Β-Glucan exacerbates allergic asthma independent of fungal sensitization and promotes steroid-resistant TH2/TH17 responses  Zhonghua Zhang, MD, Jocelyn.

Slides:



Advertisements
Similar presentations
Dysregulation of type 2 innate lymphoid cells and TH2 cells impairs pollutant-induced allergic airway responses  Katrien C. De Grove, MSc, Sharen Provoost,
Advertisements

Repeated low-dose intradermal allergen injection suppresses allergen-induced cutaneous late responses  Giuseppina Rotiroti, MD, Mohamed Shamji, PhD, Stephen.
Innate lymphoid cells contribute to allergic airway disease exacerbation by obesity  Laetitia Everaere, PhD, Saliha Ait-Yahia, PhD, Olivier Molendi-Coste,
Exposure to allergen and diesel exhaust particles potentiates secondary allergen- specific memory responses, promoting asthma susceptibility  Eric B. Brandt,
Surface availability of beta-glucans is critical determinant of host immune response to Cladosporium cladosporioides  Rachael A. Mintz-Cole, PhD, Eric.
Glucagon-like peptide 1 receptor signaling attenuates respiratory syncytial virus– induced type 2 responses and immunopathology  Melissa H. Bloodworth,
Thymic stromal lymphopoietin signaling in CD4+ T cells is required for TH2 memory  Qun Wang, PhD, Jianguang Du, PhD, Jingjing Zhu, MSc, Xiaowei Yang, MSc,
Weiguo Chen, PhD, Umasundari Sivaprasad, PhD, Aaron M
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
Umasundari Sivaprasad, PhD, David J. Askew, PhD, Mark B
Allergy prevention starts before conception: Maternofetal transfer of tolerance protects against the development of asthma  Tobias Polte, PhD, Christian.
Interferon response factor 3 is essential for house dust mite–induced airway allergy  Thomas Marichal, DVM, Denis Bedoret, DVM, PhD, Claire Mesnil, DVM,
Frank Kirstein, PhD, Natalie E
Maternal house dust mite exposure during pregnancy enhances severity of house dust mite–induced asthma in murine offspring  Phoebe K. Richgels, MS, Amnah.
Lack of autophagy induces steroid-resistant airway inflammation
Exposure to allergen and diesel exhaust particles potentiates secondary allergen- specific memory responses, promoting asthma susceptibility  Eric B. Brandt,
Nazanin Farhadi, MSc, Laura Lambert, BA, Chiara Triulzi, PhD, Peter J
Pentraxin 3 deletion aggravates allergic inflammation through a TH17-dominant phenotype and enhanced CD4 T-cell survival  Jyoti Balhara, MSc, Lianyu Shan,
Compartmentalized chemokine-dependent regulatory T-cell inhibition of allergic pulmonary inflammation  Roshi Afshar, PhD, James P. Strassner, BS, Edward.
IL-33 dysregulates regulatory T cells and impairs established immunologic tolerance in the lungs  Chien-Chang Chen, PhD, Takao Kobayashi, PhD, Koji Iijima,
Restoration of T-box–containing protein expressed in T cells protects against allergen- induced asthma  Jung Won Park, MD, Hyun Jung Min, MS, Jung Ho Sohn,
Group 2 innate lymphoid cells facilitate sensitization to local, but not systemic, TH2- inducing allergen exposures  Matthew J. Gold, BSc, Frann Antignano,
Antigen-specific effector CD8 T cells regulate allergic responses via IFN-γ and dendritic cell function  Yafang Tang, BSc, Shou Ping Guan, BSc, Benson.
CD1d restricted natural killer T cells are not required for allergic skin inflammation  Abdallah Elkhal, PhD, Muriel Pichavant, PhD, Rui He, PhD, Jordan.
Responsiveness to respiratory syncytial virus in neonates is mediated through thymic stromal lymphopoietin and OX40 ligand  Junyan Han, PhD, Azzeddine.
Eosinophils contribute to the resolution of lung-allergic responses following repeated allergen challenge  Katsuyuki Takeda, MD, PhD, Yoshiki Shiraishi,
Lung dendritic cells induce TH17 cells that produce TH2 cytokines, express GATA-3, and promote airway inflammation  Marianne Raymond, PhD, Vu Quang Van,
Signaling through FcRγ-associated receptors on dendritic cells drives IL-33–dependent TH2-type responses  Melissa Y. Tjota, BA, Cara L. Hrusch, PhD, Kelly.
Activin A and TGF-β promote TH9 cell–mediated pulmonary allergic pathology  Carla P. Jones, PhD, Lisa G. Gregory, PhD, Benjamin Causton, BSc, Gaynor A.
Chang Xiao, MD, PhD, Jocelyn M
Prime role of IL-17A in neutrophilia and airway smooth muscle contraction in a house dust mite–induced allergic asthma model  Julie Chesné, PhD, Faouzi.
CD4+CD25+ regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling  Jennifer Kearley, PhD, Douglas S. Robinson,
Experimental gastrointestinal allergy enhances pulmonary responses to specific and unrelated allergens  Eric B. Brandt, PhD, Troy A. Scribner, MD, Hiroko.
Frank Kirstein, PhD, Natalie E
Inhibition of house dust mite–induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment  Adam Collison,
Overexpression of sirtuin 6 suppresses allergic airway inflammation through deacetylation of GATA3  Hyun-Young Jang, PhD, Suna Gu, MS, Sang-Myeong Lee,
T-cell immunoglobulin and mucin domain 1 deficiency eliminates airway hyperreactivity triggered by the recognition of airway cell death  Hye Young Kim,
Surface availability of beta-glucans is critical determinant of host immune response to Cladosporium cladosporioides  Rachael A. Mintz-Cole, PhD, Eric.
IL-2–inducible T-cell kinase modulates TH2-mediated allergic airway inflammation by suppressing IFN-γ in naive CD4+ T cells  Arun K. Kannan, MS, Nisebita.
Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells  Elizabeth.
Exaggerated IL-17 response to epicutaneous sensitization mediates airway inflammation in the absence of IL-4 and IL-13  Rui He, MD, PhD, Hye Young Kim,
Jethe O. F. Nunes, PhD, Juliana de Souza Apostolico, MSc, David A. G
Regulation of allergic airway inflammation by class I–restricted allergen presentation and CD8 T-cell infiltration  James W. Wells, PhD, Christopher J.
Janus kinase 1/3 signaling pathways are key initiators of TH2 differentiation and lung allergic responses  Shigeru Ashino, PhD, Katsuyuki Takeda, MD,
Prophylactic and therapeutic inhibition of allergic airway inflammation by probiotic Escherichia coli O83  Christian Zwicker, MSc, Priya Sarate, MSc,
Allergen-induced IL-6 trans-signaling activates γδ T cells to promote type 2 and type 17 airway inflammation  Md Ashik Ullah, MPharm, Joana A. Revez,
Dysregulation of type 2 innate lymphoid cells and TH2 cells impairs pollutant-induced allergic airway responses  Katrien C. De Grove, MSc, Sharen Provoost,
Does reduced zona pellucida binding protein 2 (ZPBP2) expression on chromosome 17q21 protect against asthma?  Marina Miller, MD, PhD, Christine Vuong,
Role of B cells in TH cell responses in a mouse model of asthma
T-bet inhibits innate lymphoid cell–mediated eosinophilic airway inflammation by suppressing IL-9 production  Ayako Matsuki, MD, Hiroaki Takatori, MD,
Sarita Sehra, PhD, Weiguo Yao, PhD, Evelyn T. Nguyen, MS, Nicole L
Allergic skin sensitization promotes eosinophilic esophagitis through the IL-33–basophil axis in mice  Nicholas Venturelli, BS, Willem S. Lexmond, MD,
T-bet inhibits innate lymphoid cell–mediated eosinophilic airway inflammation by suppressing IL-9 production  Ayako Matsuki, MD, Hiroaki Takatori, MD,
IL-17A enhances IL-13 activity by enhancing IL-13–induced signal transducer and activator of transcription 6 activation  Sara L. Hall, MS, Theresa Baker,
Fms-like tyrosine kinase 3 ligand increases a lung DC subset with regulatory properties in allergic airway inflammation  Zhifei Shao, MD, Arpita S. Bharadwaj,
Staphylococcal enterotoxin A–activated regulatory T cells promote allergen-specific TH2 response to intratracheal allergen inoculation  Wei-ping Zeng,
Enhanced production of CCL18 by tolerogenic dendritic cells is associated with inhibition of allergic airway reactivity  Iris Bellinghausen, PhD, Sebastian.
IL-10–treated dendritic cells decrease airway hyperresponsiveness and airway inflammation in mice  Toshiyuki Koya, MD, PhD, Hiroyuki Matsuda, MD, PhD,
Duy Pham, PhD, Sarita Sehra, PhD, Xin Sun, PhD, Mark H. Kaplan, PhD 
IL-22 attenuates IL-25 production by lung epithelial cells and inhibits antigen-induced eosinophilic airway inflammation  Kentaro Takahashi, MD, Koichi.
MicroRNA-155 is essential for TH2-mediated allergen-induced eosinophilic inflammation in the lung  Carina Malmhäll, BSc, Sahar Alawieh, BSc, You Lu, PhD,
Breast-feeding, aeroallergen sensitization, and environmental exposures during infancy are determinants of childhood allergic rhinitis  Christopher D.
Eric B. Brandt, PhD, Melissa K. Mingler, MS, Michelle D
Repeated low-dose intradermal allergen injection suppresses allergen-induced cutaneous late responses  Giuseppina Rotiroti, MD, Mohamed Shamji, PhD, Stephen.
Salmeterol attenuates chemotactic responses in rhinovirus-induced exacerbation of allergic airways disease by modulating protein phosphatase 2A  Luke.
KIF3A genetic variation is associated with pediatric asthma in the presence of eczema independent of allergic rhinitis  Elisabet Johansson, PhD, Jocelyn.
Junqing Cui, PhD, Stephen Pazdziorko, Joy S
TNF can contribute to multiple features of ovalbumin-induced allergic inflammation of the airways in mice  Susumu Nakae, PhD, Carolina Lunderius, PhD,
IL-2–inducible T-cell kinase modulates TH2-mediated allergic airway inflammation by suppressing IFN-γ in naive CD4+ T cells  Arun K. Kannan, MS, Nisebita.
Presentation transcript:

β-Glucan exacerbates allergic asthma independent of fungal sensitization and promotes steroid-resistant TH2/TH17 responses  Zhonghua Zhang, MD, Jocelyn M. Biagini Myers, PhD, Eric B. Brandt, PhD, Patrick H. Ryan, PhD, Mark Lindsey, BS, Rachael A. Mintz- Cole, MD, PhD, Tiina Reponen, PhD, Stephen J. Vesper, PhD, Frank Forde, BS, Brandy Ruff, BS, Stacey A. Bass, AS, Grace K. LeMasters, PhD, David I. Bernstein, MD, James Lockey, MD, Alison L. Budelsky, PhD, Gurjit K. Khurana Hershey, MD, PhD  Journal of Allergy and Clinical Immunology  Volume 139, Issue 1, Pages 54-65.e8 (January 2017) DOI: 10.1016/j.jaci.2016.02.031 Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 1 Fungal exposure is associated with increased asthma prevalence at age 7 years among CCAAPS children. A, Prevalence of asthma at age 7 years in ERMI-high versus ERMI-low exposed children (P < .001). B, Data in Fig 1, A, stratified by SPT response positivity to 1 or more environmental allergens any time during ages 1 to 7 years (P < .001). C, Data in Fig 1, B, excluding children with fungus sensitivity (P = .003). **P < .01 and ***P < .001. ns, Not significant. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 2 Coexposure to HDM and A versicolor worsens AHR and inflammation, activates TH17 responses, and exacerbates HDM-induced TH2 responses. A, Airway resistance measured 48 hours after the last intratracheal challenge. B, Total BALF cell counts. C, Representative hematoxylin and eosin staining of lung tissue. Bar = 50 μm. D, BALF cell differential counts. Eos, Eosinophils; Lymph, lymphocytes; Macro, macrophages; Neutro, neutrophils. E, IL-17A expression in lung tissue was assessed by using quantitative PCR and ELISA. F, IL-4 and IL-13 mRNA levels in lung tissue were assessed by using quantitative PCR. *P < .05, **P < .01, and ***P < .001, 2-way ANOVA for AHR. ns, Not significant. Data are from 3 separate experiments (n = 6 for control and A versicolor treatment groups, n = 7 for HDM and H+A treatment groups) and presented as means ± SEMs. Asp, A versicolor; H+A, HDM plus A versicolor; SAL, saline. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 3 Coexposure to HDM and curdlan worsens AHR and inflammation, activates TH17 responses, and exacerbates HDM-induced TH2 responses. A, Airway resistance measured 48 hours after the last intratracheal challenge. B, Total BALF cell counts. C, Representative hematoxylin and eosin staining of lung tissues. Bar = 50 μm. D, BALF cell differential counts. E and F, TH17 and TH2 cytokine levels in lung tissue were assessed by using either quantitative PCR (IL-17A, IL-4, and IL-13) or ELISA (IL-17A). *P < .05, **P < .01, and ***P < .001, 2-way ANOVA for AHR. Data are from 3 separate experiments (n = 6 for control and curdlan treatment groups, n = 9 per HDM and H+C treatment groups) and presented as means ± SEMs. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 4 Effect of curdlan on HDM-induced asthma is dependent on IL-17A induction through dectin-1. A, Airway resistance and BALF neutrophil counts of wild-type and dectin-1 KO mice after exposure to HDM or coexposure to curdlan and HDM per protocol in Fig E1, A. **P < .01 and ****P < .0001, 2-way ANOVA for AHR (n = 6 per group). B, Airway resistance and BALF neutrophil counts of mice treated with either IL-17A neutralizing or control IgG in HDM and curdlan–coexposed mice. **P < .01, and ***P < .001, 2-way ANOVA for AHR (n = 8 per group). Data from 3 separate experiments are presented as means ± SEMs. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 5 Coexposure to curdlan and HDM induces steroid-resistant airway resistance and TH2/TH17 dual-positive T cells in the lungs of exposed mice. A-C, Asthma was induced per the protocol outlined in Fig E1, A, in wild-type (Fig 5, A and B) and Il17rc−/− mice (Fig 5, C), and dexamethasone (1 mg/kg) was administered intraperitoneally for the last 5 days, as indicated, before assessment of mice. Fig 5, A, Airway resistance measured in the wild-type mice (n = 4 for control and curdlan treatment groups, n = 6 for HDM and H+C treatment groups). Fig 5, B, Reduction of airway resistance and BALF eosinophil and neutrophil counts after dexamethasone treatment in the HDM and H+C groups in wild-type mice, respectively. Fig 5, C, Reduction of airway resistance and BALF eosinophil and neutrophil counts after dexamethasone treatment in the H+C group in IL-17RC−/− mice (n = 6). D-F, intracellular cytokine staining for TH2 (IL-13+) cells, TH17 (IL-17A+) cells, and TH2/TH17 dual-producing T cells (IL-13+IL-17A+), respectively. *P < .05, **P < .01, and ***P < .001, 2-way ANOVA for AHR (n = 3-5 per group). Data from 3 separate experiments are presented as means ± SEMs. DEX, Dexamethasone; H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 6 Fungal exposure is associated with increased serum IL-17A levels and increased asthma severity in children. A, Percentage of CCAAPS children with high IL-17A serum levels is increased in the group with high ERMI values versus those with low ERMI values (P = .014). B, Percentage of GCPCR children with asthma who have fungal exposure among those who have symptoms more than once a week (n = 258) versus those with symptoms less than once a week (n = 81, P = .026). C, Proposed working model. *P < .05. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E1 Coexposure to HDM and A versicolor induces enhanced HDM sensitization and mixed eosinophilia with neutrophilia. A, Experimental asthma protocol. B, HDM-specific IgG1 and IgE titers from mice represented in Fig 2. C, BALF cells from mice coexposed to HDM and A versicolor (Fig 2). **P < .01 and ***P < .001, 1-way ANOVA, followed by a Bonferroni posttest. Data are presented as means ± SEMs. Asp, A versicolor; H+A, HDM plus A versicolor; SAC, sacrifice; SAL, saline. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E2 Indicated cytokine and chemokine levels of the mice (represented in Fig 2) exposed to A versicolor, HDM, or both assessed by using either ELISA (BALF, A) or quantitative PCR (lung tissue, B-D). *P < .05, **P < .01, and ***P < .001 (both H+A vs Asp and H+A vs HDM for CCL11), 1-way ANOVA, followed by a Bonferroni posttest. ns, Not significant. Data are presented as means ± SEMs. Asp, A versicolor; H+A, HDM plus A versicolor; SAL, saline. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E3 Indicated cytokine and chemokine levels of mice (represented in Fig 3) exposed to curdlan, HDM, or both assessed by using either ELISA (BALF, A) or quantitative PCR (lung tissue, B and C). *P < .05, **P < .01, and ***P < .001 (both H+C vs Curdlan and H+C vs HDM for CCL11), 1-way ANOVA, followed by a Bonferroni posttest. Data are presented as means ± SEMs. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E4 Coexposure to HDM and curdlan induces enhanced HDM sensitization. HDM-specific IgG1 and IgE titers from mice represented in Fig 3. **P < .01, 1-way ANOVA, followed by a Bonferroni posttest. ns, Not significant. Data are presented as means ± SEMs. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E5 Representative hematoxylin and eosin staining of lung tissues from mice represented in Fig 4, A. Bar = 50 μm. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E6 Representative hematoxylin and eosin staining of lung tissues from mice represented in Fig 5, A. Bar = 50 μm. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E7 Representative fluorescence-activated cell sorting dot plots of lung cells exposed in vivo to HDM plus curdlan (H+C) and restimulated ex vivo with phorbol 12-myristate 13-acetate/ionomycin to assess intracellular cytokine accumulation in CD4+ T cells. Frequency of IL-13+ TH2 cells, IFN-γ+ TH1 cells, IL-17A+ TH17 cells, and IL-13+/IL-17A+ double producers. To properly separate the cytokine-positive cells from the negative cells, we used fluorescence minus one (FMO). Statistics done by using 1-way ANOVA with the Bonferroni multiple comparison test. *P < .05 and ***P < .001. H+C, HDM plus curdlan. Journal of Allergy and Clinical Immunology 2017 139, 54-65.e8DOI: (10.1016/j.jaci.2016.02.031) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions