophil-related clinical manifestations. Benralizumab is a monoclonal antibody against interleukin-5 receptor α, which is expressed on human eosinophils. METHODS: In this randomized, double-blind, placebo-controlled, phase 2 trial, we administered a series of three monthly subcutaneous injections of either benralizumab (at a dose of 30 mg) or placebo in 20 symptomatic patients who had PDGFRA-negative hypereosinophilic syndrome and an absolute eosinophil count of at least 1000 cells per cubic millimeter; all the patients were receiving stable therapy (drugs or dietary changes) for this disease. This regimen was followed by an open-label phase, during which the patient's background therapy could be tapered as tolerated, and an extension phase. The primary end point of the randomized phase was a reduction of at least 50% in the absolute eosinophil count at week 12. RESULTS: During the randomized phase, the primary end point occurred in more patients in the benralizumab group than in the placebo group (9 of 10 patients [90%] vs. 3 of 10 patients [30%], P = 0.02). During the open-label phase, clinical and hematologic responses were observed in 17 of 19 patients (89%) and were sustained for 48 weeks in 14 of 19 patients (74%); in the latter group, in 9 of 14 patients (64%), background therapies could be tapered. Bone marrow and tissue eosinophilia were also suppressed with benralizumab therapy. The most common drug-related adverse events, headache and an elevated lactate dehydrogenase level, occurred in 32% of the patients after the first dose of benralizumab and resolved within 48 hours in all patients. Other adverse events occurred with similar frequency in the two groups. Of the many potential predictors of response that were examined, only clinical disease subtype appeared to be associated with the initial response or relapse. CONCLUSIONS: In this small phase 2 trial, patients with PDGFRA-negative hypereosinophilic syndrome who received benralizumab for 12 weeks had lower absolute eosinophil counts than those who received placebo. During the open-label phase, clinical and hematologic responses were sustained for 48 weeks in 74% of the patients. Adverse events did not limit treatment. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov numbers, NCT00001406 and NCT02130882.).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Hypereosinophilic Syndrome/drug therapy , Interleukin-5 Receptor alpha Subunit/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Biopsy , Bone Marrow/immunology , Bone Marrow/pathology , Colon, Ascending/pathology , Double-Blind Method , Eosinophils , Female , Humans , Hypereosinophilic Syndrome/pathology , Injections, Subcutaneous , Leukocyte Count , Male , Middle Aged , Receptor, Platelet-Derived Growth Factor alpha/blood , Skin/pathology , Stomach/pathology
8.
Eur Respir J ; 59(3)2022 03.
Article in English | MEDLINE | ID: mdl-34289975

ABSTRACT

BACKGROUND: Benralizumab is a humanised, anti-interleukin-5 receptor α monoclonal antibody with anti-eosinophilic activity. Lack of fucose (afucosylation) increases its affinity to CD16a and significantly enhances antibody-dependent cell-mediated cytotoxicity by natural killer (NK) cells. Although benralizumab proved clinically efficacious in clinical trials for patients with severe asthma and hypereosinophilic syndrome, in-depth characterisation of its anti-eosinophilic mechanisms of action remains elusive. METHODS: Here, we further investigated the mechanisms involved in benralizumab's anti-eosinophilic activities by employing relevant primary human autologous cell co-cultures and real-time-lapse imaging combined with flow cytometry. RESULTS: In the presence of NK cells, benralizumab induced potent eosinophil apoptosis as demonstrated by the upstream induction of Caspase-3/7 and upregulation of cytochrome c. In addition, we uncovered a previously unrecognised mechanism whereby benralizumab can induce eosinophil phagocytosis/efferocytosis by macrophages, a process called antibody-dependent cellular phagocytosis. Using live cell imaging, we unravelled the stepwise processes leading to eosinophil apoptosis and uptake by activated macrophages. Through careful observations of cellular co-culture assays, we identified a novel role for macrophage-derived tumour necrosis factor (TNF) to further enhance benralizumab-mediated eosinophil apoptosis through activation of TNF receptor 1 on eosinophils. TNF-induced eosinophil apoptosis was associated with cytochrome c upregulation, mitochondrial membrane depolarisation and increased Caspase-3/7 activity. Moreover, activated NK cells were found to amplify this axis through the secretion of interferon-γ, subsequently driving TNF expression by macrophages. CONCLUSIONS: Our data provide deeper insights into the timely appearance of events leading to benralizumab-induced eosinophil apoptosis and suggest that additional mechanisms may contribute to the potent anti-eosinophilic activity of benralizumab in vivo. Importantly, afucosylation of benralizumab strongly enhanced its potency for all mechanisms investigated.


