Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36835202

ABSTRACT

Asthma heterogeneity complicates the search for targeted treatment against airway inflammation and remodeling. We sought to investigate relations between eosinophilic inflammation, a phenotypic feature frequent in severe asthma, bronchial epithelial transcriptome, and functional and structural measures of airway remodeling. We compared epithelial gene expression, spirometry, airway cross-sectional geometry (computed tomography), reticular basement membrane thickness (histology), and blood and bronchoalveolar lavage (BAL) cytokines of n = 40 moderate to severe eosinophilic (EA) and non-eosinophilic asthma (NEA) patients distinguished by BAL eosinophilia. EA patients showed a similar extent of airway remodeling as NEA but had an increased expression of genes involved in the immune response and inflammation (e.g., KIR3DS1), reactive oxygen species generation (GYS2, ATPIF1), cell activation and proliferation (ANK3), cargo transporting (RAB4B, CPLX2), and tissue remodeling (FBLN1, SOX14, GSN), and a lower expression of genes involved in epithelial integrity (e.g., GJB1) and histone acetylation (SIN3A). Genes co-expressed in EA were involved in antiviral responses (e.g., ATP1B1), cell migration (EPS8L1, STOML3), cell adhesion (RAPH1), epithelial-mesenchymal transition (ASB3), and airway hyperreactivity and remodeling (FBN3, RECK), and several were linked to asthma in genome- (e.g., MRPL14, ASB3) or epigenome-wide association studies (CLC, GPI, SSCRB4, STRN4). Signaling pathways inferred from the co-expression pattern were associated with airway remodeling (e.g., TGF-ß/Smad2/3, E2F/Rb, and Wnt/ß-catenin).


Subject(s)
Asthma , Pulmonary Eosinophilia , Respiratory Mucosa , Humans , Airway Remodeling/genetics , Asthma/genetics , Calmodulin-Binding Proteins , GPI-Linked Proteins , Inflammation , Pulmonary Eosinophilia/genetics , SOXB2 Transcription Factors , Transcriptome , Respiratory Mucosa/metabolism
2.
Chest ; 159(4): e181-e184, 2021 04.
Article in English | MEDLINE | ID: mdl-34022014

ABSTRACT

Some severe asthmatic patients experience frequent bacterial respiratory tract infections, which contribute significantly to their disease burden, and often are attributed to their use of systemic corticosteroids and comorbid bronchiectasis. We report a case of a 58-year-old woman who had prednisone-dependent asthma and exacerbations with intense mixed eosinophilic and neutrophilic bronchitis. Autosomal dominant hyper-IgE syndrome, which is a primary immunodeficiency characterized by elevated IgE, eosinophilia, and recurrent infections, caused by a novel pathogenic mutation in STAT3 was identified as the cause of her airway disease. We believe that this is the first report of the demonstration of an IL-5 driven eosinophilia that is associated with a STAT3 mutation that was treated successfully with an anti-IL5 biological.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , DNA/genetics , Loss of Function Mutation , Prednisone/therapeutic use , Pulmonary Eosinophilia/drug therapy , STAT3 Transcription Factor/genetics , Anti-Asthmatic Agents/therapeutic use , Asthma/genetics , Asthma/metabolism , DNA Mutational Analysis , Disease Progression , Drug Therapy, Combination , Female , Glucocorticoids/therapeutic use , Humans , Middle Aged , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/metabolism , STAT3 Transcription Factor/metabolism
3.
J Clin Invest ; 131(7)2021 04 01.
Article in English | MEDLINE | ID: mdl-33792561

ABSTRACT

Airway eosinophilia is a hallmark of allergic asthma and is associated with mucus production, airway hyperresponsiveness, and shortness of breath. Although glucocorticoids are widely used to treat asthma, their prolonged use is associated with several side effects. Furthermore, many individuals with eosinophilic asthma are resistant to glucocorticoid treatment, and they have an unmet need for novel therapies. Here, we show that UDP-glucose (UDP-G), a nucleotide sugar, is selectively released into the airways of allergen-sensitized mice upon their subsequent challenge with that same allergen. Mice lacking P2Y14R, the receptor for UDP-G, had decreased airway eosinophilia and airway hyperresponsiveness compared with wild-type mice in a protease-mediated model of asthma. P2Y14R was dispensable for allergic sensitization and for the production of type 2 cytokines in the lung after challenge. However, UDP-G increased chemokinesis in eosinophils and enhanced their response to the eosinophil chemoattractant, CCL24. In turn, eosinophils triggered the release of UDP-G into the airway, thereby amplifying eosinophilic recruitment. This positive feedback loop was sensitive to therapeutic intervention, as a small molecule antagonist of P2Y14R inhibited airway eosinophilia. These findings thus reveal a pathway that can be therapeutically targeted to treat asthma exacerbations and glucocorticoid-resistant forms of this disease.


