Luise Westernberg, PhD, Véronique Schulten, PhD, Jason A

Slides:



Advertisements
Similar presentations
Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy  Erik Wambre, PhD, Jonathan.
Advertisements

Catherine M. Counsell, MAa, Julian F. Bond, PhDa, John L
Volume 11, Pages (September 2016)
Comparison of the immunogenicity of BM32, a recombinant hypoallergenic B cell epitope–based grass pollen allergy vaccine with allergen extract–based vaccines 
Journal of Allergy and Clinical Immunology
Identification of drug- and drug-metabolite immune responses originating from both naive and memory T cells  Andrew Gibson, MRes, Lee Faulkner, PhD, Sally.
Cor a 1–reactive T cells and IgE are predominantly cross-reactive to Bet v 1 in patients with birch pollen–associated food allergy to hazelnut  Claudia.
Kuan-Wei Chen, PhD, Katharina Blatt, MSc, Wayne R
Induction of anergic allergen-specific suppressor T cells using tolerogenic dendritic cells derived from children with allergies to house dust mites 
Reduced TH1/TH17 CD4 T-cell numbers are associated with impaired purified protein derivative–specific cytokine responses in patients with HIV-1 infection 
Persistence and evolution of allergen-specific IgE repertoires during subcutaneous specific immunotherapy  Mattias Levin, PhD, Jasmine J. King, BS, Jacob.
Identification and gene cloning of a new major allergen Cha o 3 from Chamaecyparis obtusa (Japanese cypress) pollen  Toshihiro Osada, MSc, Takafumi Harada,
Designing hypoallergenic derivatives for allergy treatment by means of in silico mutation and screening  Theresa Thalhamer, PhD, Heidi Dobias, MSc, Tatjana.
IgE cross-linking impairs monocyte antiviral responses and inhibits influenza-driven TH1 differentiation  Regina K. Rowe, MD, PhD, David M. Pyle, MD,
Volume 25, Issue 2, Pages (February 2017)
Determination of allergen specificity by heavy chains in grass pollen allergen–specific IgE antibodies  Elisabeth Gadermaier, PhD, Sabine Flicker, PhD,
Catherine M. Counsell, MAa, Julian F. Bond, PhDa, John L
Cor a 1–reactive T cells and IgE are predominantly cross-reactive to Bet v 1 in patients with birch pollen–associated food allergy to hazelnut  Claudia.
Human CD19 and CD40L deficiencies impair antibody selection and differentially affect somatic hypermutation  Menno C. van Zelm, PhD, Sophinus J.W. Bartol,
Development and characterization of a recombinant, hypoallergenic, peptide-based vaccine for grass pollen allergy  Margarete Focke-Tejkl, PhD, Milena.
Blocking antibodies induced by immunization with a hypoallergenic parvalbumin mutant reduce allergic symptoms in a mouse model of fish allergy  Raphaela.
Magdalena A. Berkowska, PhD, Jorn J
Correlation of IgE/IgG4 milk epitopes and affinity of milk-specific IgE antibodies with different phenotypes of clinical milk allergy  Julie Wang, MD,
Malassezia sympodialis thioredoxin–specific T cells are highly cross-reactive to human thioredoxin in atopic dermatitis  Hari Balaji, MSc, Annice Heratizadeh,
Comparison of the immunogenicity of BM32, a recombinant hypoallergenic B cell epitope–based grass pollen allergy vaccine with allergen extract–based vaccines 
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
Characterization of human memory CD4+ T-cell responses to the dog allergen Can f 4  Aino L. Rönkä, MD, Tuure T. Kinnunen, MD, PhD, Amélie Goudet, BSc,
Classification of common variable immunodeficiencies using flow cytometry and a memory B-cell functionality assay  Amelia L. Rösel, MD, Carmen Scheibenbogen,
Jug r 2–reactive CD4+ T cells have a dominant immune role in walnut allergy  Luis Diego Archila, MSc, David Jeong, MD, Mariona Pascal, PhD, Joan Bartra,
Direct monitoring of basophil degranulation by using avidin-based probes  Régis Joulia, PhD, Claire Mailhol, MD, Salvatore Valitutti, MD, Alain Didier,
Staphylococcus aureus fibronectin-binding protein specifically binds IgE from patients with atopic dermatitis and requires antigen presentation for cellular.
Kathleen R. Bartemes, BA, Gail M. Kephart, BS, Stephanie J
Mohamed H. Shamji, PhD, Janice A. Layhadi, MSc, Guy W
Targeting Fel d 1 to FcγRI induces a novel variation of the TH2 response in subjects with cat allergy  Kathryn E. Hulse, BS, Amanda J. Reefer, MS, Victor.
Naturally processed T cell–activating peptides of the major birch pollen allergen  Sonja Mutschlechner, PhD, Matthias Egger, PhD, Peter Briza, PhD, Michael.
Dissection of the IgE and T-cell recognition of the major group 5 grass pollen allergen Phl p 5  Margarete Focke-Tejkl, PhD, Raffaela Campana, PhD, Renate.
Allison K. Martin, BS, Douglas G. Mack, PhD, Michael T
Chronic cat allergen exposure induces a TH2 cell–dependent IgG4 response related to low sensitization  Amedee Renand, PhD, Luis D. Archila, MSc, John.
Suppression of the basophil response to allergen during treatment with omalizumab is dependent on 2 competing factors  Donald W. MacGlashan, MD, PhD,
T-cell receptor contact and MHC binding residues of a major rye grass pollen allergen T- cell epitope  Matthew D. Burton, BSC, Hons, Bella Blaher, PhD,,
Peanut-specific type 1 regulatory T cells induced in vitro from allergic subjects are functionally impaired  Laurence Pellerin, PhD, Jennifer Anne Jenks,
Identification of drug- and drug-metabolite immune responses originating from both naive and memory T cells  Andrew Gibson, MRes, Lee Faulkner, PhD, Sally.
Characterization of the allergic T-cell response to Pru p 3, the nonspecific lipid transfer protein in peach  Véronique Schulten, MSc, Astrid Radakovics,
Role of IL-35 in sublingual allergen immunotherapy
Genetically engineered hybrid proteins from Parietaria judaica pollen for allergen- specific immunotherapy  Roberto González-Rioja, PhD, Ignacio Ibarrola,
Peanut T-cell epitope discovery: Ara h 1
Grass tablet sublingual immunotherapy downregulates the TH2 cytokine response followed by regulatory T-cell generation  Abel Suárez-Fueyo, PhD, Tania.
Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy  Erik Wambre, PhD, Jonathan.
Antigen presentation of the immunodominant T-cell epitope of the major mugwort pollen allergen, Art v 1, is associated with the expression of HLA-DRB1∗01 
Kuan-Wei Chen, PhD, Katharina Blatt, MSc, Wayne R
Spatial clustering of the IgE epitopes on the major timothy grass pollen allergen Phl p 1: Importance for allergenic activity  Sabine Flicker, PhD, Peter.
Different IgE recognition of mite allergen components in asthmatic and nonasthmatic children  Yvonne Resch, MSc, Sven Michel, MSc, Michael Kabesch, MD,
Skin test evaluation of a novel peptide carrier–based vaccine, BM32, in grass pollen– allergic patients  Verena Niederberger, MD, Katharina Marth, MD,
Reshaping the Bet v 1 fold modulates TH polarization
Allergen-specific CD8+ T cells in peanut-allergic individuals
Association between specific timothy grass antigens and changes in TH1- and TH2-cell responses following specific immunotherapy  Véronique Schulten, PhD,
A hypoallergenic cat vaccine based on Fel d 1–derived peptides fused to hepatitis B PreS  Katarzyna Niespodziana, MSc, Margarete Focke-Tejkl, PhD, Birgit.
Allergen-specific immunotherapy modulates the balance of circulating Tfh and Tfr cells  Véronique Schulten, PhD, Victoria Tripple, BSc, Grégory Seumois,
Inhibition of human allergic T-cell responses by IL-10–treated dendritic cells: Differences from hydrocortisone-treated dendritic cells  Iris Bellinghausen,
CCL17/thymus and activation-regulated chemokine induces calcitonin gene–related peptide in human airway epithelial cells through CCR4  Kandace Bonner,
Microarray immunoassay: Association of clinical history, in vitro IgE function, and heterogeneity of allergenic peanut epitopes  Wayne G. Shreffler, MD,
A bioinformatics approach to identify patients with symptomatic peanut allergy using peptide microarray immunoassay  Jing Lin, PhD, Francesca M. Bruni,
Gastrointestinal digestion of Bet v 1–homologous food allergens destroys their mediator- releasing, but not T cell–activating, capacity  Eva Maria Schimek,
Carrier-bound, nonallergenic Ole e 1 peptides for vaccination against olive pollen allergy  Teresa E. Twaroch, MSc, Margit Focke, PhD, Vera Civaj, Milena.
Allergy multivaccines created by DNA shuffling of tree pollen allergens  Michael Wallner, PhD, Angelika Stöcklinger, MSc, Theresa Thalhamer, MSc, Barbara.
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,
Invariant natural killer T cells from children with versus without food allergy exhibit differential responsiveness to milk-derived sphingomyelin  Soma.
Volume 25, Issue 2, Pages (February 2017)
Presentation transcript:

T-cell epitope conservation across allergen species is a major determinant of immunogenicity  Luise Westernberg, PhD, Véronique Schulten, PhD, Jason A. Greenbaum, PhD, Sara Natali, Victoria Tripple, BS, Denise M. McKinney, April Frazier, PhD, Heidi Hofer, PhD, Michael Wallner, PhD, Federica Sallusto, PhD, Alessandro Sette, PhD, Bjoern Peters, PhD  Journal of Allergy and Clinical Immunology  Volume 138, Issue 2, Pages 571-578.e7 (August 2016) DOI: 10.1016/j.jaci.2015.11.034 Copyright © 2016 The Authors Terms and Conditions

Fig 1 Conservation of timothy grass proteins across other pollens. A, Six hundred forty-eight peptides from IgE-reactive P pratense allergens listed by IUIS were examined for their conservation in other pollen species. A peptides was considered conserved in another pollen if its transcriptome encoded the peptide or a variant with up to 2 amino acid substitutions. The percentage of peptides conserved in all 4 additional grass transcriptomes (pangrass) is indicated by light gray columns, and the percentage of peptides conserved in all 10 pollens (panpollen) is indicated by dark gray columns. Antigens are sorted based on pangrass conservation from low to high. B and C, The pangrass (Fig 1, B) and panpollen (Fig 1, C) conservation of IUIS allergens was then compared with that of proteins identified in P pratense pollen based on a transcriptomic and proteomic analysis. Medians and quantile ranges are indicated by boxes and error bars. P pratense pollen proteins that were not recognized by either T-cell or B-cell responses were less conserved than both IUIS allergens and TGTAs. Asterisks indicate statistically significant differences: P < .05, 1-tailed Mann-Whitney test. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig 2 Peptide conservation correlates with immunogenicity. Panels of peptides from timothy grass proteins were previously tested for the ability to induce IL-5 responses in PBMCs from allergic patients after in vitro culture with timothy grass extract in 2 separate cohorts. Peptides from each study were separated into sets based on the number of pollen species in which they were conserved (x-axis). For each set, the average frequency of T-cell responses was calculated and normalized to 1.0 for each study (y-axis). Data for peptides derived from IUIS allergens are shown as blue diamonds, whereas peptides derived from TGTA antigens are shown as red boxes, and averages of the 2 are shown as black circles. The line depicts a linear correlation for the averaged data, which is highly significant with an r2 value of 0.796. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig 3 Conservation in the transcriptome predicts peptide cross-reactivity. For each peptide, donors with P pratense allergy were selected who reacted to the peptide after expanding PBMCs in vitro with P pratense extract. PBMCs were stimulated with individual peptides for 14 days, and IL-5 responses were measured by using an ELISpot to (1) the peptide itself, (2) P pratense extract, (3) the 9 other extracts for which the transciptomes were sequenced, and (4) peptide pools that did or did not contain the peptide as relevant and irrelevant controls. T-cell cultures that did not induce a robust response of greater than 200 spot-forming cells/million to the peptide itself were excluded (30% of cultures). Reponses to extracts and peptide pools are expressed as the relative fraction of the response to the peptide itself and capped at 100%. Each gray bar represents the average response ± SEM. Asterisks indicate P values of statistical significance, as indicated on the right according to 1-tailed Mann-Whitney tests. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig 4 T-cell clone cross-reactivity. Donor 1583 was cultured with peptide P5 and then restimulated with that same (relevant) peptide, an irrelevant peptide control, and peptides from the 4 grasses (green), 4 trees (blue), and 2 weeds (red). A, ELISpot data from the P5 cultured cell line. B-E, Proliferation of 4 representative T-cell clones derived from the same patient. SI was calculated by dividing cpm of antigen stimulated wells with cpm of unstimulated wells. Conditions with SI>2 were considered positive (∗). Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig 5 Schematic representation of allergen cross-reactive versus monospecific T-cell epitope recognition. Allergens 1 and 2 contain a conserved T-cell epitope. Therefore cross-reactive T cells that respond to this epitope will be primed more frequently (cross-reactive T-cell in red, top) than T cells that are specific for an epitope unique to a single allergen (allergen monospecific T cell in blue, bottom). Consequently, cross-reactive T cells can provide T-cell help to B cells that are specific to different allergens and promote antibody production/isotype switching to IgE. Importantly, both B cells that produce cross-reactive IgE and B cells that produce monospecific IgE can receive help from cross-reactive T cells as long as they process and present a conserved T-cell epitope. APC, Antigen-presenting cells. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig E1 Conservation of peptides in different timothy grass pollen proteins. A, Conservation of the 13 TGTAs. B, Conservation of timothy grass pollen proteins that were unreactive based on both T-cell and B-cell responses. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig E2 Cytokine release by T cells after 2 weeks of peptide culture. Shown are IFN-γ and IL-5 release by T cells restimulated either with the peptide with which they were originally cultured or restimulated with a relevant peptide pool or an irrelevant peptide pool or timothy grass extract. Each bar represents the average spot-forming cells/million (SFC) ± SEM. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig E3 Separate analysis of correlation between cross-reactivity and conservation for IUIS allergen peptides and TGTA peptides. The analysis performed is the same as in Fig 3 but splits out peptides from known IUIS allergens versus TGTA proteins. *P < .05, **P < .01, ***P < .005, and ****P < .001. n.s., Not significant. Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions

Fig E4 A, Additional T-cell clones generated from the same culture (with P pratense peptide P5) as shown in Fig 4. B, ELISpot data from the cell line (top) and proliferation from individual T-cell clones (bottom) derived from another culture with P pratense peptide P7. SI was calculated by dividing cpm of antigen stimulated wells with cpm of unstimulated wells. Conditions with SI>2 were considered positive (∗). Journal of Allergy and Clinical Immunology 2016 138, 571-578.e7DOI: (10.1016/j.jaci.2015.11.034) Copyright © 2016 The Authors Terms and Conditions