Subject(s)
Anti-Asthmatic Agents , Asthma , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Eosinophils , Humans
9.
Immunity ; 37(6): 986-997, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23219390

ABSTRACT

Toll-like receptor-9 (TLR9) is largely responsible for discriminating self from pathogenic DNA. However, association of host DNA with autoantibodies activates TLR9, inducing the pathogenic secretion of type I interferons (IFNs) from plasmacytoid dendritic cells (pDCs). Here, we found that in response to DNA-containing immune complexes (DNA-IC), but not to soluble ligands, IFN-α production depended upon the convergence of the phagocytic and autophagic pathways, a process called microtubule-associated protein 1A/1B-light chain 3 (LC3)-associated phagocytosis (LAP). LAP was required for TLR9 trafficking into a specialized interferon signaling compartment by a mechanism that involved autophagy-related proteins, but not the conventional autophagic preinitiation complex, or adaptor protein-3 (AP-3). Our findings unveil a new role for nonconventional autophagy in inflammation and provide one mechanism by which anti-DNA autoantibodies, such as those found in several autoimmune disorders, bypass the controls that normally restrict the apportionment of pathogenic DNA and TLR9 to the interferon signaling compartment.


Subject(s)
Antigen-Antibody Complex/immunology , Autophagy/immunology , DNA/immunology , Interferon Type I/biosynthesis , Animals , Humans , Immunoglobulin G/immunology , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Phagocytosis/immunology , Phagosomes/metabolism , Protein Transport , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
10.
J Physiol ; 598(23): 5541-5554, 2020 12.
Article in English | MEDLINE | ID: mdl-32924209

ABSTRACT

KEY POINTS: Type I interferon receptors are expressed by the majority of vagal C-fibre neurons innervating the respiratory tract Interferon alpha and beta acutely and directly activate vagal C-fibers in the airways. The interferon-induced activation of C-fibers occurs secondary to stimulation of type 1 interferon receptors Type 1 interferons may contribute to the symptoms as well as the spread of respiratory viral infections by causing coughing and other defensive reflexes associated with vagal C-fibre activation ABSTRACT: We evaluated the ability of type I interferons to acutely activate airway vagal afferent nerve terminals in mouse lungs. Using single cell RT-PCR of lung-specific vagal neurons we found that IFNAR1 and IFNAR2 were expressed in 70% of the TRPV1-positive neurons (a marker for vagal C-fibre neurons) and 44% of TRPV1-negative neurons. We employed an ex vivo vagal innervated mouse trachea-lung preparation to evaluate the effect of interferons in directly activating airway nerves. Utilizing 2-photon microscopy of the nodose ganglion neurons from Pirt-Cre;R26-GCaMP6s mice we found that applying IFNα or IFNß to the lungs acutely activated the majority of vagal afferent nerve terminals. When the type 1 interferon receptor, IFNAR1, was blocked with a blocking antibody the response to IFNß was largely inhibited. The type 2 interferon, IFNγ, also activated airway nerves and this was not inhibited by the IFNAR1 blocking antibody. The Janus kinase inhibitor GLPG0634 (1 µm) virtually abolished the nerve activation caused by IFNß. Consistent with the activation of vagal afferent C-fibers, infusing IFNß into the mouse trachea led to defensive breathing reflexes including apneas and gasping. These reflexes were prevented by pretreatment with an IFN type-1 receptor blocking antibody. Finally, using whole cell patch-clamp electrophysiology of lung-specific neurons we found that IFNß (1000 U ml-1 ) directly depolarized the membrane potential of isolated nodose neurons, in some cases beyond to action potential threshold. This acute non-genomic activation of vagal sensory nerve terminals by interferons may contribute to the incessant coughing that is a hallmark of respiratory viral infections.


