Skip to main content
Top
Published in: Clinical and Translational Allergy 1/2017

Open Access 01-12-2017 | Review

Application of the adverse outcome pathway (AOP) concept to structure the available in vivo and in vitro mechanistic data for allergic sensitization to food proteins

Authors: Jolanda H. M. van Bilsen, Edyta Sienkiewicz-Szłapka, Daniel Lozano-Ojalvo, Linette E. M. Willemsen, Celia M. Antunes, Elena Molina, Joost J. Smit, Barbara Wróblewska, Harry J. Wichers, Edward F. Knol, Gregory S. Ladics, Raymond H. H. Pieters, Sandra Denery-Papini, Yvonne M. Vissers, Simona L. Bavaro, Colette Larré, Kitty C. M. Verhoeckx, Erwin L. Roggen

Published in: Clinical and Translational Allergy | Issue 1/2017

Login to get access

Abstract

Background

The introduction of whole new foods in a population may lead to sensitization and food allergy. This constitutes a potential public health problem and a challenge to risk assessors and managers as the existing understanding of the pathophysiological processes and the currently available biological tools for prediction of the risk for food allergy development and the severity of the reaction are not sufficient. There is a substantial body of in vivo and in vitro data describing molecular and cellular events potentially involved in food sensitization. However, these events have not been organized in a sequence of related events that is plausible to result in sensitization, and useful to challenge current hypotheses. The aim of this manuscript was to collect and structure the current mechanistic understanding of sensitization induction to food proteins by applying the concept of adverse outcome pathway (AOP).

Main body

The proposed AOP for food sensitization is based on information on molecular and cellular mechanisms and pathways evidenced to be involved in sensitization by food and food proteins and uses the AOPs for chemical skin sensitization and respiratory sensitization induction as templates. Available mechanistic data on protein respiratory sensitization were included to fill out gaps in the understanding of how proteins may affect cells, cell–cell interactions and tissue homeostasis. Analysis revealed several key events (KE) and biomarkers that may have potential use in testing and assessment of proteins for their sensitizing potential.

