Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells  Elizabeth.

Slides:



Advertisements
Similar presentations
Poly (ADP-ribose) polymerase 14 and its enzyme activity regulates TH2 differentiation and allergic airway disease  Purvi Mehrotra, PhD, Andrew Hollenbeck,
Advertisements

Serum amyloid P attenuates M2 macrophage activation and protects against fungal spore–induced allergic airway disease  Ana Paula Moreira, PhD, Karen A.
Innate lymphoid cells contribute to allergic airway disease exacerbation by obesity  Laetitia Everaere, PhD, Saliha Ait-Yahia, PhD, Olivier Molendi-Coste,
Surface availability of beta-glucans is critical determinant of host immune response to Cladosporium cladosporioides  Rachael A. Mintz-Cole, PhD, Eric.
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,
Allergen endotoxins induce T-cell–dependent and non–IgE-mediated nasal hypersensitivity in mice  Naruhito Iwasaki, MD, Kazufumi Matsushita, PhD, Ayumi.
Let-7 microRNA–mediated regulation of IL-13 and allergic airway inflammation  Manish Kumar, MSc, Tanveer Ahmad, MSc, Amit Sharma, PhD, Ulaganathan Mabalirajan,
IL-4 blocks TH1-polarizing/inflammatory cytokine gene expression during monocyte- derived dendritic cell differentiation through histone hypoacetylation 
IL-25 and CD4+ TH2 cells enhance type 2 innate lymphoid cell–derived IL-13 production, which promotes IgE-mediated experimental food allergy  Jee-Boong.
Interferon response factor 3 is essential for house dust mite–induced airway allergy  Thomas Marichal, DVM, Denis Bedoret, DVM, PhD, Claire Mesnil, DVM,
Cigarette smoke extract induces thymic stromal lymphopoietin expression, leading to TH2-type immune responses and airway inflammation  Yuki Nakamura,
Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity  Hye Young Kim, PhD, Ya-Jen Chang, PhD, Srividya.
Innate IL-13–producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity  Jillian L. Barlow, PhD, Agustin Bellosi,
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
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,
IL-33 induces innate lymphoid cell–mediated airway inflammation by activating mammalian target of rapamycin  Robert J. Salmond, PhD, Ananda S. Mirchandani,
Neonatal rhinovirus induces mucous metaplasia and airways hyperresponsiveness through IL-25 and type 2 innate lymphoid cells  Jun Young Hong, MS, J. Kelley.
Β-Glucan exacerbates allergic asthma independent of fungal sensitization and promotes steroid-resistant TH2/TH17 responses  Zhonghua Zhang, MD, Jocelyn.
Allergic airway disease is unaffected by the absence of IL-4Rα–dependent alternatively activated macrophages  Natalie E. Nieuwenhuizen, PhD, Frank Kirstein,
Responsiveness to respiratory syncytial virus in neonates is mediated through thymic stromal lymphopoietin and OX40 ligand  Junyan Han, PhD, Azzeddine.
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.
CD4+CD25+ regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling  Jennifer Kearley, PhD, Douglas S. Robinson,
Forkhead box protein 3 demethylation is associated with tolerance induction in peanut- induced intestinal allergy  Meiqin Wang, MD, PhD, Ivana V. Yang,
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,
Ton C. Doan, BA, Brian M. Jeong, MSc, Mackenzie E. Coden, BA, Lucas F
Kathleen R. Bartemes, BA, Gail M. Kephart, BS, Stephanie J
Takao Kobayashi, PhD, Koji Iijima, PhD, Alexander L
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.
Jethe O. F. Nunes, PhD, Juliana de Souza Apostolico, MSc, David A. G
CD4-mediated regulatory T-cell activation inhibits the development of disease in a humanized mouse model of allergic airway disease  Helen Martin, MS,
Β2 integrins rather than β1 integrins mediate Alternaria-induced group 2 innate lymphoid cell trafficking to the lung  Maya R. Karta, PhD, Peter S. Rosenthal,
Does reduced zona pellucida binding protein 2 (ZPBP2) expression on chromosome 17q21 protect against asthma?  Marina Miller, MD, PhD, Christine Vuong,
T-bet inhibits innate lymphoid cell–mediated eosinophilic airway inflammation by suppressing IL-9 production  Ayako Matsuki, MD, Hiroaki Takatori, MD,
The cardiomyocyte protein αT-catenin contributes to asthma through regulating pulmonary vein inflammation  Stephen Sai Folmsbee, G.R. Scott Budinger,
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,
Src homology 2 domain–containing inositol 5-phosphatase 1 deficiency leads to a spontaneous allergic inflammation in the murine lung  Sun-Young Oh, PhD,
MicroRNA-155 is a critical regulator of type 2 innate lymphoid cells and IL-33 signaling in experimental models of allergic airway inflammation  Kristina.
T-bet inhibits innate lymphoid cell–mediated eosinophilic airway inflammation by suppressing IL-9 production  Ayako Matsuki, MD, Hiroaki Takatori, MD,
Programmed cell death ligand 2 regulates TH9 differentiation and induction of chronic airway hyperreactivity  Jerome Kerzerho, PhD, Hadi Maazi, PhD, Anneliese.
Fms-like tyrosine kinase 3 ligand increases a lung DC subset with regulatory properties in allergic airway inflammation  Zhifei Shao, MD, Arpita S. Bharadwaj,
Novel allergic asthma model demonstrates ST2-dependent dendritic cell targeting by cypress pollen  Lucia Gabriele, BS, Giovanna Schiavoni, BS, Fabrizio.
Poly (ADP-ribose) polymerase 14 and its enzyme activity regulates TH2 differentiation and allergic airway disease  Purvi Mehrotra, PhD, Andrew Hollenbeck,
Xin-Zi Tang, PhD, James B. Jung, BS, Christopher D.C. Allen, PhD 
Susceptibility to allergic lung disease regulated by recall responses of dual-receptor memory T cells∗  Mark A. Aronica, MD, Shadi Swaidani, MS, Yan H.
IL-35 production by inducible costimulator (ICOS)–positive regulatory T cells reverses established IL-17–dependent allergic airways disease  Gregory S.
Enhanced production of CCL18 by tolerogenic dendritic cells is associated with inhibition of allergic airway reactivity  Iris Bellinghausen, PhD, Sebastian.
Duy Pham, PhD, Sarita Sehra, PhD, Xin Sun, PhD, Mark H. Kaplan, PhD 
No defect in T-cell priming, secondary response, or tolerance induction in response to inhaled antigens in Fms-like tyrosine kinase 3 ligand–deficient.
Unique and overlapping gene expression patterns driven by IL-4 and IL-13 in the mouse lung  Christina C. Lewis, PhD, Bruce Aronow, PhD, John Hutton, MD,
Serum amyloid P attenuates M2 macrophage activation and protects against fungal spore–induced allergic airway disease  Ana Paula Moreira, PhD, Karen A.
Ozone activates pulmonary dendritic cells and promotes allergic sensitization through a Toll-like receptor 4–dependent mechanism  John W. Hollingsworth,
IL-22 attenuates IL-25 production by lung epithelial cells and inhibits antigen-induced eosinophilic airway inflammation  Kentaro Takahashi, MD, Koichi.
David S. Southam, BSc, Natasha Widmer, Russ Ellis, BSc, Jeremy A
MicroRNA-155 is essential for TH2-mediated allergen-induced eosinophilic inflammation in the lung  Carina Malmhäll, BSc, Sahar Alawieh, BSc, You Lu, PhD,
Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity  Hye Young Kim, PhD, Ya-Jen Chang, PhD, Srividya.
Eric B. Brandt, PhD, Melissa K. Mingler, MS, Michelle D
The extra domain A of fibronectin is essential for allergen-induced airway fibrosis and hyperresponsiveness in mice  Martin Kohan, MSc, Andres F. Muro,
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:

Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells  Elizabeth A. Oczypok, BS, Pavle S. Milutinovic, MD, PhD, John F. Alcorn, PhD, Anupriya Khare, PhD, Lauren T. Crum, BS, Michelle L. Manni, PhD, Michael W. Epperly, PhD, Adriane M. Pawluk, Anuradha Ray, PhD, Tim D. Oury, MD, PhD  Journal of Allergy and Clinical Immunology  Volume 136, Issue 3, Pages 747-756.e4 (September 2015) DOI: 10.1016/j.jaci.2015.03.011 Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 1 IL-33 upregulation in response to HDM extract is dependent on RAGE expression. A and B, Immunoblot probing for IL-33 in whole lung homogenate after 7 weeks of HDM treatments (Fig 1, A) and summary of normalized IL-33/β-actin signal intensity ratios (saline control set arbitrarily to 1.0; Fig 1, B). C, Quantitative RT-PCR of whole lung homogenate RNA after 7 weeks of HDM treatments probing for IL-33. IL-33 signal was normalized to the glycerldehyde-3-phosphate dehydrogenase signal, and results are expressed as fold change over wild-type (WT) saline values. In all cases results are expressed as means ± SEMs (n = 3-6 mice per strain per treatment group). *P < .05 versus comparison. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 2 Absence of RAGE attenuates the cellular inflammatory and airway hyperreactivity responses to A alternata in an acute model of allergic airway disease. A and B, Representative hematoxylin and eosin (Fig 2, A) and periodic acid–Schiff (Fig 2, B) stains (×200 magnification) of wild-type (WT) and RAGE KO mouse lung sections. C, Bronchoalveolar lavage fluid total cell and eosinophil counts normalized to 1 mL. D, Pulmonary function test demonstrating changes in airway resistance (Rn) during methacholine challenge. All samples/functional analyses were collected on day 10. Results are expressed as means ± SEMs (n = 7-14 mice per strain per treatment group for Fig 2, A-C; n = 3-4 mice per group for pulmonary function tests). *P < .05 versus comparison. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 3 Absence of RAGE results in blunted IL-5, IL-13, and IL-33 cytokine responses to A alternata in an acute model of allergic airway disease. A and B, ELISA analyses of IL-5 and IL-13 levels in bronchoalveolar lavage fluid at day 10 in an A alternata model. C, Immunoblot probing for IL-33 in whole lung homogenate after 10-day A alternata treatment and summary of normalized IL-33/β-actin signal intensity ratios (saline controls set arbitrarily to 1.0; right). In all cases results are expressed as means ± SEMs (n = 7-14 mice per strain/treatment group). *P < .05 versus comparison. WT, Wild-type. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 4 Development of inflammatory effects in the lung downstream of exogenous IL-33 depends on RAGE. A and B, Representative hematoxylin and eosin (Fig 4, A) and periodic acid–Schiff (Fig 4, B) stains of lung sections (×200 magnification) from wild-type (WT) and RAGE KO mice after IL-33 administration. C, Bronchoalveolar lavage fluid total cell and eosinophil counts normalized to 1 mL. D, ELISA analyses of IL-5 and IL-13 levels in bronchoalveolar lavage fluid after treatment with rIL-33. All samples were collected after 4 days of IL-33 treatment. In all cases results are expressed as means ± SEMs (n = 3-11 mice per strain per treatment group). *P < .05 versus comparison. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 5 RAGE is required for ILC2 accumulation in the lung. A, Representative flow plots of the live, lineage-negative cell populations gated for expression of CD90.2 and ST2. Numbers in each quadrant are percentages of the parent population. ILC2s were identified as lineage-negative cells that coexpress CD90.2 and ST2. B and C, Graphic summary of flow cytometrically enumerated ILC2s in one lung after 4 days of treatment with rIL-33 (Fig 5, B) or 10 days of treatment with A alternata (Fig 5, C). Results are expressed as means ± SEMs (n = 3-11 mice per strain per treatment group). *P < .05 versus comparison. WT, Wild-type. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 6 Stromal, but not hematopoietic, RAGE drives AAI. A and B, Bronchoalveolar lavage fluid total cell counts (Fig 6, A) and eosinophil counts (Fig 6, B) of bone marrow chimeric mice treated intranasally for 7 weeks with HDM extract or saline control. Treatment groups are labeled as recipient strain (bone marrow donor strain). In all cases results are expressed as means ± SEMs (n = 3-7 mice per strain per treatment group). *P < .05 versus comparison. BM, Bone marrow; ns, not significant; WT, wild-type. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 7 RAGE is not required for ILC2 induction, proliferation, or secretory function in the alimentary tract after intraperitoneal rIL-33 administration. A, Immunoblot probing for RAGE in mouse whole-organ homogenates. B, Representative periodic acid–Schiff stains of mouse ileum sections (×100 magnification). C, IL-5 and IL-13 levels in peritoneal lavage fluid. D, Representative lung sections stained with hematoxylin and eosin (×100 magnification). E, ILC2 (Lin−, CD90.2+, ST2+) counts in the spleen. All samples were collected after 3 days of IL-33 injection. Results are expressed as means ± SEMs (n = 3-5 mice per strain per treatment group). *P < .05 versus comparison. WT, Wild-type. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E1 Models of AAI. A, Chronic HDM model. B, Acute A alternata model. C, Intranasal recombinant IL-33 model. D, Intraperitoneal IL-33 model. i.n., Intranasal; i.p., intraperitoneal. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E2 TSLP and IL-25 expression are not dependent on RAGE expression in the lung. A, There is no significant difference in TSLP expression in whole-lung homogenates after 7 weeks of HDM treatment between any of the strain or treatment groups. B, In contrast, IL-25 secretion in the bronchoalveolar lavage fluid after 7 weeks of HDM exposure is upregulated in response to allergen in both wild-type (WT) and RAGE KO mice but is inadequate to precipitate a type 2 immune response in RAGE KO mice at the levels observed in this disease model.20 Results are expressed as means ± SEMs (n = 3-6 mice per strain per treatment group). *P < .05 versus comparison. ns, Not significant. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E3 Gating strategy for lung and spleen ILC2s. A, Gating strategy for lung ILC2s (used for both IL-33 intranasal and A alternata experiments). B, Gating strategy for spleen ILC2s after IL-33 administered intraperitoneally. In both organs live (green) lineage-negative (blue) cells were selected and analyzed for coexpression of the surface markers CD90.2 and ST2 (red). SSC, Side scatter. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig E4 Numbers of CD4+ T cells expressing IL-5 and IL-13 are not increased in RAGE KO mice in response to allergen. A and B, CD4+IL-5+ cells (Fig E4, A) and CD4+IL-13+ cells (Fig E4, B) are increased in wild-type (WT) mice after 10-day A alternata treatment but are not increased in RAGE KO mice. Results are expressed as means ± SEMs (n = 3-5 mice per strain per treatment group). *P < .05 versus comparison. Journal of Allergy and Clinical Immunology 2015 136, 747-756.e4DOI: (10.1016/j.jaci.2015.03.011) Copyright © 2015 American Academy of Allergy, Asthma & Immunology Terms and Conditions