Subject(s)
Interferon Type I , Nociceptors , Animals , Bronchi , Mice , Neurons, Afferent , Nodose Ganglion , Vagus Nerve
11.
Eur Respir J ; 55(5)2020 05.
Article in English | MEDLINE | ID: mdl-32060064

ABSTRACT

Although elevated blood or sputum eosinophils are present in many patients with COPD, uncertainties remain regarding the anatomical distribution pattern of lung-infiltrating eosinophils. Basophils have remained virtually unexplored in COPD. This study mapped tissue-infiltrating eosinophils, basophils and eosinophil-promoting immune mechanisms in COPD-affected lungs.Surgical lung tissue and biopsies from major anatomical compartments were obtained from COPD patients with severity grades Global Initiative for Chronic Obstructive Lung Disease stages I-IV; never-smokers/smokers served as controls. Automated immunohistochemistry and in situ hybridisation identified immune cells, the type 2 immunity marker GATA3 and eotaxins (CCL11, CCL24).Eosinophils and basophils were present in all anatomical compartments of COPD-affected lungs and increased significantly in very severe COPD. The eosinophilia was strikingly patchy, and focal eosinophil-rich microenvironments were spatially linked with GATA3+ cells, including type 2 helper T-cell lymphocytes and type 2 innate lymphoid cells. A similarly localised and interleukin-33/ST2-dependent eosinophilia was demonstrated in influenza-infected mice. Both mice and patients displayed spatially confined eotaxin signatures with CCL11+ fibroblasts and CCL24+ macrophages.In addition to identifying tissue basophilia as a novel feature of advanced COPD, the identification of spatially confined eosinophil-rich type 2 microenvironments represents a novel type of heterogeneity in the immunopathology of COPD that is likely to have implications for personalised treatment.


Subject(s)
Basophils/immunology , Eosinophils/immunology , Macrophages/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Eosinophilia/etiology , Adult , Aged , Animals , Biomarkers , Chemokine CCL11/immunology , Chemokine CCL24/immunology , Female , GATA3 Transcription Factor/immunology , Humans , Immunity, Innate , Male , Mice , Middle Aged , Smokers , Young Adult
12.
Mediators Inflamm ; 2020: 4087315, 2020.
Article in English | MEDLINE | ID: mdl-33376451

ABSTRACT

The interleukin-1 family member IL-33 participates in both innate and adaptive T helper-2 immune cell responses in models of lung disease. The IL-6-type cytokine Oncostatin M (OSM) elevates lung inflammation, Th2-skewed cytokines, alternatively activated (M2) macrophages, and eosinophils in C57Bl/6 mice in vivo. Since OSM induces IL-33 expression, we here test the IL-33 function in OSM-mediated lung inflammation using IL-33-/- mice. Adenoviral OSM (AdOSM) markedly induced IL-33 mRNA and protein levels in wild-type animals while IL-33 was undetectable in IL-33-/- animals. AdOSM treatment showed recruitment of neutrophils, eosinophils, and elevated inflammatory chemokines (KC, eotaxin-1, MIP1a, and MIP1b), Th2 cytokines (IL-4/IL-5), and arginase-1 (M2 macrophage marker) whereas these responses were markedly diminished in IL-33-/- mice. AdOSM-induced IL-33 was unaffected by IL-6-/- deficiency. AdOSM also induced IL-33R+ ILC2 cells in the lung, while IL-6 (AdIL-6) overexpression did not. Flow-sorted ILC2 responded in vitro to IL-33 (but not OSM or IL-6 stimulation). Matrix remodelling genes col3A1, MMP-13, and TIMP-1 were also decreased in IL-33-/- mice. In vitro, IL-33 upregulated expression of OSM in the RAW264.7 macrophage cell line and in bone marrow-derived macrophages. Taken together, IL-33 is a critical mediator of OSM-driven, Th2-skewed, and M2-like responses in mouse lung inflammation and contributes in part through activation of ILC2 cells.


