Effect of anti-IgE therapy on food allergen specific T cell responses in eosinophil associated gastrointestinal disorders

Barbara Foster, Shabnam Foroughi, Yuzhi Yin, Calman Prussin, Barbara Foster, Shabnam Foroughi, Yuzhi Yin, Calman Prussin

Abstract

Background: Anti-IgE therapy inhibits mast cell and basophil activation, blocks IgE binding to both FcεRI and CD23 and down regulates FcεRI expression by antigen (Ag) presenting cells (APCs). In addition to its classical role in immediate hypersensitivity, IgE has been shown in vitro to facilitate Ag presentation of allergens, whereby APC bound IgE preferentially takes up allergens for subsequent processing and presentation. The purpose of this study was to determine whether anti-IgE therapy, by blocking facilitated Ag presentation in vivo, attenuates allergen specific Th2 cell responses.

Methods: To test this hypothesis, food allergen specific T cell responses were examined during a 16-week clinical trial of omalizumab in nine subjects with eosinophilic gastroenteritis and food sensitization. Allergen specific T cell responses were measured using carboxyfluorescein succinimidyl ester dye dilution coupled with intracellular cytokine staining and polychromatic flow cytometry. Four independent indices of allergen specific T cell response (proliferation, Ag dose response, precursor frequency, and the ratio of Th2:Th1 cytokine expression) were determined.

Results: Eight of the 9 subjects had measurable food allergen specific responses, with a median proliferation index of 112-fold. Allergen specific T cell proliferation was limited to CD4 T cells, whereas CD8 T cell did not proliferate. Food allergen specific responses were Th2 skewed relative to tetanus specific responses in the same subjects. In contradistinction to the original hypothesis, anti-IgE treatment did not diminish any of the four measured indices of allergen specific T cell response.

Conclusions: In sum, using multiple indices of T cell function, this study failed to demonstrate that anti-IgE therapy broadly or potently inhibits allergen specific T cell responses. As such, these data do not support a major role for IgE facilitated Ag presentation augmenting allergen specific T cell responses in vivo.

Trial registration: ClinicalTrials.gov identifier NCT00084097.

Figures

Figure 1
Figure 1
CFSE determination of allergen specific CD4 T cell responses. PBMC were activated and stained as per the Material and Methods and then gated on CD3+, violet LIVE/DEAD negative cells (A), and subsequently gated on CD4+, CD8- or CD4-, CD8+ cells (B). After culture with media (C), or peanut antigen extract (D, E), cells were gated on viable (C, D) CD4, or (E) CD8 T cells and CFSE vs. CD4 dotplots were generated.
Figure 2
Figure 2
Effect of anti-IgE therapy on allergen specific T cell proliferation. (A) Allergen specific CD4 T cell proliferation was measured by calculating the percentage of CFSElow cells and the results compared both at baseline and at completion of a 16-week trial of omalizumab. (B) Example of an allergen dose response to peanut Ag for subject 2, performed at baseline (circles) and at study completion (squares). (C) The EC50 for allergen proliferation was compared at baseline and at study completion. (D) The precursor frequency of allergen specific T cells was determined by CFSE dye dilution and compared at baseline and at study completion. Each color/symbol combination represents one subject and one allergen; results in A and D include two allergens examined for each subject.
Figure 3
Figure 3
Allergen specific cytokine staining. PBMC were activated with peanut Ag (A-D, G) or tetanus toxoid (E, F, H), stained for intracellular cytokines, and after gating on CD4+, CFSElow cells, cytokine dots plots of the noted cytokine pairs were generated.
Figure 4
Figure 4
Effect of anti-IgE therapy on allergen specific T cell cytokines. The ratio of IL-4:IFN-γ (A, C) and IL-5:IFN-γ (B, D) producing cells were measured in cultures stimulated with either allergen (A, B) or tetanus toxoid (C, D), and compared at baseline and at study completion. Each color/symbol combination represents one subject and one allergen; results in A and B include two allergens examined for each subject.