Subject(s)
Asthma/immunology , Eosinophils/immunology , Pulmonary Eosinophilia/immunology , Receptors, Purinergic P2Y/immunology , Uridine Diphosphate Glucose/immunology , Allergens/immunology , Animals , Asthma/genetics , Asthma/pathology , Chemokine CCL24/genetics , Chemokine CCL24/immunology , Eosinophils/pathology , Male , Mice , Mice, Knockout , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/pathology , Receptors, Purinergic P2Y/deficiency , Th2 Cells/immunology , Th2 Cells/pathology , Uridine Diphosphate Glucose/genetics
5.
J Allergy Clin Immunol ; 144(3): 764-776, 2019 09.
Article in English | MEDLINE | ID: mdl-31082458

ABSTRACT

BACKGROUND: Lung eosinophilia is a hallmark of asthma, and eosinophils are believed to play a crucial role in the pathogenesis of allergic inflammatory diseases. Short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate, are produced in high amounts in the gastrointestinal tract by commensal bacteria and can be absorbed into the bloodstream. Although there is recent evidence that SCFAs are beneficial in allergic asthma models, the effect on eosinophils has remained elusive. OBJECTIVE: The role of SCFAs was investigated in human eosinophil function and a mouse model of allergic asthma. METHODS: Eosinophils were purified from self-reported allergic or healthy donors. Migration, adhesion to the endothelium, and eosinophil survival were studied in vitro. Ca2+ flux, apoptosis, mitochondrial membrane potential, and expression of surface markers were determined by using flow cytometry and in part by using real-time PCR. Allergic airway inflammation was assessed in vivo in an ovalbumin-induced asthma model by using invasive spirometry. RESULTS: For the first time, we observed that SCFAs were able to attenuate human eosinophils at several functional levels, including (1) adhesion to the endothelium, (2) migration, and (3) survival. These effects were independent from GPR41 and GPR43 but were accompanied by histone acetylation and mimicked by trichostatin A, a pan-histone deacetylase inhibitor. In vivo butyrate ameliorated allergen-induced airway and lung eosinophilia, reduced type 2 cytokine levels in bronchial fluid, and improved airway hyperresponsiveness in mice. CONCLUSION: These in vitro and in vivo findings highlight the importance of SCFAs, especially butyrate as a promising therapeutic agent in allergic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Butyrates/pharmacology , Butyrates/therapeutic use , Eosinophils/drug effects , Pulmonary Eosinophilia/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Asthma/genetics , Asthma/immunology , Cell Movement/drug effects , Eosinophils/immunology , Eosinophils/physiology , Female , Gene Expression Regulation/drug effects , Humans , Mice, Inbred BALB C , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology
6.
Respir Res ; 20(1): 14, 2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30658649

ABSTRACT

BACKGROUND: Benralizumab, a humanized, afucosylated, monoclonal antibody that targets interleukin-5 receptor α, depletes eosinophils and basophils by enhanced antibody-dependent cell-mediated cytotoxicity. It demonstrated efficacy for patients with moderate to severe asthma and, in a Phase IIa trial, for chronic obstructive pulmonary disease (COPD) with eosinophilic inflammation. We investigated effects of benralizumab 100 mg every 8 weeks (first three doses every 4 weeks) subcutaneous on blood inflammatory markers through proteomic and gene-expression analyses collected during two Phase II studies of patients with eosinophilic asthma and eosinophilic COPD. METHODS: Serum samples for proteomic analysis and whole blood for gene expression analysis were collected at baseline and 52 weeks (asthma study) or 32 weeks (COPD study) post-treatment. Proteomic analyses were conducted on a custom set of 90 and 147 Rules-Based Medicine analytes for asthma and COPD, respectively. Gene expression was profiled by Affymetrix Human Genome U133 plus 2 arrays (~ 54 K probes). Gene set variation analysis (GSVA) was used to determine transcriptomic activity of immune signatures. Treatment-related differences between analytes, genes, and gene signatures were analyzed for the overall population and for patient subgroups stratified by baseline blood eosinophil count (eosinophil-high [≥300 cells/µL] and eosinophil-low [< 300 cells/µL]) via t-test and repeated measures analysis of variance. RESULTS: Eosinophil chemokines eotaxin-1 and eotaxin-2 were significantly upregulated (false discovery rate [FDR] < 0.05) by approximately 2.1- and 1.4-fold in the asthma study and by 2.3- and 1.7-fold in the COPD study following benralizumab treatment. Magnitude of upregulation of these two chemokines was greater for eosinophil-high patients than eosinophil-low patients in both studies. Benralizumab was associated with significant reductions (FDR < 0.05) in expression of genes associated with eosinophils and basophils, such as CLC, IL-5Rα, and PRSS33; immune-signaling complex genes (FCER1A); G-protein-coupled receptor genes (HRH4, ADORA3, P2RY14); and further immune-related genes (ALOX15 and OLIG2). The magnitude of downregulation of gene expression was greater for eosinophil-high than eosinophil-low patients. GSVA on immune signatures indicated significant treatment reductions (FDR < 0.05) in eosinophil-associated signatures. CONCLUSIONS: Benralizumab is highly selective, modulating blood proteins or genes associated with eosinophils or basophils. Modulated protein and gene expression patterns are most prominently altered in eosinophil-high vs. eosinophil-low patients. TRIAL REGISTRATION: NCT01227278 and NCT01238861 .


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Inflammation Mediators/blood , Pulmonary Eosinophilia/blood , Pulmonary Eosinophilia/drug therapy , Adolescent , Adult , Aged , Anti-Asthmatic Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Biomarkers/blood , Double-Blind Method , Female , Humans , Inflammation Mediators/antagonists & inhibitors , Male , Middle Aged , Proteomics/methods , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Eosinophilia/genetics , Young Adult
7.
Sci Rep ; 8(1): 18052, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575775

ABSTRACT

Certain proteases derived from house dust mites and plants are considered to trigger initiation of allergic airway inflammation by disrupting tight junctions between epithelial cells. It is known that inhalation of proteases such as house dust mite-derived Der p1 and/or papaya-derived papain caused airway eosinophilia in naïve mice and even in Rag-deficient mice that lack acquired immune cells such as T, B and NKT cells. In contrast, little is known regarding the possible involvement of proteases derived from Aspergillus species (fungal-associated proteases; FAP), which are ubiquitous saprophytic fungi in the environment, in the development of allergic airway eosinophilia. Here, we found that inhalation of FAP by naïve mice led to airway eosinophilia that was dependent on protease-activated receptor-2 (PAR2), but not TLR2 and TLR4. Those findings suggest that the protease activity of FAP, but not endotoxins in FAP, are important in the setting. In addition, development of that eosinophilia was mediated by innate immune cells (ILCs) such as innate lymphoid cells, but not by acquired immune cells such as T, B and NKT cells. Whereas IL-33, IL-25 and thymic stromal lymphopoietin (TSLP) are involved in induction of FAP-induced ILC-mediated airway eosinophilia, IL-33-rather than IL-25 and/or TSLP-was critical for the eosinophilia in our model. Our findings improve our understanding of the molecular mechanisms involved in induction of airway inflammation by FAP.


Subject(s)
Aspergillus/immunology , Cytokines/physiology , Immunity, Innate/physiology , Interleukin-33/physiology , Interleukins/physiology , Peptide Hydrolases/immunology , Pneumonia/immunology , Allergens/immunology , Animals , Aspergillus/enzymology , Aspergillus/metabolism , Immunity, Cellular/physiology , Lung Diseases, Fungal/complications , Lung Diseases, Fungal/enzymology , Lung Diseases, Fungal/genetics , Lung Diseases, Fungal/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Hydrolases/metabolism , Pneumonia/genetics , Pneumonia/metabolism , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Thymic Stromal Lymphopoietin
8.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1058-L1068, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28798252

ABSTRACT

A mucosal oxidative burst is a hallmark response to pollen exposure that promotes allergic inflammatory responses. Reactive species constituents of oxidative stress signal via the modification of cellular molecules including nucleic acids. One of the most abundant forms of oxidative genomic base damage is 8-oxo-7,8-dihydroguanine (8-oxoG), which is removed from DNA by 8-oxoguanine DNA glycosylase 1 (OGG1). OGG1 in complex with 8-oxoG acts as a GDP-GTP exchange factor and induces acute inflammation; however, the mechanism(s) by which OGG1 signaling regulates allergic airway inflammation is not known. Here, we postulate that the OGG1 signaling pathway differentially altered the levels of small regulatory RNAs and increased the expression of T helper 2 (Th2) cytokines in ragweed pollen extract (RWPE)-challenged lungs. To determine this, the lungs of sensitized mice expressing or lacking OGG1 were challenged with RWPE and/or with OGG1's excision product 8-oxoG. The responses in lungs were assessed by next-generation sequencing, as well as various molecular and histological approaches. The results showed that RWPE challenge induced oxidative burst and damage to DNA and activated OGG1 signaling, resulting in the differential expression of 84 micro-RNAs (miRNAs), which then exacerbated antigen-driven allergic inflammation and histological changes in the lungs. The exogenous administration of the downregulated let-7b-p3 mimetic or inhibitors of upregulated miR-23a or miR-27a decreased eosinophil recruitment and mucus and collagen production via controlling the expression of IL-4, IL-5, and IL-13. Together, these data demonstrate the roles of OGG1 signaling in the regulation of antigen-driven allergic immune responses via differential expression of miRNAs upstream of Th2 cytokines and eosinophils.


Subject(s)
Antigens, Plant/toxicity , DNA Damage , Hypersensitivity/immunology , MicroRNAs/immunology , Plant Extracts/toxicity , Pulmonary Eosinophilia/immunology , Th2 Cells/immunology , Animals , Cell Line, Transformed , Cytokines/genetics , Cytokines/immunology , DNA Glycosylases/genetics , DNA Glycosylases/immunology , Hypersensitivity/genetics , Hypersensitivity/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/genetics , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/pathology , Th2 Cells/pathology
9.
Am J Respir Cell Mol Biol ; 57(6): 674-682, 2017 12.
Article in English | MEDLINE | ID: mdl-28700253

ABSTRACT

IgE contributes to disease exacerbations but not to baseline airway hyperresponsiveness (AHR) in human asthma. In rodent allergic airway disease (AAD), mast cell and IgE dependence for the induction of AHR has only been observed when mice are immunized with a relatively weak allergen without adjuvant. To evaluate the role of IgE in murine AAD that is induced by a potent allergen, we inoculated BALB/c and FVB/N background wild-type and IgE- or FcεRIα-deficient mice intratracheally with large or limiting doses of house dust mite extract (HDM) and evaluated AHR, pulmonary eosinophilia, goblet cell metaplasia, serum IgE, and lung mastocytosis. We found that neither IgE nor FcεRIα contributed to AAD, even in mice inoculated with the lowest dose of HDM, which readily induced detectable disease, but did not increase serum IgE or pulmonary mast cell levels. In contrast, high doses of HDM strikingly increased serum IgE and pulmonary mast cells, although both AHR and airway mast cell degranulation were equally elevated in wild-type and IgE-deficient mice. Surprisingly, allergen challenge of mice with severe AAD and pulmonary mastocytosis failed to acutely increase airway resistance, lung Newtonian resistance, or hysteresis. Overall, this study shows that, although mice may not reliably model acute asthma exacerbations, mechanisms that are IgE and FcεRIα independent are responsible for AHR and airway inflammation when low doses of a potent allergen are inhaled repetitively.


Subject(s)
Allergens/immunology , Asthma/immunology , Immunoglobulin E/immunology , Pulmonary Eosinophilia/immunology , Pyroglyphidae/immunology , Receptors, IgE/immunology , Animals , Asthma/genetics , Asthma/pathology , Goblet Cells/immunology , Goblet Cells/pathology , Humans , Mastocytosis/immunology , Mastocytosis/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pulmonary Eosinophilia/genetics , Receptors, IgE/genetics
10.
Klin Padiatr ; 228(6-07): 319-324, 2016 Nov.
Article in German | MEDLINE | ID: mdl-27846662

ABSTRACT

Based on a case report an overview on the differential diagnostic considerations with respect to blood hypereosinophilia (HE) and hypereosinophilic syndromes (HES) in childhood is given. A 13-year-old boy was admitted for the clarification of an asthma. In the blood count an increased HE with 3 500/µl (30%) was found along with elevated total serum IgE and IL-5 level (2 000 IU/ml and 17 pg/ml). Lung function showed an obstruction (FEV1 38%). Radiologically the picture of bronchiectasis and mucus pluggine appeared. In the BAL a HE (76%) with raised IL-5 level was apparent. Histologically asthma was diagnosed with mucostasis, hypertrophy of the bronchial wall musculature and a lung HE. Differential-diagnostically an ABPA, a Churg-Strauss-Syndrome, a parasitosis, drug associated HE, allergies and malignant disease could be excluded. An aberrant T-cell clone in peripheral blood was detected by flow cytometry and T-cell receptor clonal rearrangements by PCR, leading to the diagnosis of a lymphoid variant of HES (L-HES). Failure to detect the FIP1L1-PDGFRA gene fusion and a normal bone marrow examination could exclude a neoplastic HES (HESN). After steroid initiation, prompt decrease of blood eosinophilia with resolution of symptoms was observed. Steroid discontinuation led to eosinophilia recurrence associated with disease symptoms. As steroid-sparing agent the immunosuppressive azathioprine was additionally given; steroid doses could be decreased and stopped in the course. This case demonstrated the range of HE evaluation in infancy. With asthma one should also consider the possibility of a L-HES.


Subject(s)
Asthma/diagnosis , Asthma/immunology , Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/immunology , T-Lymphocytes/immunology , Adolescent , Asthma/genetics , Asthma/pathology , Azathioprine/therapeutic use , Biopsy, Needle , Bone Marrow/pathology , Bronchi/pathology , Diagnosis, Differential , Flow Cytometry , Forced Expiratory Volume/physiology , Humans , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/pathology , Immunoglobulin E/blood , Interleukin-5/blood , Lung/pathology , Oncogene Proteins, Fusion/genetics , Pulmonary Eosinophilia/diagnosis , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
11.
J Allergy Clin Immunol ; 137(2): 591-600.e1, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26602165

ABSTRACT

BACKGROUND: The transcription factor promyelocytic leukemia zinc finger (PLZF) is transiently expressed during development of type 2 innate lymphoid cells (ILC2s) but is not present at the mature stage. We hypothesized that PLZF-deficient ILC2s have functional defects in the innate allergic response and represent a tool for studying innate immunity in a mouse with a functional adaptive immune response. OBJECTIVE: We determined the consequences of PLZF deficiency on ILC2 function in response to innate and adaptive immune stimuli by using PLZF(-/-) mice and mixed wild-type:PLZF(-/-) bone marrow chimeras. METHODS: PLZF(-/-) mice, wild-type littermates, or mixed bone marrow chimeras were treated with the protease allergen papain or the cytokines IL-25 and IL-33 or infected with the helminth Nippostrongylus brasiliensis to induce innate type 2 allergic responses. Mice were sensitized with intraperitoneal ovalbumin-alum, followed by intranasal challenge with ovalbumin alone, to induce adaptive TH2 responses. Lungs were analyzed for immune cell subsets, and alveolar lavage fluid was analyzed for ILC2-derived cytokines. In addition, ILC2s were stimulated ex vivo for their capacity to release type 2 cytokines. RESULTS: PLZF-deficient lung ILC2s exhibit a cell-intrinsic defect in the secretion of IL-5 and IL-13 in response to innate stimuli, resulting in defective recruitment of eosinophils and goblet cell hyperplasia. In contrast, the adaptive allergic inflammatory response to ovalbumin and alum was unimpaired. CONCLUSIONS: PLZF expression at the innate lymphoid cell precursor stage has a long-range effect on the functional properties of mature ILC2s and highlights the importance of these cells for innate allergic responses in otherwise immunocompetent mice.


Subject(s)
Hypersensitivity/genetics , Hypersensitivity/immunology , Immunity, Innate/genetics , Kruppel-Like Transcription Factors/genetics , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Adoptive Transfer , Allergens/immunology , Animals , Antigens, Surface/metabolism , Biomarkers , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Disease Models, Animal , Helminthiasis/genetics , Helminthiasis/immunology , Helminthiasis/pathology , Helminths/immunology , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Immunophenotyping , Interleukin-33/administration & dosage , Interleukin-33/pharmacology , Interleukins/administration & dosage , Interleukins/pharmacology , Kruppel-Like Transcription Factors/deficiency , Lymphocyte Activation , Lymphocyte Subsets/drug effects , Mice , Mice, Knockout , Ovalbumin/immunology , Papain/administration & dosage , Papain/pharmacology , Promyelocytic Leukemia Zinc Finger Protein , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Th2 Cells/immunology , Th2 Cells/metabolism
12.
J Immunol ; 195(8): 3537-45, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26342029

ABSTRACT

Aspirin-exacerbated respiratory disease (AERD), a severe eosinophilic inflammatory disorder of the airways, involves overproduction of cysteinyl leukotrienes (cysLTs), activation of airway mast cells (MCs), and bronchoconstriction in response to nonselective cyclooxygenase inhibitors that deplete homeostatic PGE2. The mechanistic basis for MC activation in this disorder is unknown. We now demonstrate that patients with AERD have markedly increased epithelial expression of the alarmin-like cytokine IL-33 in nasal polyps, as compared with polyps from aspirin-tolerant control subjects. The murine model of AERD, generated by dust mite priming of mice lacking microsomal PGE2 synthase (ptges(-/-) mice), shows a similar upregulation of IL-33 protein in the airway epithelium, along with marked eosinophilic bronchovascular inflammation. Deletion of leukotriene C4 synthase, the terminal enzyme needed to generate cysLTs, eliminates the increased IL-33 content of the ptges(-/-) lungs and sharply reduces pulmonary eosinophilia and basal secretion of MC products. Challenges of dust mite-primed ptges(-/-) mice with lysine aspirin induce IL-33-dependent MC activation and bronchoconstriction. Thus, IL-33 is a component of a cysLT-driven innate type 2 immune response that drives pathogenic MC activation and contributes substantially to AERD pathogenesis.


Subject(s)
Asthma, Aspirin-Induced/immunology , Immunity, Innate , Interleukin-33/immunology , Leukotrienes/immunology , Mast Cells/immunology , Adolescent , Adult , Aged , Animals , Asthma, Aspirin-Induced/genetics , Asthma, Aspirin-Induced/pathology , Dinoprostone/genetics , Dinoprostone/immunology , Female , Humans , Interleukin-33/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Leukotrienes/genetics , Male , Mast Cells/pathology , Mice , Mice, Knockout , Middle Aged , Prostaglandin-E Synthases , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology
13.
J Immunol ; 194(8): 3583-93, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25780046

ABSTRACT

Type-2 innate lymphoid cells (ILC2s) and the acquired CD4(+) Th2 and Th17 cells contribute to the pathogenesis of experimental asthma; however, their roles in Ag-driven exacerbation of chronic murine allergic airway diseases remain elusive. In this study, we report that repeated intranasal rechallenges with only OVA Ag were sufficient to trigger airway hyperresponsiveness, prominent eosinophilic inflammation, and significantly increased serum OVA-specific IgG1 and IgE in rested mice that previously developed murine allergic airway diseases. The recall response to repeated OVA inoculation preferentially triggered a further increase of lung OVA-specific CD4(+) Th2 cells, whereas CD4(+) Th17 and ILC2 cell numbers remained constant. Furthermore, the acquired CD4(+) Th17 cells in Stat6(-/-)/IL-17-GFP mice, or innate ILC2s in CD4(+) T cell-ablated mice, failed to mount an allergic recall response to OVA Ag. After repeated OVA rechallenge or CD4(+) T cell ablation, the increase or loss of CD4(+) Th2 cells resulted in an enhanced or reduced IL-13 production by lung ILC2s in response to IL-25 and IL-33 stimulation, respectively. In return, ILC2s enhanced Ag-mediated proliferation of cocultured CD4(+) Th2 cells and their cytokine production, and promoted eosinophilic airway inflammation and goblet cell hyperplasia driven by adoptively transferred Ag-specific CD4(+) Th2 cells. Thus, these results suggest that an allergic recall response to recurring Ag exposures preferentially triggers an increase of Ag-specific CD4(+) Th2 cells, which facilitates the collaborative interactions between acquired CD4(+) Th2 cells and innate ILC2s to drive the exacerbation of a murine allergic airway diseases with an eosinophilic phenotype.


Subject(s)
Asthma/immunology , Cell Communication/immunology , Immunity, Innate , Pulmonary Eosinophilia/immunology , Th2 Cells/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cell Communication/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-33 , Interleukins/genetics , Interleukins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/toxicity , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/pathology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/pathology
14.
Toxicology ; 326: 74-85, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25456268

ABSTRACT

This study examines the immunological responses in rats following inhalation to titanium dioxide nanoparticles (TiO2 NPs), in naïve rats and in rats with induced allergic airway disease. The responses of two different inbred rat strains were compared: the Dark Aguoti (DA), susceptible to chronic inflammatory disorders, and the Brown Norwegian (BN), susceptible to atopic allergic inflammation. Naïve rats were exposed to an aerosol of TiO2 NPs once daily for 10 days. Another subset of rats was sensitized to the allergen ovalbumin (OVA) in order to induce airway inflammation. These sensitized rats were exposed to TiO2 NPs before and during the allergen challenge. Naïve rats exposed to TiO2 NPs developed an increase of neutrophils and lymphocytes in both rat strains. Airway hyperreactivity and production of inflammatory mediators typical of a T helper 1 type immune response were significantly increased, only in DA rats. Sensitization of the rats induced a prominent OVA-specific-IgE and IgG response in the BN rat while DA rats only showed an increased IgG response. Sensitized rats of both strains developed airway eosinophilia following allergen challenge, which declined upon exposure to TiO2 NPs. The level of neutrophils and lymphocytes increased upon exposure to TiO2 NPs in the airways of DA rats but remained unchanged in the airways of BN rats. In conclusion, the responses to TiO2 NPs were strain-dependent, indicating that genetics play a role in both immune and airway reactivity. DA rats were found to be higher responder compared to BN rats, both when it comes to responses in naïve and sensitized rats. The impact of genetically determined factors influencing the inflammatory reactions pinpoints the complexity of assessing health risks associated with nanoparticle exposures.


Subject(s)
Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/genetics , Genetic Variation , Lung/drug effects , Metal Nanoparticles/toxicity , Pneumonia/chemically induced , Pneumonia/genetics , Titanium/toxicity , Aerosols , Animals , Bronchial Hyperreactivity/blood , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Cytokines/metabolism , Disease Models, Animal , Genotype , Immunoglobulin E/blood , Immunoglobulin G/blood , Inflammation Mediators/metabolism , Inhalation Exposure/adverse effects , Lung/immunology , Lung/physiopathology , Male , Neutrophils/drug effects , Neutrophils/immunology , Ovalbumin , Phenotype , Pneumonia/blood , Pneumonia/immunology , Pneumonia/physiopathology , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Rats, Inbred BN , Risk Factors , Species Specificity , Th1 Cells/drug effects , Th1 Cells/immunology
15.
Respir Res ; 15: 132, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25344652

ABSTRACT

BACKGROUND: CD86-CD28 interaction has been suggested as the principal costimulatory pathway for the activation and differentiation of naïve T cells in allergic inflammation. However, it remains uncertain whether this pathway also has an essential role in the effector phase. We sought to determine the contribution of CD86 on dendritic cells in the reactivation of allergen-specific Th2 cells. METHODS: We investigated the effects of the downregulation of CD86 by short interfering RNAs (siRNAs) on Th2 cytokine production in the effector phase in vitro and on asthma phenotypes in ovalbumin (OVA)-sensitized and -challenged mice. RESULTS: Treatment of bone marrow-derived dendritic cells (BMDCs) with CD86 siRNA attenuated LPS-induced upregulation of CD86. CD86 siRNA treatment impaired BMDCs' ability to activate OVA-specific Th2 cells. Intratracheal administration of CD86 siRNA during OVA challenge downregulated CD86 expression in the airway mucosa. CD86 siRNA treatment ameliorated OVA-induced airway eosinophilia, airway hyperresponsiveness, and the elevations of OVA-specific IgE in the sera and IL-5, IL-13, and CCL17 in the bronchoalveolar lavage fluid, but not the goblet cell hyperplasia. CONCLUSION: These results suggest that local administration of CD86 siRNA during the effector phase ameliorates lines of asthma phenotypes. Targeting airway dendritic cells with siRNA suppresses airway inflammation and hyperresponsiveness in an experimental model of allergic asthma.


Subject(s)
Asthma/prevention & control , B7-2 Antigen/metabolism , Bronchial Hyperreactivity/prevention & control , Lung/metabolism , RNAi Therapeutics , Th2 Cells/metabolism , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , B7-2 Antigen/genetics , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Bronchoconstriction , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Immunoglobulin E/blood , Lung/immunology , Lung/physiopathology , Lymphocyte Activation , Mice, Inbred BALB C , Ovalbumin , Phenotype , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Pulmonary Eosinophilia/prevention & control , RNA Interference , Th2 Cells/immunology , Transfection
16.
Allergol Int ; 63 Suppl 1: 29-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24809373

ABSTRACT

BACKGROUND: The eosinophil is deeply associated with the pathogenesis of bronchial asthma and other allergic diseases. We recently identified a novel eosinophil-specific cell surface molecule, major facilitator super family domain containing 10 (Mfsd10). A monoclonal antibody (mAb) against Mfsd10 (M2) showed selective binding and neutralizing activities for eosinophils. However, the relative potency of the blockage of Mfsd10 and other eosinophil-specific molecules for the treatment of allergic diseases has not been evaluated. Therefore, in this study, the effects of M2 and an anti-Siglec-F mAb on antigen-immunized and antigen-specific Th2 cell-transferred murine eosinophilic inflammation models were comparatively investigated. METHODS: Ovalbumin (OVA)-specific Th2 cells were differentiated from naïve CD4+ T cells of DO11.10/RAG-2-/- mice in vitro and cytokine producing activity of the Th2 cells was examined. OVA-immunized and Th2 cell-transferred BALB/c mice were treated with M2 or anti-Siglec-F and challenged with OVA. Then the number of inflammatory cells and the concentration of IL-5 in the bronchoalveolar lavage fluid (BALF) were determined. RESULTS: Antigen-specific Th2 cells produced large amounts of IL-4, IL-5 and IL-13 but not IL-17A or IFN-γ. Administration of M2 significantly suppressed antigen-induced lung eosinophil infiltration both in OVA-immunized and Th2 cell-transferred mice. The potency as well as selectivity of M2 for down-regulating eosinophils was quite similar to that of anti-Siglec-F. Both mAbs did not affect antigen-induced IL-5 production in the lungs. CONCLUSIONS: Mfsd10 as well as Siglec-F could be an effective target to treat eosinophil-related disorders including bronchial asthma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunosuppressive Agents/pharmacology , Membrane Proteins/antagonists & inhibitors , Pulmonary Eosinophilia/immunology , Th2 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Disease Models, Animal , Eosinophils/immunology , Female , Interleukin-5/biosynthesis , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Mice , Mice, Knockout , Pulmonary Eosinophilia/drug therapy , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/pathology , Sialic Acid Binding Immunoglobulin-like Lectins , Th2 Cells/metabolism
17.
Pediatr Pulmonol ; 49(3): E52-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24106060

ABSTRACT

Dedicator of cytokinesis 8 (DOCK8) deficiency is an autosomal recessive type of combined immunodeficiency with elevated IgE. In this report, we describe a Japanese girl of non-consanguineous family suffering from acute eosinophilic pneumonia (AEP) as a presenting feature of DOCK8 deficiency. Although AEP was self-limiting, consecutively experienced recurrent respiratory infections, severe atopic dermatitis, and vulnerability to viral infections, prompted us to evaluate the possibility of DOCK8 deficiency. Immunological assessments demonstrated decreased IgM, increased IgE, T lymphocytepenia, especially in CD4 T cells, decreased PHA blastogenesis, and decreased CD27(+) CD19(+) memory B cells. Western blotting revealed the absence of DOCK8 protein. Investigation of genomic DNA by multiplex ligation-dependent probe amplification (MLPA) revealed a heterozygous large deletion of 77 kb spanning from intron 5 to exon 22. DOCK8 cDNA sequencing revealed a nonsense mutation at position 740 (E740X). As far as we know, this is the first Japanese case of DOCK8 deficiency.


Subject(s)
Base Sequence , Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/genetics , Pulmonary Eosinophilia/genetics , Sequence Deletion/genetics , Candidiasis/complications , Candidiasis/diagnosis , Candidiasis/genetics , Child, Preschool , Female , Guanine Nucleotide Exchange Factors/deficiency , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/diagnosis , Lung/diagnostic imaging , Lung/pathology , Lung Diseases, Fungal/complications , Lung Diseases, Fungal/diagnosis , Lung Diseases, Fungal/genetics , Moraxellaceae Infections/complications , Moraxellaceae Infections/diagnosis , Moraxellaceae Infections/genetics , Otitis Media/complications , Otitis Media/diagnosis , Otitis Media/genetics , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/genetics , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/diagnosis , Pneumonia, Pneumococcal/genetics , Pulmonary Eosinophilia/complications , Pulmonary Eosinophilia/diagnosis , Radiography , Sequence Analysis, DNA , Sinusitis/complications , Sinusitis/diagnosis , Sinusitis/genetics , Staphylococcal Infections/complications , Staphylococcal Infections/diagnosis , Staphylococcal Infections/genetics , Streptococcal Infections/complications , Streptococcal Infections/diagnosis , Streptococcal Infections/genetics
18.
PLoS One ; 8(11): e79985, 2013.
Article in English | MEDLINE | ID: mdl-24244587

ABSTRACT

Ovarian cancer G protein-coupled receptor 1 (OGR1) stimulation by extracellular protons causes the activation of G proteins and subsequent cellular functions. However, the physiological and pathophysiological roles of OGR1 in airway responses remain largely unknown. In the present study, we show that OGR1-deficient mice are resistant to the cardinal features of asthma, including airway eosinophilia, airway hyperresponsiveness (AHR), and goblet cell metaplasia, in association with a remarkable inhibition of Th2 cytokine and IgE production, in an ovalbumin (OVA)-induced asthma model. Intratracheal transfer to wild-type mice of OVA-primed bone marrow-derived dendritic cells (DCs) from OGR1-deficient mice developed lower AHR and eosinophilia after OVA inhalation compared with the transfer of those from wild-type mice. Migration of OVA-pulsed DCs to peribronchial lymph nodes was also inhibited by OGR1 deficiency in the adoption experiments. The presence of functional OGR1 in DCs was confirmed by the expression of OGR1 mRNA and the OGR1-sensitive Ca(2+) response. OVA-induced expression of CCR7, a mature DC chemokine receptor, and migration response to CCR7 ligands in an in vitro Transwell assay were attenuated by OGR1 deficiency. We conclude that OGR1 on DCs is critical for migration to draining lymph nodes, which, in turn, stimulates Th2 phenotype change and subsequent induction of airway inflammation and AHR.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Dendritic Cells/immunology , Pulmonary Eosinophilia/immunology , Receptors, G-Protein-Coupled/immunology , Adoptive Transfer , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/pathology , Calcium/metabolism , Cell Movement , Dendritic Cells/pathology , Dendritic Cells/transplantation , Female , Gene Expression Regulation , Goblet Cells/immunology , Goblet Cells/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Lung/immunology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Knockout , Ovalbumin , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/pathology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Th1-Th2 Balance , Th2 Cells/immunology , Th2 Cells/pathology
19.
Mol Immunol ; 56(4): 480-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911404

ABSTRACT

Neuroimmune semaphorin 4D (Sema4D) was found to be expressed and function in the nervous and immune systems. In the immune system, Sema4D is constitutively expressed on T cells and regulates T cell priming. In addition, it displays a stimulatory function on macrophages, DC, NK cells, and neutrophils. As all these cells are deeply involved in asthma pathology, we hypothesized that Sema4D plays a critical non-redundant regulatory role in allergic airway response. To test our hypothesis, we exposed Sema4D(-/-) and WT mice to OVA injections and challenges in the well-defined mouse model of OVA-induced experimental asthma. We observed a significant decrease in eosinophilic airway infiltration in allergen-treated Sema4D(-/-) mice relative to WT mice. This reduced allergic inflammatory response was associated with decreased BAL IL-5, IL-13, TGFß1, IL-6, and IL-17A levels. In addition, T cell proliferation in OVA323₋339-restimulated Sema4D(-/-) cell cultures was downregulated. We also found increased Treg numbers in spleens of Sema4D(-/-) mice. However, airway hyperreactivity (AHR) to methacholine challenges was not affected by Sema4D deficiency in either acute or chronic experimental disease setting. Surprisingly, lung DC number and activation were not affected by Sema4D deficiency. These data provide a new insight into Sema4D biology and define Sema4D as an important regulator of Th2-driven lung pathophysiology and as a potential target for a combinatory disease immunotherapy.


Subject(s)
Antigens, CD/immunology , Hypersensitivity/immunology , Lung/immunology , Pneumonia/immunology , Semaphorins/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Lung/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Pneumonia/genetics , Pneumonia/metabolism , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
20.
PLoS One ; 8(6): e67115, 2013.
Article in English | MEDLINE | ID: mdl-23805294

ABSTRACT

Experimental evidence and epidemiological studies indicate that exposure to endotoxin lipopolysaccharide (eLPS) or other TLR agonists prevent asthma. We have previously shown in the OVA-model of asthma that eLPS administration during alum-based allergen sensitization blocked the development of lung TH2 immune responses via MyD88 pathway and IL-12/IFN-γ axis. In the present work we determined the effect of eLPS exposure during sensitization to a natural airborne allergen extract derived from the house dust mite Blomia tropicalis (Bt). Mice were subcutaneously sensitized with Bt allergens co-adsorbed onto alum with or without eLPS and challenged twice intranasally with Bt. Cellular and molecular parameters of allergic lung inflammation were evaluated 24 h after the last Bt challenge. Exposure to eLPS but not to ultrapure LPS (upLPS) preparation during sensitization to Bt allergens decreased the influx of eosinophils and increased the influx of neutrophils to the airways. Inhibition of airway eosinophilia was not observed in IFN-γdeficient mice while airway neutrophilia was not observed in IL-17RA-deficient mice as well in mice lacking MyD88, CD14, TLR4 and, surprisingly, TLR2 molecules. Notably, exposure to a synthetic TLR2 agonist (PamCSK4) also induced airway neutrophilia that was dependent on TLR2 and TLR4 molecules. In the OVA model, exposure to eLPS or PamCSK4 suppressed OVA-induced airway inflammation. Our results suggest that B. tropicalis allergens engage TLR4 that potentiates TLR2 signaling. This dual TLR activation during sensitization results in airway neutrophilic inflammation associated with increased frequency of lung TH17 cells. Our work highlight the complex interplay between bacterial products, house dust mite allergens and TLR signaling in the induction of different phenotypes of airway inflammation.


Subject(s)
Allergens/toxicity , Asthma/immunology , Lipopolysaccharides/toxicity , Neutrophils/immunology , Pyroglyphidae/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Allergens/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Disease Models, Animal , Eosinophils/immunology , Eosinophils/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Th17 Cells/pathology , Th2 Cells/pathology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...