Subject(s)
Interleukin-33/physiology , Oncostatin M/physiology , Pneumonia/etiology , Animals , Female , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Th2 Cells/immunology
13.
Ann Rheum Dis ; 78(7): 957-966, 2019 07.
Article in English | MEDLINE | ID: mdl-31040119

ABSTRACT

OBJECTIVES: The presence of proinflammatory low-density granulocytes (LDG) has been demonstrated in autoimmune and infectious diseases. Recently, regulatory neutrophilic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) were identified in systemic lupus erythematosus (SLE). Because LDG and PMN-MDSC share a similar phenotype with contrasting functional effects, we explored these cells in a cohort of patients with SLE. METHODS: LDG and normal-density granulocytes (NDG) were isolated from fresh blood of healthy donors (HD) and patients with SLE. Associations between LDG and clinical manifestations were analysed. Multicolor flow cytometry and confocal imaging were performed to immunophenotype the cells. The ability of LDG and NDG to suppress T cell function and induce cytokine production was quantified. RESULTS: LDG prevalence was elevated in SLE versus HD, associated with the interferon (IFN) 21-gene signature and disease activity. Also, the LDG-to-lymphocyte ratio associated better with SLE disease activity index than neutrophil-to-lymphocyte ratio. SLE LDG exhibited significantly heightened surface expression of various activation markers and also of lectin-like oxidised low-density lipoprotein receptor-1, previously described to be associated with PMN-MDSC. Supernatants from SLE LDG did not restrict HD CD4+ T cell proliferation in an arginase-dependent manner, suggesting LDG are not immunosuppressive. SLE LDG supernatants induced proinflammatory cytokine production (IFN gamma, tumour necrosis factor alpha and lymphotoxin alpha) from CD4+ T cells. CONCLUSIONS: Based on our results, SLE LDG display an activated phenotype, exert proinflammatory effects on T cells and do not exhibit MDSC function. These results support the concept that LDG represent a distinct proinflammatory subset in SLE with pathogenic potential, at least in part, through their ability to activate type 1 helper responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Granulocytes/immunology , Lupus Erythematosus, Systemic/immunology , Neutrophils/immunology , Adolescent , Adult , Aged , Case-Control Studies , Cell Proliferation , Female , Flow Cytometry , Humans , Immunophenotyping , Lupus Erythematosus, Systemic/blood , Lymphocyte Activation , Male , Middle Aged , Phenotype , Young Adult
14.
Ann Rheum Dis ; 78(2): 228-237, 2019 02.
Article in English | MEDLINE | ID: mdl-30459279

ABSTRACT

OBJECTIVE: Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS: VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS: We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS: VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigen-Antibody Complex/drug effects , Autoimmune Diseases/drug therapy , Immunologic Factors/pharmacology , Receptors, IgG/immunology , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antigen-Antibody Complex/immunology , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Humans , Immunoglobulin G/immunology , Interleukin-6/immunology , Macaca fascicularis , Mice , Mice, Transgenic , Neutrophils/immunology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology
15.
J Immunol ; 199(9): 3316-3325, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28939759

ABSTRACT

Dysfunction of the epithelial barrier is a hallmark of inflammatory intestinal diseases. The intestinal epithelial barrier is maintained by expression of tight junctions that connect adjacent epithelial cells and seal the paracellular space. IL-22 is critical for the maintenance of intestinal barrier function through promoting antipathogen responses and regeneration of epithelial tissues in the gut. However, little is known about the effects of IL-22 on the regulation of tight junctions in the intestinal epithelium. In this study we report that IL-22 signals exclusively through the basolateral side of polarized Caco-2 cell monolayers. IL-22 treatment does not affect the flux of uncharged macromolecules across cell monolayers but significantly reduces transepithelial electrical resistance (TEER), indicating an increase of paracellular permeability for ions. IL-22 treatment on Caco-2 monolayers and on primary human intestinal epithelium markedly induces the expression of Claudin-2, a cation-channel-forming tight junction protein. Furthermore, treatment of IL-22 in mice upregulates Claudin-2 protein in colonic epithelial cells. Knocking down Claudin-2 expression with small interfering RNA reverses the reduction of TEER in IL-22-treated cells. Moreover, IL-22-mediated upregulation of Claudin-2 and loss of TEER can be suppressed with the treatment of JAK inhibitors. In summary, our results reveal that IL-22 increases intestinal epithelial permeability by upregulating Claudin-2 expression through the JAK/STAT pathway. These results provide novel mechanistic insights into the role of IL-22 in the regulation and maintenance of the intestinal epithelial barrier.


