Repeated FcεRI triggering reveals modified mast cell function related to chronic allergic responses in tissue  Jolien Suurmond, MSc, Kim L.L. Habets,

Slides:



Advertisements
Similar presentations
Multiple-checkpoint inhibition of thymic stromal lymphopoietin–induced TH2 response by TH17-related cytokines  Sofia I. Bogiatzi, BSc, Maude Guillot-Delost,
Advertisements

MicroRNA signature in patients with eosinophilic esophagitis, reversibility with glucocorticoids, and assessment as disease biomarkers  Thomas X. Lu,
Triggering of specific Toll-like receptors and proinflammatory cytokines breaks allergen- specific T-cell tolerance in human tonsils and peripheral blood 
Dorothea Dijkstra, MSc, Rob Leurs, PhD, Paul Chazot, PhD, Fiona C
Early suppression of basophil activation during allergen-specific immunotherapy by histamine receptor 2  Natalija Novak, MD, Nihal Mete, MD, Caroline.
Changes in markers associated with dendritic cells driving the differentiation of either TH2 cells or regulatory T cells correlate with clinical benefit.
Barbara Stanic, PhD, Willem van de Veen, PhD, Oliver F
Defective calcium signaling and disrupted CD20–B-cell receptor dissociation in patients with common variable immunodeficiency disorders  Annick A.J.M.
CD4+CD25+ regulatory T cells suppress contact hypersensitivity reactions through a CD39, adenosine-dependent mechanism  Sabine Ring, PhD, Stephen J. Oliver,
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
Ryan H. Dougherty, MD, Sukhvinder S
The activating protein 1 transcription factor basic leucine zipper transcription factor, ATF- like (BATF), regulates lymphocyte- and mast cell–driven immune.
Antigen-driven basophil activation is indicative of early Necator americanus infection in IgE-seronegative patients  Franco H. Falcone, PhD, Gary Telford,
IgE cross-linking impairs monocyte antiviral responses and inhibits influenza-driven TH1 differentiation  Regina K. Rowe, MD, PhD, David M. Pyle, MD,
Differential cytokine induction by the human skin–associated autoallergen thioredoxin in sensitized patients with atopic dermatitis and healthy control.
Oral immunotherapy induces IgG antibodies that act through FcγRIIb to suppress IgE- mediated hypersensitivity  Oliver T. Burton, PhD, Stephanie L. Logsdon,
MicroRNA signature in patients with eosinophilic esophagitis, reversibility with glucocorticoids, and assessment as disease biomarkers  Thomas X. Lu,
Frank Kirstein, PhD, Natalie E
Assessing basophil activation by using flow cytometry and mass cytometry in blood stored 24 hours before analysis  Kaori Mukai, PhD, Nicolas Gaudenzio,
Inhibition of differentiation, amplification, and function of human TH17 cells by intravenous immunoglobulin  Mohan S. Maddur, DVM, Janakiraman Vani,
Dawn C. Newcomb, PhD, Madison G
Human dendritic cell subset 4 (DC4) correlates to a subset of CD14dim/−CD16++ monocytes  Federica Calzetti, BS, Nicola Tamassia, PhD, Alessandra Micheletti,
Human IL-31 is induced by IL-4 and promotes TH2-driven inflammation
Clara cell 16-kd protein downregulates TH2 differentiation of human naive neonatal T cells  Sofi Johansson, MSc, Göran Wennergren, MD, PhD, Nils Åberg,
Frank Kirstein, PhD, Natalie E
Small RNA profiling reveals deregulated phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinase (PI3K)/Akt pathway in bronchial smooth muscle.
Miriam Wittmann, MD, Jana Zeitvogel, Dong Wang, MD, Thomas Werfel, MD 
Jerome A. Sigua, MD, Becky Buelow, MD, Dorothy S
Food allergy herbal formula 2 protection against peanut anaphylactic reaction is via inhibition of mast cells and basophils  Ying Song, MD, Chunfeng Qu,
Thymic stromal lymphopoietin converts human epidermal Langerhans cells into antigen- presenting cells that induce proallergic T cells  Susanne Ebner, PhD,
Human mast cells drive memory CD4+ T cells toward an inflammatory IL-22+ phenotype  Nicolas Gaudenzio, PhD, Camille Laurent, MD, Salvatore Valitutti,
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.
Inhibition of human B-cell development into plasmablasts by histone deacetylase inhibitor valproic acid  Anne-Kathrin Kienzler, MSc, Marta Rizzi, MD,
Multiple-checkpoint inhibition of thymic stromal lymphopoietin–induced TH2 response by TH17-related cytokines  Sofia I. Bogiatzi, BSc, Maude Guillot-Delost,
IL-33 promotes food anaphylaxis in epicutaneously sensitized mice by targeting mast cells  Claire Galand, PhD, Juan Manuel Leyva-Castillo, PhD, Juhan.
Targeting allergen to FcγRI reveals a novel TH2 regulatory pathway linked to thymic stromal lymphopoietin receptor  Kathryn E. Hulse, PhD, Amanda J. Reefer,
Superior anti-inflammatory effects of narrow-spectrum kinase inhibitors in airway smooth muscle cells from subjects with chronic obstructive pulmonary.
Jill A. Poole, MD, Neil E. Alexis, PhD, Conrad Parks, BS, Amy K
Mammalian target of rapamycin inhibition counterbalances the inflammatory status of immune cells in patients with chronic granulomatous disease  Aurélie.
Notch signaling confers antigen-presenting cell functions on mast cells  Nobuhiro Nakano, PhD, Chiharu Nishiyama, PhD, Hideo Yagita, PhD, Akemi Koyanagi,
Yilin Qi, MSc, Darwin J. Operario, PhD, Christopher M
The mannose receptor negatively modulates the Toll-like receptor 4–aryl hydrocarbon receptor–indoleamine 2,3-dioxygenase axis in dendritic cells affecting.
Staphylococcal exotoxins are strong inducers of IL-22: A potential role in atopic dermatitis  Margarete Niebuhr, MD, Helena Scharonow, MS, Merle Gathmann,
Josée Lamoureux, PhD, Jana Stankova, PhD, Marek Rola-Pleszczynski, MD 
IL-9 is a key component of memory TH cell peanut-specific responses from children with peanut allergy  Helen A. Brough, MSc, FRCPCH, David J. Cousins,
Oktay Kirak, MD, Gert Riethmüller, MD 
IL-17E upregulates the expression of proinflammatory cytokines in lung fibroblasts  Séverine Létuvé, PhD, Stéphane Lajoie-Kadoch, MSc, Séverine Audusseau,
Allergen-specific CD8+ T cells in peanut-allergic individuals
Matthew J. Loza, PhD, Susan Foster, PhD, Stephen P
Sara Paveglio, PhD, MS, Erin Bennett, MS, Kelly L. Hawley, PhD, Adam P
Sputum inflammatory cells from patients with allergic rhinitis and asthma have decreased inflammasome gene expression  Willie June Brickey, PhD, Neil.
Allergen-specific immunotherapy modulates the balance of circulating Tfh and Tfr cells  Véronique Schulten, PhD, Victoria Tripple, BSc, Grégory Seumois,
Impairment of T-regulatory cells in cord blood of atopic mothers
Toll-like receptor 2 ligands activate human basophils for both IgE-dependent and IgE- independent secretion  Anja P. Bieneman, BS, Kristin L. Chichester,
Duy Pham, PhD, Sarita Sehra, PhD, Xin Sun, PhD, Mark H. Kaplan, PhD 
Jenny Seltmann, PhD, Lennart M
Expression of chemokines and chemokine receptors in lesional and nonlesional upper skin of patients with atopic dermatitis  Eva Gros, MSc, Caroline Bussmann,
The mannose receptor negatively modulates the Toll-like receptor 4–aryl hydrocarbon receptor–indoleamine 2,3-dioxygenase axis in dendritic cells affecting.
CCL17/thymus and activation-regulated chemokine induces calcitonin gene–related peptide in human airway epithelial cells through CCR4  Kandace Bonner,
Dorothea Dijkstra, MSc, Rob Leurs, PhD, Paul Chazot, PhD, Fiona C
Defective calcium signaling and disrupted CD20–B-cell receptor dissociation in patients with common variable immunodeficiency disorders  Annick A.J.M.
IL-10–producing monocytes differentiate to alternatively activated macrophages and are increased in atopic patients  Antje Prasse, MD, Martin Germann,
Julie Negri, BA, S. Brandon Early, BA, John W
Thymic stromal lymphopoietin does not activate human basophils
Soybean isoflavones regulate dendritic cell function and suppress allergic sensitization to peanut  Madhan Masilamani, PhD, John Wei, BA, Shiven Bhatt,
IgG4 production is confined to human IL-10–producing regulatory B cells that suppress antigen-specific immune responses  Willem van de Veen, MSc, Barbara.
Large-scale gene expression profiling reveals distinct type 2 inflammatory patterns in chronic rhinosinusitis subtypes  Matthew A. Tyler, MD, Chris B.
Induction of anergic allergen-specific suppressor T cells using tolerogenic dendritic cells derived from children with allergies to house dust mites 
CCL17/thymus and activation-regulated chemokine induces calcitonin gene–related peptide in human airway epithelial cells through CCR4  Kandace Bonner,
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:

Repeated FcεRI triggering reveals modified mast cell function related to chronic allergic responses in tissue  Jolien Suurmond, MSc, Kim L.L. Habets, PhD, Zuotian Tatum, MSc, Joris J. Schonkeren, BASc, Peter A.C.'t Hoen, PhD, Tom W.J. Huizinga, MD, PhD, Jeroen F.J. Laros, PhD, René E.M. Toes, PhD, Fina Kurreeman, PhD  Journal of Allergy and Clinical Immunology  Volume 138, Issue 3, Pages 869-880 (September 2016) DOI: 10.1016/j.jaci.2016.01.017 Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 1 Repeated mast cell stimulation through FcεRI. A, Experimental setup of repeated stimulation of mast cells. Human cord blood–derived mast cells were stimulated for 2 weeks with 0.2 μg/mL anti-IgE (Repeated αIgE) or control medium (Naive). Twenty-four hours before each stimulation, mast cells were sensitized with human IgE. At the end of 2 weeks, mast cells were either left untreated (N and R) or stimulated with anti-IgE (N+αIgE and R+αIgE). B, Representative flow cytometric plots for CD63 of naive mast cells (N) or mast cells 3 hours after their first anti-IgE stimulation (R), showing degranulation in response to the first anti-IgE stimulation on day 0. PE, Phycoerythrin. C, Summary of surface CD63 expression as in Fig 1, B, and IL-8 secretion 24 hours after the first anti-IgE stimulation or control medium. MC, Mast cells. D and E, Surface CD63 and CD203c expression in response to 24 hours of anti-IgE stimulation in naive mast cells or mast cells exposed to 2 weeks of stimulation (Repeated αIgE). CD63 staining as in Fig 1, C, is shown, and the CD203c median fluorescence intensity (MFI) ratio is calculated as a ratio of the MFI of CD203c staining divided by the MFI of the isotype control for each individual sample. Data are represented as means ± SEMs from 3 (Fig 1, C) or 6 (Fig 1, D and E) independent experiments. Numbers indicate that no significant differences were found between naive mast cells (N) and cells exposed for 2 weeks to anti-IgE (R) by using the paired t test. Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 2 Dampened genes after repeated anti-IgE. A, Gene expression determined by using RNA sequencing in response to a single 6-hour anti-IgE stimulation (N+αIgE) compared with that seen in control mast cells (N). Log fold change (LogFC) relative to naive (N) mast cells of the differentially expressed genes is shown. B, Genes that were significantly upregulated after a single anti-IgE stimulation were selected as shown in Fig 2, A, and compared with their upregulation after repeated stimulation (R+αIgE). LogFC values were calculated compared with unstimulated mast cells at the end of the 2-week stimulation (N+αIgE/N and R+αIgE/R). C, Genes that were significantly upregulated after a single anti-IgE stimulation were selected as shown in Fig 2, A, where each dot indicates a different gene. Those genes significantly upregulated (top) and tolerized (bottom) after repeated anti-IgE (R+αIgE) compared with a single anti-IgE (N+αIgE) stimulation are indicated. Data are shown as fold change relative to N+αIgE. D, Genes that are significantly downregulated after repeated anti-IgE stimulation compared with a single anti-IgE stimulation, as determined by using differential expression analysis. Each line represents a single gene, and data are shown as relative reads per kilobase per million (RPKM) normalized to single anti-IgE stimulation (N+αIgE). E and H, Gene expression of CSF2 and CCL4 determined by using RNA sequencing. Data are shown as RPKM fold change relative to single anti-IgE stimulation (N+αIgE). F and I, Gene expression of CSF2 and CCL4 determined by using qPCR. Data are shown as relative expression normalized to the housekeeping gene RPL5 as a fold change to single anti-IgE stimulation (N+αIgE). G and J, Protein levels of CSF2 (GM-CSF) and CCL4 (macrophage inflammatory protein 1β [MIP-1β]) in supernatant 24 hours after stimulation with anti-IgE of naive mast cells or mast cells that were exposed to anti-IgE for 2 weeks. K, Expression of genes in the arachidonic acid (AA) metabolism pathway determined by using RNA sequencing. Log fold change (LogFC) relative to naive (N) mast cells of the differentially expressed genes is shown. L and M, Protein levels of leukotriene C4 (LTC4) and prostaglandin D2 (PGD2) in the supernatant 1 hour after stimulation with anti-IgE of naive mast cells or mast cells that were exposed to anti-IgE for 2 weeks. Data are represented as means ± SEMs from 3 (Fig 2, B, E, H, and K), 5 (Fig 2, F, G, I, and J), or 6 (Fig 2, L and M) independent experiments. Asterisks indicate significant differences determined by using the paired t test (P < .05; Fig 2, B), a differential expression analysis false discovery rate of less than .05 (Fig 2, C, E, and H), or repeated-measures ANOVA with the Bonferroni post hoc test (P < .05; Fig 2, F, G, I, L, and M). Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 3 Persistent upregulation of genes after repeated anti-IgE stimulation. A, Overlap of genes upregulated after repeated anti-IgE stimulation without an additional stimulation (R > N) or with an additional stimulation (R+αIgE > N+αIgE), as determined by using differential expression analysis. B, Heat map showing relative expression of all genes in Fig 3, A. Each row represents a single gene. Relative expression in 3 independent experiments is shown. C, E, and G, Gene expression of TREM2, OLFM4, and CCL18, as obtained by using RNA sequencing. Data are shown as reads per kilobase per million (RPKM) fold change relative to naive mast cells (N). D, F, and H, Gene expression of TREM2, OLFM4, and CCL18, as determined by using qPCR. Data are shown as relative expression normalized to the housekeeping gene RPL5 as a fold change to naive mast cells (N). I, Protein levels of CCL18 in the supernatant 24 hours after stimulation with anti-IgE of naive mast cells or mast cells that were exposed to anti-IgE for 2 weeks. J, Protein levels of CCL18 in the supernatant 6 hours after stimulation with anti-IgE of naive mast cells (left) or 2 weeks after stimulation with repeated anti-IgE or control medium (right). Data are represented as means ± SEMs from 3 (Fig 3, C, E, G, and J), 5 (Fig 3, D, F, and H), or 4 (Fig 3, I) independent experiments. Asterisks indicate significant differences determined by using a differential expression analysis false discovery rate of less than 0.05 (Fig 3, C, E, and G), repeated-measures ANOVA with the Bonferroni post hoc test (P < .05; Fig 3, D, F, H, and I), or the paired t test (P < .05; Fig 3, J). N, Naive mast cells; N+αIgE, single 6-hour stimulation of naive mast cells with anti-IgE; R, repeated anti-IgE stimulation for 2 weeks; R+αIgE, 6-hour stimulation with anti-IgE after repeated anti-IgE stimulation. Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 4 Overlap in genes and enriched pathways between repeated and single stimulation of mast cells. A and B, Interaction network of GO terms enriched in genes upregulated after repeated anti-IgE (Fig 4, A) or single anti-IgE (Fig 4, B) stimulation. Node size reflects the significance/number of genes, and line thickness indicates the connectivity between 2 GO terms. Pathways that are unique for either of the gene sets are highlighted in red. C and D, Protein expression of HLA-DR determined by using flow cytometry. Data are shown as Δ median fluorescence intensity (MFI) of HLA-DR compared with the isotype control. Data are represented as means ± SEMs from 5 (Fig 4, C) independent experiments. Asterisks indicate significant differences determined by using repeated-measures ANOVA with the Bonferroni post hoc test (P < .05; Fig 4, C). Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 5 Autocrine signaling and transcription factors. A-F, Mast cells were stimulated for 2 weeks with 0.2 μg/mL anti-IgE (Repeated αIgE) or control medium (Naive) through direct stimulation (left) or using the supernatant from activated mast cells collected 24 hours after activation (right). Gene expression of CCL4, CSF2, EMR3, TMEM45B, TREM2, and CCL18 determined by using qPCR. Data are shown as relative expression normalized to the housekeeping gene RPL5 as a fold change to cells stimulated for 6 hours with anti-IgE (CCL4, CSF2, EMR3, and TMEM45B) or naive mast cells (TREM2 and CCL18). G and H, RNA expression analysis of the transcription factors BATF (Fig 5, G) and RXRA (Fig 5, H) and their target genes as identified by using Cscan enrichment analysis. Expression of these genes in mast cells exposed to repeated anti-IgE stimulation is shown as fold change relative to naive mast cells. Data are represented as means ± SEMs from 4 independent experiments (Fig 5, A-F) or from 3 independent experiments (Fig 5, G and H). Asterisks indicate significant differences determined by using repeated-measures ANOVA with the Bonferroni post hoc test (P < .05). Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions

Fig 6 Association of upregulated genes with chronic allergy. A and C, Heat map showing relative expression of genes upregulated in patients with atopic allergy or those with psoriasis in mast cells exposed to repeated anti-IgE stimulation. B and D, Overlap of genes upregulated in patients with atopic allergy or those with psoriasis with random genes (top), genes most highly expressed by mast cells (second row), genes upregulated in mast cells after a single anti-IgE stimulus (third row), or genes upregulated in mast cells after repeated anti-IgE (bottom). Overlap was calculated as a percentage of the mast cell or random gene list. Asterisks indicate significant differences determined by using the Fisher exact test (P < .05; Fig 6, B and D). Journal of Allergy and Clinical Immunology 2016 138, 869-880DOI: (10.1016/j.jaci.2016.01.017) Copyright © 2016 American Academy of Allergy, Asthma & Immunology Terms and Conditions