References

    1. Stone KD, Prussin C, Metcalfe DD. IgE, mast cells, basophils, and eosinophils. J Allergy Clin Immunol. 2010;125:S73–80. doi: 10.1016/j.jaci.2009.11.017.
    1. Maurer D, Ebner C, Reininger B, Fiebiger E, Kraft D, Kinet JP, Stingl G. The high affinity IgE receptor (Fc epsilon RI) mediates IgE-dependent allergen presentation. J Immunol. 1995;154:6285–6290.
    1. van der Heijden FL, Joost van Neerven RJ, van Katwijk M, Bos JD, Kapsenberg ML. Serum-IgE-facilitated allergen presentation in atopic disease. J Immunol. 1993;150:3643–3650.
    1. Schroeder JT, Bieneman AP, Xiao H, Chichester KL, Vasagar K, Saini S, Liu MC. TLR9- and FcepsilonRI-mediated responses oppose one another in plasmacytoid dendritic cells by down-regulating receptor expression. J Immunol. 2005;175:5724–5731.
    1. Gill MA, Bajwa G, George TA, Dong CC, Dougherty II, Jiang N, Gan VN, Gruchalla RS. Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells. J Immunol. 2010;184:5999–6006. doi: 10.4049/jimmunol.0901194.
    1. Grayson MH, Cheung D, Rohlfing MM, Kitchens R, Spiegel DE, Tucker J, Battaile JT, Alevy Y, Yan L, Agapov E, Kim EY, Holtzman MJ. Induction of high-affinity IgE receptor on lung dendritic cells during viral infection leads to mucous cell metaplasia. J Exp Med. 2007;204:2759–2769. doi: 10.1084/jem.20070360.
    1. Khan SH, Grayson MH. Cross-linking IgE augments human conventional dendritic cell production of CC chemokine ligand 28. J Allergy Clin Immunol. 2010;125:265–267. doi: 10.1016/j.jaci.2009.09.038.
    1. Holgate S, Casale T, Wenzel S, Bousquet J, Deniz Y, Reisner C. The anti-inflammatory effects of omalizumab confirm the central role of IgE in allergic inflammation. J Allergy Clin Immunol. 2005;115:459–465. doi: 10.1016/j.jaci.2004.11.053.
    1. Prussin C, Griffith DT, Boesel KM, Lin H, Foster B, Casale TB. Omalizumab treatment downregulates dendritic cell FcepsilonRI expression. J Allergy Clin Immunol. 2003;112:1147–1154. doi: 10.1016/j.jaci.2003.10.003.
    1. Schroeder JT, Bieneman AP, Chichester KL, Hamilton RG, Xiao H, Saini SS, Liu MC. Decreases in human dendritic cell-dependent T(H)2-like responses after acute in vivo IgE neutralization. J Allergy Clin Immunol. 2010;125:896–901. doi: 10.1016/j.jaci.2009.10.021. e896.
    1. Klunker S, Saggar LR, Seyfert-Margolis V, Asare AL, Casale TB, Durham SR, Francis JN. Combination treatment with omalizumab and rush immunotherapy for ragweed-induced allergic rhinitis: Inhibition of IgE-facilitated allergen binding. J Allergy Clin Immunol. 2007;120:688–695. doi: 10.1016/j.jaci.2007.05.034.
    1. Roederer M. Multiparameter FACS analysis. Curr Protoc Immunol 2002. Unit 5 8.
    1. Foroughi S, Foster B, Kim N, Bernardino LB, Scott LM, Hamilton RG, Metcalfe DD, Mannon PJ, Prussin C. Anti-IgE treatment of eosinophil-associated gastrointestinal disorders. J Allergy Clin Immunol. 2007;120:594–601. doi: 10.1016/j.jaci.2007.06.015.
    1. Prussin C, Lee J, Foster B. Eosinophilic gastrointestinal disease and peanut allergy are alternatively associated with IL-5+ and IL-5(-) T(H)2 responses. J Allergy Clin Immunol. 2009;124:1326–1332. doi: 10.1016/j.jaci.2009.09.048. e1326.
    1. Turcanu V, Maleki SJ, Lack G. Characterization of lymphocyte responses to peanuts in normal children, peanut-allergic children, and allergic children who acquired tolerance to peanuts. J Clin Invest. 2003;111:1065–1072.
    1. Foster B, Prussin C. In: Unit 6.24, Detection of Intracellular Cytokines by Flow Cytometry In Current Protocols in Immunology. Coligan JE, Kruisbeek AM, Margulies DH, Shevach EM, editor. Strober W: Wiley; 2003. pp. 6.24.21–26.24.16.
    1. Givan AL, Fisher JL, Waugh MG, Bercovici N, Wallace PK. Use of cell-tracking dyes to determine proliferation precursor frequencies of antigen-specific T cells. Methods Mol Biol. 2004;263:109–124.
    1. Noga O, Hanf G, Brachmann I, Klucken AC, Kleine-Tebbe J, Rosseau S, Kunkel G, Suttorp N, Seybold J. Effect of omalizumab treatment on peripheral eosinophil and T-lymphocyte function in patients with allergic asthma. J Allergy Clin Immunol. 2006;117:1493–1499. doi: 10.1016/j.jaci.2006.02.028.
    1. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005;23:307–336. doi: 10.1146/annurev.immunol.23.021704.115843.
    1. Sokol CL, Medzhitov R. Role of basophils in the initiation of Th2 responses. Curr Opin Immunol. 2010;22:73–77. doi: 10.1016/j.coi.2010.01.012.
    1. Spergel JM. Eosinophilic esophagitis in adults and children: evidence for a food allergy component in many patients. Curr Opin Allergy Clin Immunol. 2007;7:274–278. doi: 10.1097/ACI.0b013e32813aee4a.
    1. Chehade M, Aceves SS. Food allergy and eosinophilic esophagitis. Curr Opin Allergy Clin Immunol. 2010;10:231–237. doi: 10.1097/ACI.0b013e328338cbab.
    1. Liacouras CA, Spergel JM, Ruchelli E, Verma R, Mascarenhas M, Semeao E, Flick J, Kelly J, Brown-Whitehorn T, Mamula P, Markowitz JE. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol. 2005;3:1198–1206. doi: 10.1016/S1542-3565(05)00885-2.
    1. Roy-Ghanta S, Larosa DF, Katzka DA. Atopic characteristics of adult patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2008;6:531–535. doi: 10.1016/j.cgh.2007.12.045.
    1. Easterbrook PJ, Berlin JA, Gopalan R, Matthews DR. Publication bias in clinical research. Lancet. 1991;337:867–872. doi: 10.1016/0140-6736(91)90201-Y.
    1. Sridharan L, Greenland P. Editorial policies and publication bias: the importance of negative studies. Arch Intern Med. 2009;169:1022–1023. doi: 10.1001/archinternmed.2009.100.

Source: PubMed

3
구독하다