Innate immune responses during respiratory tract infection with a bacterial pathogen induce allergic airway sensitization  Nicolas W.J. Schröder, MD,

Slides:



Advertisements
Similar presentations
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,
Advertisements

Maternal immune response to helminth infection during pregnancy determines offspring susceptibility to allergic airway inflammation  Kathrin Straubinger,
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model  Sylvie Amu, PhD, Sean P.
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
Virus-specific IgE enhances airway responsiveness on reinfection with respiratory syncytial virus in newborn mice  Azzeddine Dakhama, PhD, Young-Mok Lee,
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,
Selective control of SIRP-α–positive airway dendritic cell trafficking through CD47 is critical for the development of TH2-mediated allergic inflammation 
Innate IL-13–producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity  Jillian L. Barlow, PhD, Agustin Bellosi,
Expression of IL-4 receptor α on smooth muscle cells is not necessary for development of experimental allergic asthma  Frank Kirstein, PhD, William G.C.
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
Protective role of nuclear factor of activated T cells 2 in CD8+ long-lived memory T cells in an allergy model  Roman Karwot, PhD, Joachim H. Maxeiner,
Elisabeth Roesler, MSc, Richard Weiss, PhD, Esther E
Nazanin Farhadi, MSc, Laura Lambert, BA, Chiara Triulzi, PhD, Peter J
Phosphodiesterase 4B is essential for TH2-cell function and development of airway hyperresponsiveness in allergic asthma  S.-L. Catherine Jin, PhD, Sho.
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,
Airway inflammation after epicutaneous sensitization of mice requires protease activity of low-dose allergen inhalation  Izumi Nishioka, MD, PhD, Toshiro.
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,
Crosstalk of regulatory T cells and tolerogenic dendritic cells prevents contact allergy in subjects with low zone tolerance  Ulrike Luckey, PhD, Talkea.
Antigen-specific effector CD8 T cells regulate allergic responses via IFN-γ and dendritic cell function  Yafang Tang, BSc, Shou Ping Guan, BSc, Benson.
Neonatal rhinovirus induces mucous metaplasia and airways hyperresponsiveness through IL-25 and type 2 innate lymphoid cells  Jun Young Hong, MS, J. Kelley.
CCR10 regulates balanced maintenance and function of resident regulatory and effector T cells to promote immune homeostasis in the skin  Mingcan Xia,
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,
Activated glycoprotein A repetitions predominant (GARP)–expressing regulatory T cells inhibit allergen-induced intestinal inflammation in humanized mice 
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.
Notch signaling in T cells is essential for allergic airway inflammation, but expression of the Notch ligands Jagged 1 and Jagged 2 on dendritic cells.
Surfactant protein D inhibits TNF-α production by macrophages and dendritic cells in mice  László Hortobágyi, MS, Sonja Kierstein, PhD, Kateryna Krytska,
Early-life chlamydial lung infection enhances allergic airways disease through age- dependent differences in immunopathology  Jay C. Horvat, PhD, Malcolm.
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,
Frank Kirstein, PhD, Natalie E
Specific epicutaneous immunotherapy prevents sensitization to new allergens in a murine model  Lucie Mondoulet, PhD, Vincent Dioszeghy, PhD, Emilie Puteaux,
T-cell immunoglobulin and mucin domain 1 deficiency eliminates airway hyperreactivity triggered by the recognition of airway cell death  Hye Young Kim,
Takao Kobayashi, PhD, Koji Iijima, PhD, Alexander L
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.
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,
Peanut-induced intestinal allergy is mediated through a mast cell–IgE–FcεRI–IL-13 pathway  Meiqin Wang, MD, PhD, Katsuyuki Takeda, MD, PhD, Yoshiki Shiraishi,
A novel allergen-specific therapy with CD40-silenced B cells and dendritic cells  Motohiko Suzuki, MD, PhD, Makoto Yokota, MD, PhD, Yoshihisa Nakamura,
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
T-bet inhibits innate lymphoid cell–mediated eosinophilic airway inflammation by suppressing IL-9 production  Ayako Matsuki, MD, Hiroaki Takatori, MD,
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,
Intravenous immunoglobulin attenuates airway inflammation through induction of forkhead box protein 3–positive regulatory T cells  Amir H. Massoud, MSc,
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,
Sustained increases in numbers of pulmonary dendritic cells after respiratory syncytial virus infection  Marc Beyer, MD, Holger Bartz, MD, Katharina Hörner,
Duy Pham, PhD, Sarita Sehra, PhD, Xin Sun, PhD, Mark H. Kaplan, PhD 
Rhinovirus infection interferes with induction of tolerance to aeroantigens through OX40 ligand, thymic stromal lymphopoietin, and IL-33  Amit K. Mehta,
No defect in T-cell priming, secondary response, or tolerance induction in response to inhaled antigens in Fms-like tyrosine kinase 3 ligand–deficient.
DNA methylation of TH1/TH2 cytokine genes affects sensitization and progress of experimental asthma  Stephanie Brand, PhD, Dörthe Andrea Kesper, 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,
TH17 cells mediate pulmonary collateral priming
Eric B. Brandt, PhD, Melissa K. Mingler, MS, Michelle D
Epicutaneous immunization with ovalbumin and CpG induces TH1/TH17 cytokines, which regulate IgE and IgG2a production  Monika Majewska-Szczepanik, PhD,
Nrf2 activation by sulforaphane restores the age-related decrease of TH1 immunity: Role of dendritic cells  Hyon-Jeen Kim, PhD, Berenice Barajas, BS,
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:

Innate immune responses during respiratory tract infection with a bacterial pathogen induce allergic airway sensitization  Nicolas W.J. Schröder, MD, Timothy R. Crother, PhD, Yoshikazu Naiki, PhD, Shuang Chen, MD, Michelle H. Wong, MD, Atilla Yilmaz, MD, Anatoly Slepenkin, PhD, Danica Schulte, MD, Randa Alsabeh, MD, Terence M. Doherty, BA, Ellena Peterson, MD, Andre E. Nel, MD, Moshe Arditi, MD  Journal of Allergy and Clinical Immunology  Volume 122, Issue 3, Pages 595-602.e5 (September 2008) DOI: 10.1016/j.jaci.2008.06.038 Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 1 Low-dose live C pneumoniae infection induces allergic airway sensitization to HSA. A, Groups of mice were infected as indicated. Starting with day 0, mice received either 3 consecutive intranasal injections of HSA (n = 13 for low dose and n = 9 for high dose) or PBS control (n = 13 for low dose and n = 8 for high dose). Some mice were infected with 0.5 × 106C pneumoniae, and sensitization with HSA (day 0) was started 10 days after infection (n = 9) or PBS control (n = 10). Mice were challenged with HSA starting on day 15. One control group received PBS during the challenge period (n = 10). B, Eosinophil (eos) numbers per lung section, with representative lung sections (100-fold magnification) are shown at the right of the graph. C, Goblet cell numbers related to bronchial basal membrane length. Representative periodic acid–Schiff–stained sections (200-fold magnification) are shown. D, HSA-specific IgE, IgG1, and IgG2a titers of mice sensitized and challenged with HSA after infection. ∗P ≤ .05, ∗∗P ≤ .01. ns, Not significant compared with control animals. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 2 Characteristics of pulmonary DCs in C pneumoniae–infected mice. A, Paraffin-embedded sections were stained with MIDC-8 to detect DCs. Total numbers of MIDC-8+ cells in 4 separate sections were counted (6 mice per group). B, Upregulation of costimulatory molecules assessed by means of staining of lung leukocytes with antibodies against CD11c, CD11b, and MHC class II. Shown are uninfected control animals (gray), mice infected with high-dose C pneumoniae (dotted line) or low-dose C pneumoniae (bold line) 5 days before analysis, and mice infected with low-dose C pneumoniae 10 days before analysis (thin line). These are representative results from 1 of 3 experiments performed with 5 mice. C, Numbers of FITC+ DCs in mediastinal lymph nodes (LN) after infection and intratracheal injection of 100 μg of FITC–HSA 24 hours before analysis. Cells were gated for CD11b. Shown are representative dot blots of cells (FITC–HSA × CD11c), as well as combined results of 3 separate experiments (n = 4 mice per experiment). ∗P ≤ .05. ND, Not determined. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 3 DCs from C pneumoniae–infected mice pulsed with HSA can adoptively transfer allergic airway sensitization. A, DCs were isolated from lungs of mice 5 or 10 days after infection with low-dose C pneumoniae or sham infection. DC expression of MHC class II expression by infected mice and control animals was determined by means of FACS. Cells were incubated with either PBS or HSA overnight and transferred to recipient mice by means of intratracheal inoculation (500,000 cells per mouse) on day 0. Mice were challenged with HSA starting on day 15. B, Eosinophil (eos) numbers in lung sections from mice that received control DCs pulsed with HSA (n = 5) or DCs from infected mice pulsed with either HSA (n = 7) or PBS (n = 8). C, Goblet cell numbers assessed by means of periodic acid–Schiff staining. Shown are combined results from 3 experiments. ∗P ≤ .05 D, Cytokine production by lymph node (LN) cells after in vitro restimulation with HSA. Shown are representative results from 1 of 3 experiments performed. E, IL-6 RNA content of pulmonary DCs of mice 5 days and 10 days after infection. IL-6 RNA was measured by using TaqMan real-time RT-PCR and compared with that seen in DCs of uninfected control mice. Combined data of 4 mice per group are shown. F, Percentage of lung CD4+CD25+Foxp3+ Treg cells in naive wild-type mice (n = 6-9 mice) 5 days after infection with either 5 × 105 or 5 × 106C pneumoniae or 10 days after infection with 0.5 × 106 administered intranasally. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 4 Treg cells regulate airway sensitization induced by C pneumoniae (CP) infection. A, Infection, Treg cell depletion, and sensitization/challenge protocol: mice were infected with 0.5 × 106C pneumoniae on day −10, followed by a single intraperitoneal injection of 100 μg of PC61 antibody (n = 8) or rat IgG1 isotype control (n = 7) at day −2. Mice were then exposed to HSA on 3 consecutive days, followed by HSA challenge 2 weeks later. Control mice were injected with PC61 or isotype control (n = 8 and n = 7, respectively) without prior infection, followed by HSA injection on days 0 to 2. B and C, Eosinophil (eos) numbers and goblet cell numbers. D, HSA-specific IgE titers. ∗P ≤ .05. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 5 MyD88-dependent signaling in DCs is required for airway sensitization. A and B, Eosinophil (eos) numbers and goblet cell numbers in lung sections from wild-type mice (n = 7) infected with 0.5 × 106 live C pneumoniae, exposed to HSA on 3 consecutive days, and challenged with HSA, as shown in Fig 1, A. Data obtained from MyD88−/− mice (n = 8 mice are shown in comparison). ∗P ≤ .05, ∗∗P ≤ .01. ns, Not significant. Wild-type mice (n = 7) and MyD88−/− mice (n = 6) were infected with 0.5 × 106C pneumoniae and killed after 5 days. C, Upregulation of MHC class II, CD80, CD86, and CD40 in DCs of infected mice (bold line, wild-type mice; dotted line, MyD88−/− mice) versus uninfected control animals (gray, wild-type mice; dark gray, MyD88−/− mice) determined by means of FACS. DCs were identified as CD11c+/CD11b+. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Characteristics of pneumonia caused by low-dose and high-dose C pneumoniae at day 5 or low-dose C pneumoniae at day 10. Mice (n = 6 per group) were infected by means of intranasal injection of either 0.5 × 106 or 5 × 106 IFU of C pneumoniae 5 days or 0.5 × 106C pneumoniae 10 days before analysis. A, Representative hematoxylin and eosin–stained sections of paraffin-embedded lungs. B, Inflammatory scores obtained from hematoxylin and eosin–stained sections and levels of IL-6, TNF-α, IFN-γ, and IL-5 in the BAL fluid. ∗P ≤ .05, ∗∗P ≤ .01. ns, Not significant. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Cytokine production and expression of costimulatory molecules in DCs isolated from C pneumoniae–infected mice. Mice were infected with either 0.5 × 106 or 5 × 106 IFU of C pneumoniae, and lung leukocytes were prepared as described in the Methods section. A, Cells were stained for expression of CD11b and CD11c, and staining for intracellular cytokines was performed with antibodies against IL-10–phycoerythrin or IL-12–phycoerythrin. B, Upregulation of costimulatory molecules assessed by staining of lung leukocytes with antibodies against CD11c, CD11b, CD40, CD80, and CD86. Shown are uninfected control animals (gray), mice infected with high-dose C pneumoniae (dotted line) or low-dose C pneumoniae (bold line) 5 days before analysis, and mice infected with low-dose C pneumoniae 10 days before analysis (thin line). Shown are representative results from 1 of 3 experiments performed with 5 mice. C, Total numbers of MHC class IIhigh DCs in mediastinal lymph nodes (LN) 5 days after infection. Shown are representative dot blots of cells gated for CD11b, as well as combined results of 3 separate experiments (n = 4 per group). D, Percentages of DCs in infected mice undergoing apoptosis. DCs were identified by using CD11c, CD11b, and side-scatter. Apoptotic cells were identified with the Vybrant Apoptosis Assay Kit (Invitrogen). Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Depletion of CD4+CD25+ Treg cells in the lungs after injection of anti-CD25 antibody. Mice received intraperitoneal injections of 100 μg of either anti-CD25 antibody (clone PC61) or rat IgG1 isotype control on day 0. Mice were killed 2 or 4 days after injection. Splenocytes and lung leukocytes were prepared as described in the Methods section and were then analyzed for Foxp3+ Treg cells by means of flow cytometry with antibodies against CD4, CD25 (clone 7D4), and Foxp3. Percentages refer to total CD4+ lymphocytes. Shown are representative results from 1 of 3 experiments performed with 3 mice. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

MyD88-dependent signaling in DCs is required for airway sensitization MyD88-dependent signaling in DCs is required for airway sensitization. BMDCs from wild-type (wt) and MyD88−/− mice were exposed to C pneumoniae (multiplicity of infection = 2.5) and HSA (50 μg/mL) in the presence of gentamicin for 16 hours, followed by intratracheal transfer into recipient mice. Wild-type mice were injected with wild-type DCs (n = 8) or MyD88−/− DCs (n = 11). One group of MyD88−/− mice received wild-type DCs (n = 10). A, Inflammatory scores determined by using hematoxylin and eosin–stained lung sections. B and C, Eosinophil (eos) numbers and goblet cell numbers in lung sections. ∗P ≤ .05, ∗∗P ≤ .01, ∗∗∗P ≤ .001. D, BMDCs generated from wild-type and MyD88−/− mice were infected with C pneumoniae (multiplicity of infection = 2.5), and content of C pneumoniae was determined after 24 hours by means of immunofluorescence with the Pathfinder staining kit (Sigma). Shown are representative results from 1 of 3 separate experiments. Journal of Allergy and Clinical Immunology 2008 122, 595-602.e5DOI: (10.1016/j.jaci.2008.06.038) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions