Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Elife ; 132024 May 09.
Article in English | MEDLINE | ID: mdl-38722677

ABSTRACT

Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 microRNA (Mirlet7 miRNA) family is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the Mirlet7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here, we show that overall expression of the Mirlet7 clusters, Mirlet7b/Mirlet7c2 and Mirlet7a1/Mirlet7f1/Mirlet7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the Mirlet7b/Mirlet7c2 cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the Mirlet7b/Mirlet7c2 cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing Mirlet7g in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of Mirlet7 in T cells. Overall, our findings shed light on the Mirlet7/RORγt axis with Mirlet7 acting as a molecular brake in the generation of Tc17 cells and suggest a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.


Subject(s)
Cell Differentiation , Down-Regulation , MicroRNAs , Nuclear Receptor Subfamily 1, Group F, Member 3 , Pulmonary Emphysema , Th17 Cells , Animals , Female , Humans , Male , Mice , Interleukin-17/metabolism , Interleukin-17/genetics , Lung/pathology , Lung/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Th17 Cells/immunology , Th17 Cells/metabolism
2.
bioRxiv ; 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-37905101

ABSTRACT

Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 family of miRNAs is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the let-7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here we show that overall expression of the let-7 miRNA clusters, let-7b/let-7c2 and let-7a1/let-7f1/let-7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the let-7b/let-7c2-cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the let-7b/let-7c2-cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing let-7 in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of let-7 miRNA in T cells. Overall, our findings shed light on the let-7/RORγt axis with let-7 acting as a molecular brake in the generation of Tc17 cells and suggests a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.

3.
Sci Adv ; 8(46): eabq0615, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36383649

ABSTRACT

Chronic exposure to airborne carbon black ultrafine (nCB) particles generated from incomplete combustion of organic matter drives IL-17A-dependent emphysema. However, whether and how they alter the immune responses to lung cancer remains unknown. Here, we show that exposure to nCB particles increased PD-L1+ PD-L2+ CD206+ antigen-presenting cells (APCs), exhausted T cells, and Treg cells. Lung macrophages that harbored nCB particles showed selective mitochondrial structure damage and decreased oxidative respiration. Lung macrophages sustained the HIF1α axis that increased glycolysis and lactate production, culminating in an immunosuppressive microenvironment in multiple mouse models of non-small cell lung cancers. Adoptive transfer of lung APCs from nCB-exposed wild type to susceptible mice increased tumor incidence and caused early metastasis. Our findings show that nCB exposure metabolically rewires lung macrophages to promote immunosuppression and accelerates the development of lung cancer.


Subject(s)
Lung Neoplasms , Soot , Mice , Animals , Soot/metabolism , Particulate Matter/adverse effects , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Macrophages , Lung/metabolism , Carbon/metabolism , Tumor Microenvironment
4.
J Clin Invest ; 129(10): 4290-4304, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31483291

ABSTRACT

Electronic nicotine delivery systems (ENDS) or e-cigarettes have emerged as a popular recreational tool among adolescents and adults. Although the use of ENDS is often promoted as a safer alternative to conventional cigarettes, few comprehensive studies have assessed the long-term effects of vaporized nicotine and its associated solvents, propylene glycol (PG) and vegetable glycerin (VG). Here, we show that compared with smoke exposure, mice receiving ENDS vapor for 4 months failed to develop pulmonary inflammation or emphysema. However, ENDS exposure, independent of nicotine, altered lung lipid homeostasis in alveolar macrophages and epithelial cells. Comprehensive lipidomic and structural analyses of the lungs revealed aberrant phospholipids in alveolar macrophages and increased surfactant-associated phospholipids in the airway. In addition to ENDS-induced lipid deposition, chronic ENDS vapor exposure downregulated innate immunity against viral pathogens in resident macrophages. Moreover, independent of nicotine, ENDS-exposed mice infected with influenza demonstrated enhanced lung inflammation and tissue damage. Together, our findings reveal that chronic e-cigarette vapor aberrantly alters the physiology of lung epithelial cells and resident immune cells and promotes poor response to infectious challenge. Notably, alterations in lipid homeostasis and immune impairment are independent of nicotine, thereby warranting more extensive investigations of the vehicle solvents used in e-cigarettes.


Subject(s)
Electronic Nicotine Delivery Systems , Immunity, Innate/drug effects , Lipid Metabolism/drug effects , Lung/drug effects , Lung/metabolism , Adolescent , Adult , Animals , Disease Models, Animal , Female , Homeostasis , Humans , Lipidomics , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nicotine/administration & dosage , Nicotine/adverse effects , Phospholipids/metabolism , Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Smoke/adverse effects , Solvents/administration & dosage , Solvents/adverse effects
5.
J Immunol ; 203(2): 349-359, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31182478

ABSTRACT

Loss of immune tolerance to self-antigens can promote chronic inflammation and disrupt the normal function of multiple organs, including the lungs. Degradation of elastin, a highly insoluble protein and a significant component of the lung structural matrix, generates proinflammatory molecules. Elastin fragments (EFs) have been detected in the serum of smokers with emphysema, and elastin-specific T cells have also been detected in the peripheral blood of smokers with emphysema. However, an animal model that could recapitulate T cell-specific autoimmune responses by initiating and sustaining inflammation in the lungs is lacking. In this study, we report an animal model of autoimmune emphysema mediated by the loss of tolerance to elastin. Mice immunized with a combination of human EFs plus rat EFs but not mouse EFs showed increased infiltration of innate and adaptive immune cells to the lungs and developed emphysema. We cloned and expanded mouse elastin-specific CD4+ T cells from the lung and spleen of immunized mice. Finally, we identified TCR sequences from the autoreactive T cell clones, suggesting possible pathogenic TCRs that can cause loss of immune tolerance against elastin. This new autoimmune model of emphysema provides a useful tool to examine the immunological factors that promote loss of immune tolerance to self.


Subject(s)
Autoimmunity/immunology , Elastin/immunology , Lung/immunology , Pulmonary Emphysema/immunology , Adaptive Immunity/immunology , Animals , Cell Line , Disease Models, Animal , Female , HEK293 Cells , Humans , Immune Tolerance/immunology , Immunity, Innate/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/immunology
6.
J Biol Chem ; 294(22): 8834-8847, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30992366

ABSTRACT

Proteinases are essential drivers of allergic airway disease and innate antifungal immunity in part through their ability cleave the clotting factor fibrinogen (FBG) into fibrinogen cleavage products (FCPs) that signal through Toll-like receptor 4 (TLR4). However, the mechanism by which FCPs engage TLR4 remains unknown. Here, we show that the proteinases from Aspergillus melleus (PAM) and other allergenic organisms rapidly hydrolyze FBG to yield relatively few FCPs that drive distinct antifungal mechanisms through TLR4. Functional FCPs, termed cryptokines, were characterized by rapid loss of the FBG α chain with substantial preservation of the ß and γ chains, including a γ chain sequence (Fibγ390-396) that binds the integrin Mac-1 (CD11b/CD18). PAM-derived cryptokines could be generated from multiple FBG domains, and the ability of cryptokines to induce fungistasis in vitro and innate allergic airway disease in vivo strongly depended on both Mac-1 and the Mac-1-binding domain of FBG (Fibγ390-396). Our findings illustrate the essential concept of proteinase-activated immune responses and for the first time link Mac-1, cryptokines, and TLR4 to innate antifungal immunity and allergic airway disease.


Subject(s)
Aspergillus/immunology , CD11b Antigen/metabolism , Fibrinogen/metabolism , Fungal Proteins/metabolism , Immunity, Innate , Peptide Hydrolases/metabolism , Animals , Aspergillus/enzymology , CD11b Antigen/deficiency , CD11b Antigen/genetics , Disease Models, Animal , Fibrinogen/chemistry , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism , Toll-Like Receptor 4/metabolism
7.
Nat Commun ; 10(1): 58, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30610193

ABSTRACT

Bloodborne infections with Candida albicans are an increasingly recognized complication of modern medicine. Here, we present a mouse model of low-grade candidemia to determine the effect of disseminated infection on cerebral function and relevant immune determinants. We show that intravenous injection of 25,000 C. albicans cells causes a highly localized cerebritis marked by the accumulation of activated microglial and astroglial cells around yeast aggregates, forming fungal-induced glial granulomas. Amyloid precursor protein accumulates within the periphery of these granulomas, while cleaved amyloid beta (Aß) peptides accumulate around the yeast cells. CNS-localized C. albicans further activate the transcription factor NF-κB and induce production of interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor (TNF), and Aß peptides enhance both phagocytic and antifungal activity from BV-2 cells. Mice infected with C. albicans display mild memory impairment that resolves with fungal clearance. Our results warrant additional studies to understand the effect of chronic cerebritis on cognitive and immune function.


Subject(s)
Candidemia/complications , Cerebrum/pathology , Memory Disorders/microbiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Astrocytes/metabolism , Astrocytes/microbiology , Astrocytes/pathology , Candida albicans , Candidemia/metabolism , Candidemia/pathology , Cerebrum/microbiology , Cerebrum/physiopathology , Interleukin-1beta/metabolism , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Microglia/metabolism , Microglia/microbiology , Microglia/pathology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha
8.
Ann Am Thorac Soc ; 15(Suppl 3): S198-S204, 2018 11.
Article in English | MEDLINE | ID: mdl-30431347

ABSTRACT

Asthma, chronic rhinosinusitis, and related incurable allergic afflictions of the upper and lower airways are medically important because of their association with the disabling symptom of dyspnea and, at least for asthma, the potential to cause fatal asphyxiation. Extensive research over the past two decades has uncovered both the physiological basis of airway obstruction in asthma and key governing molecular pathways. Exaggerated airway constriction in response to diverse provocative stimuli, termed airway hyperresponsiveness, is mediated through the cytokines interleukin 4 (IL-4) and IL-13 and the transcription factor signal transducer and activator of transcription 6 (STAT6). Overproduction of mucus has long been known to be an essential second component of airway obstruction and is also mediated in part through the IL-4/IL-13/STAT6 pathway. In this review, we discuss a second major signaling pathway which underlies mucus production that is mediated through proteinase-cleaved fibrinogen signaling through Toll-like receptor 4. Unexpectedly, our analysis of human sputum and paranasal sinus fluid indicates that in most cases of severe allergic airway disease, a unique type of airway fungal infection, termed airway mycosis, is pathogenically linked to these conditions. We further discuss how fungal and endogenous proteinases mediate the fibrinogenolysis that is essential to both Toll-like receptor 4 signaling and fibrin deposition that, together with mucus, contribute to airway obstruction.


Subject(s)
Lung Diseases, Obstructive/microbiology , Mycoses/etiology , Antifungal Agents/therapeutic use , Fibrinogen , Humans , Lung Diseases, Obstructive/diagnosis , Lung Diseases, Obstructive/therapy , Mucus , Mycoses/diagnosis , Mycoses/therapy , Peptide Hydrolases , Toll-Like Receptor 4
9.
Methods Mol Biol ; 1799: 1-9, 2018.
Article in English | MEDLINE | ID: mdl-29956139

ABSTRACT

Allergic airway diseases (asthma and chronic rhinosinusitis) are among the most common of all human diseases in heavily industrialized societies. Animal models of asthma have provided remarkable insight into allergic disease pathogenesis and will continue to drive the discovery of new therapeutic insights. We provide in this chapter a detailed protocol for inducing allergic immunity in the lungs of mice using a purified fungal protease and include related protocols for assessing immune endpoints.


Subject(s)
Allergens/immunology , Fungi/immunology , Hypersensitivity/etiology , Immunity , Lung/immunology , Peptide Hydrolases/immunology , Animals , Biomarkers , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Fungi/enzymology , Hypersensitivity/pathology , Lung/pathology , Mice , Plethysmography , Th2 Cells/immunology , Th2 Cells/metabolism
10.
Cancer Immunol Res ; 6(6): 645-657, 2018 06.
Article in English | MEDLINE | ID: mdl-29653981

ABSTRACT

Somatic mutations can promote malignant transformation of airway epithelial cells and induce inflammatory responses directed against resultant tumors. Tumor-infiltrating T lymphocytes (TIL) in early-stage non-small cell lung cancer (NSCLC) secrete distinct proinflammatory cytokines, but the contribution of these TILs to tumor development and metastasis remains unknown. We show here that TILs in early-stage NSCLC are biased toward IL17A expression (Th17) when compared with adjacent tumor-free tissue, whereas Th17 cells are decreased in tumor infiltrating locoregional lymph nodes in advanced NSCLC. Mice in which Pten and Smad4 (Pts4d/d ) are deleted from airway epithelial cells develop spontaneous tumors, that share genetic signatures with squamous- (SQ.2b), and adeno- (AD.1) subtypes of human NSCLC. Pts4d/d mice globally lacking in IL17a (Pts4d/dIl17a-/- ) showed decreased tumor latency and increased metastasis. Th17 cells were required for recruitment of CD103+ dendritic cells, and adoptive transfer of IL17a-sufficient CD4+ T cells reversed early tumor development and metastasis in Pts4d/dIl17a-/- mice. Together, these findings support a key role for Th17 cells in TILs associated with the Pts4d/d model of NSCLC and suggest therapeutic and biomarker strategies for human SQ2b and AD1 lung cancer. Cancer Immunol Res; 6(6); 645-57. ©2018 AACR.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Interleukin-17/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/genetics , Animals , Biomarkers , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/genetics , Cell Cycle/genetics , Disease Models, Animal , Disease Progression , Female , Genomics/methods , Humans , Immunohistochemistry , Immunophenotyping , Interleukin-17/genetics , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Staging , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology
11.
Semin Respir Crit Care Med ; 39(1): 64-81, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29427987

ABSTRACT

Allergic asthma is a heterogeneous disorder that defies a unanimously acceptable definition, but is generally recognized through its highly characteristic clinical expression of dyspnea and cough accompanied by clinical data that document reversible or exaggerated airway constriction and obstruction. The generally rising prevalence of asthma in highly industrialized societies despite significant therapeutic advances suggests that the fundamental cause(s) of asthma remain poorly understood. Detailed analyses of both the indoor (built) and outdoor environments continue to support the concept that not only inhaled particulates, especially carbon-based particulate pollution, pollens, and fungal elements, but also many noxious gases and chemicals, especially biologically derived byproducts such as proteinases, are essential to asthma pathogenesis. Phthalates, another common class of chemical pollutant found in the built environment, are emerging as potentially important mediators or attenuators of asthma. Other biological products such as endotoxin have also been confirmed to be protective in both the indoor and outdoor contexts. Proasthmatic factors are believed to activate, and in some instances initiate, pathologic inflammatory cascades through complex interactions with pattern recognition receptors (PRRs) expressed on many cell types, but especially airway epithelial cells. PRRs initiate the release of proallergic cytokines such as interleukin (IL)-33, IL-25, and others that coordinate activation of innate lymphoid cells type 2 (ILC2), T helper type 2 cells, and immunoglobulin E-secreting B cells that together promote additional inflammation and the major airway remodeling events (airway hyperresponsiveness, mucus hypersecretion) that promote airway obstruction. Proteinases, with airway fungi and viruses being potentially important sources, are emerging as critically important initiators of these inflammatory cascades in part through their effects on clotting factors such as fibrinogen. Recent clinical trials have demonstrated that targeting inflammatory pathways orchestrated through IL-4, IL-5, IL-13, and the prostaglandin receptor CRTH2 is potentially highly effective in adult asthma.


Subject(s)
Asthma/drug therapy , Asthma/physiopathology , Inflammation/complications , Animals , Asthma/epidemiology , Cytokines/drug effects , Cytokines/metabolism , Environmental Exposure/adverse effects , Humans , Inflammation/microbiology , Respiratory System/microbiology , Th2 Cells/drug effects , Th2 Cells/immunology
12.
J Allergy Clin Immunol ; 142(2): 530-541.e6, 2018 08.
Article in English | MEDLINE | ID: mdl-29038008

ABSTRACT

BACKGROUND: Inhaled protease allergens preferentially trigger TH2-mediated inflammation in allergic asthma. The role of dendritic cells (DCs) on induction of TH2 cell responses in allergic asthma has been well documented; however, the mechanism by which protease allergens induce TH2-favorable DCs in the airway remains unclear. OBJECTIVE: We sought to determine a subset of DCs responsible for TH2 cell responses in allergic asthma and the mechanism by which protease allergens induce the DC subset in the airway. METHODS: Mice were challenged intranasally with protease allergens or fibrinogen cleavage products (FCPs) to induce allergic airway inflammation. DCs isolated from mediastinal lymph nodes were analyzed for surface phenotype and T-cell stimulatory function. Anti-Thy1.2 and Mas-TRECK mice were used to deplete innate lymphoid cells and mast cells, respectively. Adoptive cell transfer, bone marrow DC culture, anti-IL-13, and Toll-like receptor (TLR) 4-deficient mice were used for further mechanistic studies. RESULTS: Protease allergens induced a remarkable accumulation of TH2-favorable programmed cell death 1 ligand 2 (PD-L2)+ DCs in mediastinal lymph nodes, which was significantly abolished in mice depleted of mast cells and, to a lesser extent, innate lymphoid cells. Mechanistically, FCPs generated by protease allergens triggered IL-13 production from wild-type mast cells but not from TLR4-deficient mast cells, which resulted in an increase in the number of PD-L2+ DCs. Intranasal administration of FCPs induced an increase in numbers of PD-L2+ DCs in the airway, which was significantly abolished in TLR4- and mast cell-deficient mice. Injection of IL-13 restored the PD-L2+ DC population in mice lacking mast cells. CONCLUSION: Our findings unveil the "protease-FCP-TLR4-mast cell-IL-13" axis as a molecular mechanism for generation of TH2-favorable PD-L2+ DCs in allergic asthma and suggest that targeting the PD-L2+ DC pathway might be effective in suppressing allergic T-cell responses in the airway.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Fibrinogen/metabolism , Hypersensitivity/immunology , Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , Toll-Like Receptor 4/metabolism , Allergens/immunology , Animals , Cell Differentiation , Disease Models, Animal , Fibrinogen/immunology , Humans , Immunity, Innate , Interleukin-13/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Th2 Cells/immunology , Toll-Like Receptor 4/genetics
13.
Curr Opin Allergy Clin Immunol ; 16(1): 51-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26658015

ABSTRACT

PURPOSE OF REVIEW: The purpose is to review the important recent advances made in how innate immune cells, microbes, and the environment contribute to the expression of allergic disease, emphasizing the allergen-related signals that drive allergic responses. RECENT FINDINGS: The last few years have seen crucial advances in how innate immune cells such as innate lymphoid cells group 2 and airway epithelial cells and related molecular pathways through organismal proteinases and innate immune cytokines, such as thymic stromal lymphopoietin, IL-25, and IL-33 contribute to allergy and asthma. Simultaneously with these advances, important progress has been made in our understanding of how the environment, and especially pathogenic organisms, such as bacteria, viruses, helminths, and especially fungi derived from the natural and built environments, either promote or inhibit allergic inflammation and disease. Of specific interest are how lipopolysaccharide mediates its antiallergic effect through the ubiquitin modifying factor A20 and the antiallergic activity of both helminths and protozoa. SUMMARY: Innate immune cells and molecular pathways, often activated by allergen-derived proteinases acting on airway epithelium and macrophages as well as additional unknown factors, are essential to the expression of allergic inflammation and disease. These findings suggest numerous future research opportunities and new opportunities for therapeutic intervention in allergic disease.


Subject(s)
Allergens/immunology , Hypersensitivity/immunology , Immunity, Innate , Signal Transduction/immunology , Animals , Humans , Hypersensitivity/pathology
14.
Cell Rep ; 11(6): 957-966, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25937283

ABSTRACT

Micro- and nanometer-size particles have become popular candidates for cancer vaccine adjuvants. However, the mechanism by which such particles enhance immune responses remains unclear. Here, we report a porous silicon microparticle (PSM)-based cancer vaccine that greatly enhances cross-presentation and activates type I interferon (IFN-I) response in dendritic cells (DCs). PSM-loaded antigen exhibited prolonged early endosome localization and enhanced cross-presentation through both proteasome- and lysosome-dependent pathways. Phagocytosis of PSM by DCs induced IFN-I responses through a TRIF- and MAVS-dependent pathway. DCs primed with PSM-loaded HER2 antigen produced robust CD8 T cell-dependent anti-tumor immunity in mice bearing HER2+ mammary gland tumors. Importantly, this vaccination activated the tumor immune microenvironment with elevated levels of intra-tumor IFN-I and MHCII expression, abundant CD11c+ DC infiltration, and tumor-specific cytotoxic T cell responses. These findings highlight the potential of PSM as an immune adjuvant to potentiate DC-based cancer immunotherapy.


Subject(s)
Cross-Priming/immunology , Immunity, Innate , Interferon Type I/immunology , Microspheres , Neoplasms/immunology , Silicon/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Female , Mammary Neoplasms, Animal/pathology , Mice, Inbred C57BL , Neoplasms/pathology , Ovalbumin/immunology , Phagocytosis , Porosity , Receptor, ErbB-2/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment
15.
J Interferon Cytokine Res ; 34(3): 141-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24028396

ABSTRACT

Interleukin-33 (IL-33) appears to play a crucial role in the expression of allergic diseases, but its cellular source and regulatory mechanisms remain to be fully elucidated. Mast cells, one of the major effecter cell populations in mediating allergy, express high levels of IL-33 receptor, ST2, and have been shown to express IL-33 transcripts. In this study, we aimed to examine the secretion of IL-33 in mast cells and their response to IL-33. We have successfully detected secreted IL-33 from cell supernatants through a modified enzyme-linked immunosorbent assay (ELISA) technique-cell-based ELISA. Activation of bone marrow-derived cultured mast cells (BMMCs) by crosslinkage of an antigen [ovalbumin (OVA)] and OVA-specific IgE mAbs significantly induced the expression of IL-33 transcripts, cytosolic and secreted proteins. In addition, the Toll-like receptor (TLR) 2 and TLR-9 ligands could trigger IL-33 mRNA expression. Exposure of BMMCs to IL-33 significantly increased the levels of IL-13 and IL-6 expression, concomitant with enhanced activation of mitogen-activated protein kinase (MAPKs) (ERK, p38, and JNK) and nuclear factor-kappa B. These results suggest that mouse BMMCs are capable of producing and serving as endogenous sources of IL-33, and that IL-33 plays an important role in regulating mast cell functions.


Subject(s)
Bone Marrow Cells/immunology , Interleukins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/immunology , Immunoglobulin E/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-13/biosynthesis , Interleukin-33 , Interleukin-6/biosynthesis , Interleukins/genetics , Interleukins/metabolism , JNK Mitogen-Activated Protein Kinases/immunology , Mast Cells/cytology , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , Ovalbumin/immunology , RNA, Messenger/genetics , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 9/immunology , p38 Mitogen-Activated Protein Kinases/immunology
16.
Blood ; 121(16): 3195-204, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23462117

ABSTRACT

We propose that the aryl hydrocarbon receptor (AhR), a unique chemical sensor, is critical in controlling mast cell differentiation, growth, and function in vitro and in vivo. In antigen-stimulated mast cells, exposure to AhR ligands resulted in a calcium- and reactive oxygen species (ROS)-dependent increase of reversible oxidation in and reduced activity of SHP-2 phosphatase, leading to enhanced mast cell signaling, degranulation, and mediator and cytokine release, as well as the in vivo anaphylactic response. Surprisingly, significant mast cell deficiency was noted in AhR-null mice due to defective calcium signaling and mitochondrial function, concomitant with reduced expression of c-kit and cytosolic STAT proteins, as well as enhanced intracellular ROS and apoptosis. Consequently, AhR-null mast cells responded poorly to stimulation, demonstrating a critical role of AhR signaling in maintaining mast cell homeostasis.


Subject(s)
Calcium/immunology , Immunoglobulin E/immunology , Mast Cells/physiology , Reactive Oxygen Species/immunology , Receptors, Aryl Hydrocarbon/immunology , Animals , Antigens/immunology , Apoptosis , Carbazoles/pharmacology , Cell Degranulation , Cells, Cultured , Gene Deletion , Homeostasis , Humans , Mast Cells/drug effects , Mast Cells/pathology , Mice , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...