Subject(s)
Claudins/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Signal Transduction/immunology , Tight Junctions/immunology , Up-Regulation/immunology , Animals , Caco-2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Permeability , Interleukin-22
16.
J Immunol ; 197(10): 4118-4126, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798148

ABSTRACT

Nucleic acid recognition is an important mechanism that enables the innate immune system to detect microbial infection and tissue damage. To minimize the recognition of self-derived nucleic acids, all nucleic acid-sensing signaling receptors are sequestered away from the cell surface and are activated in the cytoplasm or in endosomes. Nucleic acid sensing in endosomes relies on members of the TLR family. The receptor for advanced glycation end-products (RAGE) was recently shown to bind DNA at the cell surface, facilitating DNA internalization and subsequent recognition by TLR9. In this article, we show that RAGE binds RNA molecules in a sequence-independent manner and enhances cellular RNA uptake into endosomes. Gain- and loss-of-function studies demonstrate that RAGE increases the sensitivity of all ssRNA-sensing TLRs (TLR7, TLR8, TLR13), suggesting that RAGE is an integral part of the endosomal nucleic acid-sensing system.


Subject(s)
Endosomes/metabolism , RNA/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Toll-Like Receptors/immunology , DNA/genetics , DNA/metabolism , HEK293 Cells , Humans , Immunity, Innate , Polymerase Chain Reaction , RNA/genetics , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/immunology , Toll-Like Receptor 8/metabolism
17.
J Allergy Clin Immunol ; 140(6): 1604-1615.e5, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28216433

ABSTRACT

BACKGROUND: A number of food allergies (eg, fish, shellfish, and nuts) are lifelong, without any disease-transforming therapies, and unclear in their underlying immunology. Clinical manifestations of food allergy are largely mediated by IgE. Although persistent IgE titers have been attributed conventionally to long-lived IgE+ plasma cells (PCs), this has not been directly and comprehensively tested. OBJECTIVE: We sought to evaluate mechanisms underlying persistent IgE and allergic responses to food allergens. METHODS: We used a model of peanut allergy and anaphylaxis, various knockout mice, adoptive transfer experiments, and in vitro assays to identify mechanisms underlying persistent IgE humoral immunity over almost the entire lifespan of the mouse (18-20 months). RESULTS: Contrary to conventional paradigms, our data show that clinically relevant lifelong IgE titers are not sustained by long-lived IgE+ PCs. Instead, lifelong reactivity is conferred by allergen-specific long-lived memory B cells that replenish the IgE+ PC compartment. B-cell reactivation requires allergen re-exposure and IL-4 production by CD4 T cells. We define the half-lives of antigen-specific germinal centers (23.3 days), IgE+ and IgG1+ PCs (60 and 234.4 days, respectively), and clinically relevant cell-bound IgE (67.3 days). CONCLUSIONS: These findings can explain lifelong food allergies observed in human subjects as the consequence of allergen exposures that recurrently activate memory B cells and identify these as a therapeutic target with disease-transforming potential.


Subject(s)
Anaphylaxis/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Food Hypersensitivity/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Arachis/immunology , Cells, Cultured , Humans , Immunity, Humoral , Immunoglobulin E/metabolism , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
J Allergy Clin Immunol ; 137(6): 1651-1661, 2016 06.
Article in English | MEDLINE | ID: mdl-27264000

ABSTRACT

There is accumulating evidence to suggest that IgE plays a significant role in autoimmunity. The presence of circulating self-reactive IgE in patients with autoimmune disorders has been long known but, at the same time, largely understudied. However, studies have shown that the increased IgE concentration is not associated with higher prevalence for atopy and allergy in patients with autoimmune diseases, such as systemic lupus erythematosus. IgE-mediated mechanisms are conventionally known to facilitate degranulation of mast cells and basophils and promote TH2 immunity, mechanisms that are not only central to mounting an appropriate defense against parasitic worms, noxious substances, toxins, venoms, and environmental irritants but that also trigger exuberant allergic reactions in patients with allergies. More recently, IgE autoantibodies have been recognized to participate in the self-inflicted damaging immune responses that characterize autoimmunity. Such autoimmune responses include direct damage on tissue-containing autoantigens, activation and migration of basophils to lymph nodes, and, as observed most recently, induction of type 1 interferon responses from plasmacytoid dendritic cells. The importance of IgE as a central pathogenic mechanism in autoimmunity has now been clinically validated by the approval of omalizumab, an anti-IgE mAb, for patients with chronic spontaneous urticaria and for the clinical benefit of patients with bullous pemphigoid. In this review we summarize recent reports describing the prevalence of self-reactive IgE and discuss novel findings that incriminate IgE as central in the pathogenesis of inflammatory autoimmune disorders.


Subject(s)
Autoimmunity , Immunoglobulin E/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/metabolism , Urticaria/immunology , Urticaria/metabolism
19.
J Immunol ; 193(6): 3134-45, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25092891

ABSTRACT

Cigarette smoke has a broad impact on the mucosal environment with the ability to alter host defense mechanisms. Within the context of a bacterial infection, this altered host response is often accompanied by exacerbated cellular inflammation, characterized by increased neutrophilia. The current study investigated the mechanisms of neutrophil recruitment in a murine model of cigarette smoke exposure and, subsequently, a model of both cigarette smoke exposure and bacterial infection. We investigated the role of IL-1 signaling in neutrophil recruitment and found that cigarette smoke-induced neutrophilia was dependent on IL-1α produced by alveolar macrophages. In addition to being the crucial source of IL-1α, alveolar macrophages isolated from smoke-exposed mice were primed for excessive IL-1α production in response to bacterial ligands. To test the relevance of exaggerated IL-1α production in neutrophil recruitment, a model of cigarette smoke exposure and nontypeable Haemophilus influenzae infection was developed. Mice exposed to cigarette smoke elaborated an exacerbated CXCR2-dependent neutrophilia in response to nontypeable Haemophilus influenzae. Exacerbated neutrophilia was dependent on IL-1α priming of the pulmonary environment by cigarette smoke as exaggerated neutrophilia was dependent on IL-1 signaling. These data characterize a novel mechanism of cigarette smoke priming the lung mucosa toward greater IL-1-driven neutrophilic responses to bacteria, with a central role for the alveolar macrophage in this process.


Subject(s)
Haemophilus influenzae/immunology , Interleukin-1alpha/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Receptors, Interleukin-8B/immunology , Smoke/adverse effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Chemokine CXCL1/biosynthesis , Chemokine CXCL5/biosynthesis , Chemokine CXCL5/genetics , Chemokine CXCL5/immunology , Female , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Inflammation/immunology , Leukocyte Count , Lung/pathology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/biosynthesis , Receptors, Interleukin-8B/biosynthesis , Receptors, Interleukin-8B/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Nicotiana/adverse effects
20.
Eur J Immunol ; 44(12): 3669-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25229885

ABSTRACT

Anaphylatoxin C5a released upon complement activation is associated with both acute and chronic inflammations such as gout. The pathogenesis of gout was identified as uric acid crystal deposition in the joints that activates inflammasome, leading to IL-1ß release. However, little is known about the interaction between complement activation and monosodium urate/uric acid (MSU) crystal-induced inflammasome activation or IL-1ß production. Here, we report that MSU crystal-induced proinflammatory cytokines/chemokines in human whole blood is predominantly regulated by C5a through its interaction with C5a receptor. C5a induces pro-IL-1ß and IL-1ß production in human primary monocytes, and potentiates MSU or cholesterol crystals in IL-1ß production. This potentiation is caspase-1 dependent and requires intracellular Ca(2+) mobilization, K(+) efflux, and cathepsin B activity. Our results provide insight into the role of C5a as an endogenous priming signal that is required for the initiation of uric acid crystal-induced IL-1ß production. C5a could potentially be a therapeutic target together with IL-1ß antagonists for the treatment of complement-dependent and inflammasome-associated diseases.


Subject(s)
Antioxidants/pharmacology , Calcium Signaling/drug effects , Complement C5a/immunology , Interleukin-1beta/immunology , Monocytes/immunology , Uric Acid/pharmacology , Antioxidants/adverse effects , Calcium/immunology , Calcium Signaling/immunology , Caspase 1/immunology , Female , Humans , Inflammasomes/immunology , Male , Monocytes/pathology , Potassium/immunology , Uric Acid/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL