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,

Slides:



Advertisements
Similar presentations
Innate lymphoid cells contribute to allergic airway disease exacerbation by obesity  Laetitia Everaere, PhD, Saliha Ait-Yahia, PhD, Olivier Molendi-Coste,
Advertisements

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.
Therapeutic reversal of food allergen sensitivity by mature retinoic acid–differentiated dendritic cell induction of LAG3+CD49b−Foxp3− regulatory T cells 
Effective prevention and therapy of experimental allergic asthma using a GATA-3– specific DNAzyme  Serdar Sel, MD, Michael Wegmann, PhD, Tanja Dicke, MSc,
CD4 T-helper cells engineered to produce IL-10 prevent allergen-induced airway hyperreactivity and inflammation  Jae-Won Oh, MD, PhD, Christine M. Seroogy,
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,
Increased expression of nuclear factor of activated T cells 1 drives IL-9–mediated allergic asthma  Sonja Koch, PhD, Anna Graser, PhD, Hooman Mirzakhani,
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
Elisabeth Roesler, MSc, Richard Weiss, PhD, Esther E
Exposure to allergen and diesel exhaust particles potentiates secondary allergen- specific memory responses, promoting asthma susceptibility  Eric B. Brandt,
Protein corona–mediated targeting of nanocarriers to B cells allows redirection of allergic immune responses  Limei Shen, PhD, Stefan Tenzer, PhD, Wiebke.
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,
Antigen-specific effector CD8 T cells regulate allergic responses via IFN-γ and dendritic cell function  Yafang Tang, BSc, Shou Ping Guan, BSc, Benson.
Leukocyte nicotinamide adenine dinucleotide phosphate-reduced oxidase is required for isocyanate-induced lung inflammation  Si-Yen Liu, PhD, Wei-Zhi Wang,
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.
Eosinophils contribute to the resolution of lung-allergic responses following repeated allergen challenge  Katsuyuki Takeda, MD, PhD, Yoshiki Shiraishi,
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.
Surfactant protein D inhibits TNF-α production by macrophages and dendritic cells in mice  László Hortobágyi, MS, Sonja Kierstein, PhD, Kateryna Krytska,
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.
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,
Oliver T. Burton, PhD, Jaciel M. Tamayo, PhD, Amanda J
Frank Kirstein, PhD, Natalie E
Kathleen R. Bartemes, BA, Gail M. Kephart, BS, Stephanie J
Prostaglandin E2 suppresses allergic sensitization and lung inflammation by targeting the E prostanoid 2 receptor on T cells  Zbigniew Zasłona, PhD, Katsuhide.
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.
A fusion protein of flagellin and ovalbumin suppresses the TH2 response and prevents murine intestinal allergy  Stefan Schülke, PhD, Manja Burggraf, MSc,
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,
IL-33 promotes food anaphylaxis in epicutaneously sensitized mice by targeting mast cells  Claire Galand, PhD, Juan Manuel Leyva-Castillo, PhD, Juhan.
Prostaglandin E2 suppresses allergic sensitization and lung inflammation by targeting the E prostanoid 2 receptor on T cells  Zbigniew Zasłona, PhD, Katsuhide.
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
The relative contribution of IL-4 and IL-13 to human IgE synthesis induced by activated CD4+ or CD8+ T cells  Juha Punnonen, MD, PhD, Hans Yssel, PhD,
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,
Effects of established allergen sensitization on immune and airway responses after secondary allergen sensitization  Katharina Blumchen, MD, Kerstin Gerhold,
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,
12/15-Lipoxygenase deficiency protects mice from allergic airways inflammation and increases secretory IgA levels  Amanda R. Hajek, BS, Alexa R. Lindley,
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,
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.
Blocking IL-25 prevents airway hyperresponsiveness in allergic asthma
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
Ovalbumin-specific IgE modulates ovalbumin-specific T-cell response after repetitive oral antigen administration  Nemuko Omata, MD, Yusei Ohshima, MD,
The extra domain A of fibronectin is essential for allergen-induced airway fibrosis and hyperresponsiveness in mice  Martin Kohan, MSc, Andres F. Muro,
Epicutaneous immunization with ovalbumin and CpG induces TH1/TH17 cytokines, which regulate IgE and IgG2a production  Monika Majewska-Szczepanik, PhD,
Corticosteroids enhance CD8+ T cell–mediated airway hyperresponsiveness and allergic inflammation by upregulating leukotriene B4 receptor 1  Hiroshi Ohnishi,
TNF can contribute to multiple features of ovalbumin-induced allergic inflammation of the airways in mice  Susumu Nakae, PhD, Carolina Lunderius, PhD,
Presentation transcript:

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, Steffen Schmitt, PhD, Petra Scholtes, Michael Hausding, PhD, Hans A. Lehr, MD, Laurie H. Glimcher, MD, Susetta Finotto, PhD  Journal of Allergy and Clinical Immunology  Volume 121, Issue 4, Pages 992-999.e6 (April 2008) DOI: 10.1016/j.jaci.2007.12.1172 Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 1 Increased airway lymphocytes, collagen, and AHR in NFATc2(−/−) mice. AHR was measured in wt and NFATc2(−/−) BALB/c mice 6 to 8 weeks old and transpulmonary RI measured 24 hours after the last treatment. Dose-response curves to methacholine (MCH) were obtained after administering indicated doses of intratracheally aerosolized MCH. A, Eight to 10 BALB/c mice per group were sensitized at days 0 and 7 to ovalbumin (OVA)-alum (OVA-sensitized mice) followed by 3 days local treatment with PBS aerosol on days 14, 15, and 16. NFATc2(−/−) OVA-sensitized mice displayed increased AHR at higher doses of MCH compared with wt littermates (n = 11; P = .018, P = .0065, and P = .015 after 3, 10, and 30 mg/mL of 3 minutes MCH aerosol, respectively). B, Increased serum IgE in NFATc2(−/−) mice after OVA sensitization without OVA challenge (n = 4-6; P = .02826). C, Increased peribronchial collagen deposition in NFATc2(−/−) lung by Goldner staining (compare right vs left panel and quantified in far right graph). Eight mice and 3 sections/mouse were analyzed at 400× magnification on a Zeiss microscope (Carl Zeiss, Göttingen, Germany). The images were all taken at the same magnification. The width of the boxes is exactly 200 μm, and the space bar represents 100 μm. ∗P < .05; ∗∗P < .01. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 2 NFATc2(−/−) lung CD8+ T cells produce less IFN-γ but more IL-10 than WT lung CD8+ T cells. A, Intracellular IFN-γ production by lung CD8+ T cells from ovalbumin-sensitized and challenged mice. Left panel, Dot plot analysis of IFN-γ–producing CD8+ T cells; right panel, quantification of IFN-γ+ cells (n = 4; P = .00103 for CD8/IFN-γ). B, ELISA of IFN-γ released by lung and spleen CD8+ T cells (n = 4; P = .028, P = .0065). C,Left panel, ELISA for IL-10 production by lung NFATc2(−/−) CD8+ T cells from ovalbumin-sensitized and unchallenged mice (n = 4; P = .03, P = .04); right panel, IL-10 production is also increased in IFN-γ(−/−) lung CD8+ T cells (n = 4; P = .02, P = .006) as measured by ELISA. ∗P < .05; ∗∗P < .01. D, Neutralization of IL-10 by the indicated concentration of anti–IL-10 antibodies did not influence IFN-γ release by wild-type or NFATc2(−/−) CD8+ T cells. Values of anti–IL-10 antibodies (αIL-10; R & D Systems) are given as μg/mL of medium (IgG was present in the cell culture at the final concentration of 1 μg/mL). Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 3 Increased number of CD8+CD122+ CD127high (IL-7Rα) cells in the lung and spleen of NFATc2(−/−) and IFN-γ(−/−) mice. A, NFATc2(−/−) mice have increased numbers of CD8+CD122+ T cells both in the lung and in the spleen before ovalbumin (OVA) challenge (OVA/PBS) as shown by dot plot analysis (left panel) and depicted quantitatively (right panel; n = 4; P = .0012, P = .036). B, Increased numbers of CD8+CD122+ T cells in lungs and spleen of IFN-γ(−/−) mice (middle panel; n = 4; P = .048, P = .016). C, CD8+CD122+ cells also expressed the IL-7R α chain (CD127) and were thereafter defined as long-lived memory cells. Long-lived memory cells were increased in the lungs of NFATc2(−/−) mice. A representative dot plot is shown in the left panel (quantification is depicted on the right; n = 4; P = .0022, P = .00015). D, The CD8+CD25+ T effector population is downregulated in NFATc2(−/−) lung 24 hours after allergen challenge (n = 4; P = .0078, respectively). E, NFATc2(−/−) CD8+ lung T cells produced less IL-2 compared with those isolated from wild-type mice (n = 4; P = .037). F, NFATc2(−/−) CD8+CD122- lung T cells produced less IFN-γ than wt (n = 4; P = .0068) as shown by FACS analysis before allergen challenge (OVA/PBS). G, Graph depicting the hypothetical sequence of events in the lung NFATc2(−/−) CD8+ T-cell population. In the absence of NFATc2(−/−), CD8+ T cells display changes characteristic of anergic T cells and produce less IL-2 (1). Because IL-2 controls CD25 expression, CD8+CD25+ T cells are also decreased in the absence of NFATc2. This event leads to an expansion of the CD8+CD122+ T-cell population (2), which releases IL-10 (3). Finally, CD8+CD122+ T cells inhibit IFN-γ production by CD8+CD122− T cells (4). ∗P < .05; ∗∗P < .01. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 4 The CD8+CD122+ subset inhibits IFN-γ production in NFATc2(−/−) CD8+CD122- cells and induces AHR on adoptive cotransfer of NFATc2(−/−) CD4+ and CD8+ T cells. A and B, Adoptive transfer of CD8+ T cells from NFATc2(−/−) compared with WT mice increased the AHR generated by NFATc2(−/−) CD4+ T cells (n = 4; P = .028, P= .014). A, The NFATc2(−/−) CD8+ T cells used to reconstitute these SCID mice released significantly less IFN-γ per cell compared with wt CD8+ T cells. The total amount of IFN-γ released by both NFATc2(−/−) CD4+ and NFATc2(−/−) CD8+ T cells (right panel) was significantly lower than wt (left panel). B, AHR is ameliorated by cotransfer of wild-type but exacerbated by cotransfer of NFATc2(−/−) CD8+ T cells. SCID mice were reconstituted with NFATc2(−/−) CD4+ T cells alone or together with either wild-type CD8+ T cells or NFATc2(−/−) CD8+ T cells from OVA-sensitized and challenged mice and AHR measured in reconstituted mice. As reference, the AHR of SCID mice reconstituted with NFATc2(−/−) CD4+ T cells alone is shown. C, Both CD4+ and CD8+ T cells homed to the lung of reconstituted mice as demonstrated by FACS analysis. D, Increased number of eosinophils in SCID mice reconstituted with NFATc2(−/−) CD4+ T cells together with NFATc2(−/−) CD8+ cells (n = 4; P = .036). E and F, Decreased IFN-γ (n = 4; P = .03) and increased IL-17 (n = 4; P = .023) in lungs of SCID mice reconstituted with NFATc2(−/−) CD4+ and CD8+ T cells. BAL, Bronchoalveolar lavage; MCH, methacholine; KO, knockout. ∗P < .05; ∗∗P < .01. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 5 The sorted CD8+CD122+ subset induces AHR on adoptive cotransfer of NFATc2(−/−) CD4+ and CD8+ T cells. A, FACS of splenic CD8+ NFATc2(−/−) T cells that had been depleted of CD8+CD122+ T cells. The 2 resulting cell populations, CD8+CD122+ and CD8+CD122−, were used to reconstitute SCID mice along with CD4+ NFATc2(−/−) T cells. B, SCID mice that received CD8+CD122− NFATc2(−/−) cells but not CD8+CD122+ NFATc2(−/−) cells were protected from AHR. C and D, Decreased numbers of eosinophils and levels of eotaxin in SCID mice reconstituted with NFATc2(−/−) CD4+ T cells together with NFATc2(−/−) CD8+CD122− cells (n = 4; P = .0057 and n = 4-6; P = .0344) E, Increased IFN-γ levels in BALF of SCID mice reconstituted as described on depletion of CD8+CD122+ T cells before transfer (n = 4; P = .0298). F, Lung CD8+ T cells from both wt and NFATc2(−/−) mice were depleted of CD8+CD122+ T cells, and the 4 resulting cell populations CD8+CD122+ and CD8+CD122−, were cultured with anti-CD3 and anti-CD3/CD28 overnight and the supernatant analyzed for IFN-γ by ELISA (n = 5-9; P = .04, P = .005). As shown, the inhibition of IFN-γ production by CD8+CD122− T cells was reversed by CD8+CD122+ T-cell depletion. MCH, Methacholine; FL, fluorescence. ∗P < .05; ∗∗P < .01; ∗∗∗P < .001. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Left, Naive mice lacking NFATc2 show comparable AHR to the wild-type littermates (n = 5). Right, Addition of different amounts of recombinant IL-10 (R&D Systems, Minneapolis, Minn) did not alter IFN-γ levels in lung CD8+ T cells isolated from unchallenged mice (n = 4). MCH, Methacholine. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Upper lanes, Ovalbumin (OVA) challenge after OVA sensitization in NFATc2(−/−) mice led to increased numbers of CD8+CD122+ T cells compared with wild-type littermates (n = 4; P = .012, P = .037). Left, Isotype control. Lower lanes, OVA challenge after OVA sensitization in NFATc2(−/−) mice led to increased numbers of CD8+CD122+CD127+ T cells compared with wild-type littermates (n = 4; P = .0022). Left, Isotype control. FCS, Forward scatter; ISO, isotype control; FL, fluorescence. ∗P < .05; ∗∗∗P < .001. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

CD8+CD25+ cells were decreased in NFATc2(−/−) mice after ovalbumin (OVA) sensitization (n = 4; P = .0078) and OVA sensitization and OVA challenge (n = 4; P = .0027). ∗∗P < .01. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Increased number of CD8+CD122+ lung T cells expressing intracellular IL-10 in NFATc2(−/−) and IFN-γ(−/−) mice. Increased numbers of CD8+CD122+ lung T cells from NFATc2(−/−) mice (n = 4; P = .037, P = .011; upper panels) and IFN-γ(−/−) mice (n = 4; P = .013; lower panels) after allergen challenge as compared to the number of CD8+CD122+ T cells isolated from the NFATc2(+/+) and IFN-γ(+/+) mice, respectively. These cells express intracellular IL-10 as shown by FACS analysis. ∗P < .05. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions

SCID mice reconstituted with either wild-type or NFATc2(−/−) CD8+ T cells (1.5 × 106 CD8+ T cells) do not show increased AHR (n = 5). MCH, Methacholine; OVA, ovalbumin; KO, knockout. Journal of Allergy and Clinical Immunology 2008 121, 992-999.e6DOI: (10.1016/j.jaci.2007.12.1172) Copyright © 2008 American Academy of Allergy, Asthma & Immunology Terms and Conditions