Conclusion

The application of the AOP concept to structure mechanistic in vivo and in vitro knowledge has made it possible to identify a number of methods, each addressing a specific KE, that provide information about the food allergenic potential of new proteins. When applied in the context of an integrated strategy these methods may reduce, if not replace, current animal testing approaches. The proposed AOP will be shared at the www.​aopwiki.​org platform to expand the mechanistic data, improve the confidence in each of the proposed KE and key event relations (KERs), and allow for the identification of new, or refinement of established KE and KERs.
Literature
1.
go back to reference Nwaru BI, Hickstein L, Panesar SS, Muraro A, Werfel T, Cardona V, Dubois AE, Halken S, Hoffmann-Sommergruber K, Poulsen LK, Roberts G, Van Ree R, Vlieg-Boerstra BJ, Sheikh A. EAACI food allergy and anaphylaxis guidelines group: the epidemiology of food allergy in Europe: a systematic review and meta-analysis. Allergy. 2014;69(1):62–75.PubMedCrossRef Nwaru BI, Hickstein L, Panesar SS, Muraro A, Werfel T, Cardona V, Dubois AE, Halken S, Hoffmann-Sommergruber K, Poulsen LK, Roberts G, Van Ree R, Vlieg-Boerstra BJ, Sheikh A. EAACI food allergy and anaphylaxis guidelines group: the epidemiology of food allergy in Europe: a systematic review and meta-analysis. Allergy. 2014;69(1):62–75.PubMedCrossRef
2.
go back to reference Vetander M, Ly DH, Hakansson N, Lilja G, Nilsson C, Ostblom E, Wickman M, Bergstrom A. Recurrent reactions to food among children at paediatric emergency departments: epidemiology of allergic disease. Clin Exp Allergy. 2014;44(1):113–20.PubMedCrossRef Vetander M, Ly DH, Hakansson N, Lilja G, Nilsson C, Ostblom E, Wickman M, Bergstrom A. Recurrent reactions to food among children at paediatric emergency departments: epidemiology of allergic disease. Clin Exp Allergy. 2014;44(1):113–20.PubMedCrossRef
3.
go back to reference Jansson SA, Heibert-Arnlind M, Middelveld RJ, Bengtsson UJ, Sundqvist AC, Kallstrom-Bengtsson I, Marklund B, Rentzos G, Akerstrom J, Ostblom E, Dahlen SE, Ahlstedt S. Health-related quality of life, assessed with a disease-specific questionnaire, in Swedish adults suffering from well-diagnosed food allergy to staple foods. Clin Transl Allergy 2013;3:21-7022-3-21 (eCollection 2013). Jansson SA, Heibert-Arnlind M, Middelveld RJ, Bengtsson UJ, Sundqvist AC, Kallstrom-Bengtsson I, Marklund B, Rentzos G, Akerstrom J, Ostblom E, Dahlen SE, Ahlstedt S. Health-related quality of life, assessed with a disease-specific questionnaire, in Swedish adults suffering from well-diagnosed food allergy to staple foods. Clin Transl Allergy 2013;3:21-7022-3-21 (eCollection 2013).
4.
go back to reference Mills ENC, Mackie AR, Burney P, Beyer K, Frewer L, Madsen C, Botjes E, Crevel RWR, Van Ree R. The prevalence, cost and basis of food allergy across Europe. Allergy Eur J Allergy Clin Immunol. 2007;62(7):717–22.CrossRef Mills ENC, Mackie AR, Burney P, Beyer K, Frewer L, Madsen C, Botjes E, Crevel RWR, Van Ree R. The prevalence, cost and basis of food allergy across Europe. Allergy Eur J Allergy Clin Immunol. 2007;62(7):717–22.CrossRef
6.
go back to reference Untersmayr E, Jensen-Jarolim E. Mechanisms of type I food allergy. Pharmacol Ther. 2006;112(3):787–98.PubMedCrossRef Untersmayr E, Jensen-Jarolim E. Mechanisms of type I food allergy. Pharmacol Ther. 2006;112(3):787–98.PubMedCrossRef
8.
go back to reference Minekus M, Alminger M, Alvito P, Ballance S, Bohn T, Bourlieu C, Carriere F, Boutrou R, Corredig M, Dupont D, Dufour C, Egger L, Golding M, Karakaya S, Kirkhus B, Le Feunteun S, Lesmes U, Macierzanka A, Mackie A, Marze S, McClements DJ, Menard O, Recio I, Santos CN, Singh RP, Vegarud GE, Wickham MS, Weitschies W, Brodkorb A. A standardised static in vitro digestion method suitable for food—an international consensus. Food Funct. 2014;5(6):1113–24.PubMedCrossRef Minekus M, Alminger M, Alvito P, Ballance S, Bohn T, Bourlieu C, Carriere F, Boutrou R, Corredig M, Dupont D, Dufour C, Egger L, Golding M, Karakaya S, Kirkhus B, Le Feunteun S, Lesmes U, Macierzanka A, Mackie A, Marze S, McClements DJ, Menard O, Recio I, Santos CN, Singh RP, Vegarud GE, Wickham MS, Weitschies W, Brodkorb A. A standardised static in vitro digestion method suitable for food—an international consensus. Food Funct. 2014;5(6):1113–24.PubMedCrossRef
9.
go back to reference Bischoff S, Crowe SE. Gastrointestinal food allergy: new insights into pathophysiology and clinical perspectives. Gastroenterology. 2005;128(4):1089–113.PubMedCrossRef Bischoff S, Crowe SE. Gastrointestinal food allergy: new insights into pathophysiology and clinical perspectives. Gastroenterology. 2005;128(4):1089–113.PubMedCrossRef
10.
go back to reference Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ J 2015;8(1):17-015-0063-2 (eCollection 2015). Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ J 2015;8(1):17-015-0063-2 (eCollection 2015).
11.
go back to reference OECD: Users’ Handbook supplement to the Guidance Document for developing and assessing Adverse Outcome Pathways. OECD Series on Adverse Outcome Pathways 2016. OECD: Users’ Handbook supplement to the Guidance Document for developing and assessing Adverse Outcome Pathways. OECD Series on Adverse Outcome Pathways 2016.
12.
go back to reference OECD: The Adverse Outcome Pathway for Skin Sensitisation Initiated by Covalent Binding to Proteins: Organisation for Economic Co-operation and Development; 2014. OECD: The Adverse Outcome Pathway for Skin Sensitisation Initiated by Covalent Binding to Proteins: Organisation for Economic Co-operation and Development; 2014.
14.
go back to reference Wills-Karp M, Nathan A, Page K, Karp CL. New insights into innate immune mechanisms underlying allergenicity. Mucosal Immunol. 2010;3(2):104–10.PubMedCrossRef Wills-Karp M, Nathan A, Page K, Karp CL. New insights into innate immune mechanisms underlying allergenicity. Mucosal Immunol. 2010;3(2):104–10.PubMedCrossRef
15.
go back to reference Menard S, Cerf-Bensussan N, Heyman M. Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal Immunol. 2010;3(3):247–59.PubMedCrossRef Menard S, Cerf-Bensussan N, Heyman M. Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal Immunol. 2010;3(3):247–59.PubMedCrossRef
16.
go back to reference Heyman M. Gut barrier dysfunction in food allergy. Eur J Gastroenterol Hepatol. 2005;17(12):1279–85.PubMedCrossRef Heyman M. Gut barrier dysfunction in food allergy. Eur J Gastroenterol Hepatol. 2005;17(12):1279–85.PubMedCrossRef
17.
go back to reference Kondoh M, Takahashi A, Yagi K. Spiral progression in the development of absorption enhancers based on the biology of tight junctions. Adv Drug Deliv Rev. 2012;64(6):515–22.PubMedCrossRef Kondoh M, Takahashi A, Yagi K. Spiral progression in the development of absorption enhancers based on the biology of tight junctions. Adv Drug Deliv Rev. 2012;64(6):515–22.PubMedCrossRef
18.
go back to reference Bevilacqua C, Montagnac G, Benmerah A, Candalh C, Brousse N, Cerf-Bensussan N, Perdue MH, Heyman M. Food allergens are protected from degradation during CD23-mediated transepithelial transport. Int Arch Allergy Immunol. 2004;135(2):108–16.PubMedCrossRef Bevilacqua C, Montagnac G, Benmerah A, Candalh C, Brousse N, Cerf-Bensussan N, Perdue MH, Heyman M. Food allergens are protected from degradation during CD23-mediated transepithelial transport. Int Arch Allergy Immunol. 2004;135(2):108–16.PubMedCrossRef
19.
20.
go back to reference Sander GR, Cummins AG, Henshall T, Powell BC. Rapid disruption of intestinal barrier function by gliadin involves altered expression of apical junctional proteins. FEBS Lett. 2005;579(21):4851–5.PubMedCrossRef Sander GR, Cummins AG, Henshall T, Powell BC. Rapid disruption of intestinal barrier function by gliadin involves altered expression of apical junctional proteins. FEBS Lett. 2005;579(21):4851–5.PubMedCrossRef
21.
go back to reference Song CH, Liu ZQ, Huang S, Zheng PY, Yang PC. Probiotics promote endocytic allergen degradation in gut epithelial cells. Biochem Biophys Res Commun. 2012;426(1):135–40.PubMedCrossRef Song CH, Liu ZQ, Huang S, Zheng PY, Yang PC. Probiotics promote endocytic allergen degradation in gut epithelial cells. Biochem Biophys Res Commun. 2012;426(1):135–40.PubMedCrossRef
22.
go back to reference Grozdanovic MM, Cavic M, Nesic A, Andjelkovic U, Akbari P, Smit JJ, Gavrovic-Jankulovic M. Kiwifruit cysteine protease actinidin compromises the intestinal barrier by disrupting tight junctions. Biochim Biophys Acta. 2016;1860(3):516–26.PubMedCrossRef Grozdanovic MM, Cavic M, Nesic A, Andjelkovic U, Akbari P, Smit JJ, Gavrovic-Jankulovic M. Kiwifruit cysteine protease actinidin compromises the intestinal barrier by disrupting tight junctions. Biochim Biophys Acta. 2016;1860(3):516–26.PubMedCrossRef
23.
go back to reference Price DB, Ackland ML, Burks W, Knight MI, Suphioglu C. Peanut allergens alter intestinal barrier permeability and tight junction localisation in Caco-2 cell cultures. Cell Physiol Biochem. 2014;33(6):1758–77.PubMedCrossRef Price DB, Ackland ML, Burks W, Knight MI, Suphioglu C. Peanut allergens alter intestinal barrier permeability and tight junction localisation in Caco-2 cell cultures. Cell Physiol Biochem. 2014;33(6):1758–77.PubMedCrossRef
24.
go back to reference Hong JH, Lee SI, Kim KE, Yong TS, Seo JT, Sohn MH, Shin DM. German cockroach extract activates protease-activated receptor 2 in human airway epithelial cells. J Allergy Clin Immunol. 2004;113(2):315–9.PubMedCrossRef Hong JH, Lee SI, Kim KE, Yong TS, Seo JT, Sohn MH, Shin DM. German cockroach extract activates protease-activated receptor 2 in human airway epithelial cells. J Allergy Clin Immunol. 2004;113(2):315–9.PubMedCrossRef
25.
go back to reference Jacob C, Yang PC, Darmoul D, Amadesi S, Saito T, Cottrell GS, Coelho AM, Singh P, Grady EF, Perdue M, Bunnett NW. Mast cell tryptase controls paracellular permeability of the intestine. Role of protease-activated receptor 2 and beta-arrestins. J Biol Chem. 2005;280(36):31936–48.PubMedCrossRef Jacob C, Yang PC, Darmoul D, Amadesi S, Saito T, Cottrell GS, Coelho AM, Singh P, Grady EF, Perdue M, Bunnett NW. Mast cell tryptase controls paracellular permeability of the intestine. Role of protease-activated receptor 2 and beta-arrestins. J Biol Chem. 2005;280(36):31936–48.PubMedCrossRef
26.
go back to reference Berin MC, Yang PC, Ciok L, Waserman S, Perdue MH. Role for IL-4 in macromolecular transport across human intestinal epithelium. Am J Physiol Cell Physiol. 1999;276(5):C1046–52. Berin MC, Yang PC, Ciok L, Waserman S, Perdue MH. Role for IL-4 in macromolecular transport across human intestinal epithelium. Am J Physiol Cell Physiol. 1999;276(5):C1046–52.
27.
go back to reference Heller F, Florian P, Bojarski C, Richter J, Christ M, Hillenbrand B, Mankertz J, Gitter AH, Burgel N, Fromm M, Zeitz M, Fuss I, Strober W, Schulzke JD. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology. 2005;129(2):550–64.PubMedCrossRef Heller F, Florian P, Bojarski C, Richter J, Christ M, Hillenbrand B, Mankertz J, Gitter AH, Burgel N, Fromm M, Zeitz M, Fuss I, Strober W, Schulzke JD. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology. 2005;129(2):550–64.PubMedCrossRef
28.
go back to reference Ferrier L, Mazelin L, Cenac N, Desreumaux P, Janin A, Emilie D, Colombel JF, Garcia-Villar R, Fioramonti J, Bueno L. Stress-induced disruption of colonic epithelial barrier: role of interferon-gamma and myosin light chain kinase in mice. Gastroenterology. 2003;125(3):795–804.PubMedCrossRef Ferrier L, Mazelin L, Cenac N, Desreumaux P, Janin A, Emilie D, Colombel JF, Garcia-Villar R, Fioramonti J, Bueno L. Stress-induced disruption of colonic epithelial barrier: role of interferon-gamma and myosin light chain kinase in mice. Gastroenterology. 2003;125(3):795–804.PubMedCrossRef
29.
go back to reference Willemsen LE, Hoetjes JP, van Deventer SJ, van Tol EA. Abrogation of IFN-gamma mediated epithelial barrier disruption by serine protease inhibition. Clin Exp Immunol. 2005;142(2):275–84.PubMedPubMedCentralCrossRef Willemsen LE, Hoetjes JP, van Deventer SJ, van Tol EA. Abrogation of IFN-gamma mediated epithelial barrier disruption by serine protease inhibition. Clin Exp Immunol. 2005;142(2):275–84.PubMedPubMedCentralCrossRef
30.
go back to reference Dhawan S, Hiemstra IH, Verseijden C, Hilbers FW, Te Velde AA, Willemsen LE, Stap J, den Haan JM, de Jonge WJ. Cholinergic receptor activation on epithelia protects against cytokine-induced barrier dysfunction. Acta Physiol (Oxf). 2015;213(4):846–59.CrossRef Dhawan S, Hiemstra IH, Verseijden C, Hilbers FW, Te Velde AA, Willemsen LE, Stap J, den Haan JM, de Jonge WJ. Cholinergic receptor activation on epithelia protects against cytokine-induced barrier dysfunction. Acta Physiol (Oxf). 2015;213(4):846–59.CrossRef
31.
go back to reference Brandt EB, Strait RT, Hershko D, Wang Q, Muntel EE, Scribner TA, Zimmermann N, Finkelman FD, Rothenberg ME. Mast cells are required for experimental oral allergen-induced diarrhea. J Clin Invest. 2003;112(11):1666–77.PubMedPubMedCentralCrossRef Brandt EB, Strait RT, Hershko D, Wang Q, Muntel EE, Scribner TA, Zimmermann N, Finkelman FD, Rothenberg ME. Mast cells are required for experimental oral allergen-induced diarrhea. J Clin Invest. 2003;112(11):1666–77.PubMedPubMedCentralCrossRef
32.
go back to reference Kurashima Y, Kiyono H. New era for mucosal mast cells: their roles in inflammation, allergic immune responses and adjuvant development. Exp Mol Med. 2014;46:e83.PubMedPubMedCentralCrossRef Kurashima Y, Kiyono H. New era for mucosal mast cells: their roles in inflammation, allergic immune responses and adjuvant development. Exp Mol Med. 2014;46:e83.PubMedPubMedCentralCrossRef
33.
go back to reference Heyman M, Desjeux JF. Significance of intestinal food protein transport. J Pediatr Gastroenterol Nutr. 1992;15(1):48–57.PubMedCrossRef Heyman M, Desjeux JF. Significance of intestinal food protein transport. J Pediatr Gastroenterol Nutr. 1992;15(1):48–57.PubMedCrossRef
34.
go back to reference Caillard I, Tome D. Transport of beta-lactoglobulin and alpha-lactalbumin in enterocyte-like Caco-2 cells. Reprod Nutr Dev. 1995;35(2):179–88.PubMedCrossRef Caillard I, Tome D. Transport of beta-lactoglobulin and alpha-lactalbumin in enterocyte-like Caco-2 cells. Reprod Nutr Dev. 1995;35(2):179–88.PubMedCrossRef
35.
go back to reference Bernasconi E, Fritsche R, Corthesy B. Specific effects of denaturation, hydrolysis and exposure to Lactococcus lactis on bovine beta-lactoglobulin transepithelial transport, antigenicity and allergenicity. Clin Exp Allergy. 2006;36(6):803–14.PubMedCrossRef Bernasconi E, Fritsche R, Corthesy B. Specific effects of denaturation, hydrolysis and exposure to Lactococcus lactis on bovine beta-lactoglobulin transepithelial transport, antigenicity and allergenicity. Clin Exp Allergy. 2006;36(6):803–14.PubMedCrossRef
36.
go back to reference Sewekow E, Bimczok D, Kahne T, Faber-Zuschratter H, Kessler LC, Seidel-Morgenstern A, Rothkotter HJ. The major soyabean allergen P34 resists proteolysis in vitro and is transported through intestinal epithelial cells by a caveolae-mediated mechanism. Br J Nutr. 2012;108(9):1603–11.PubMedCrossRef Sewekow E, Bimczok D, Kahne T, Faber-Zuschratter H, Kessler LC, Seidel-Morgenstern A, Rothkotter HJ. The major soyabean allergen P34 resists proteolysis in vitro and is transported through intestinal epithelial cells by a caveolae-mediated mechanism. Br J Nutr. 2012;108(9):1603–11.PubMedCrossRef
37.
go back to reference Rytkonen J, Valkonen KH, Virtanen V, Foxwell RA, Kyd JM, Cripps AW, Karttunen TJ. Enterocyte and M-cell transport of native and heat-denatured bovine beta-lactoglobulin: significance of heat denaturation. J Agric Food Chem. 2006;54(4):1500–7.PubMedCrossRef Rytkonen J, Valkonen KH, Virtanen V, Foxwell RA, Kyd JM, Cripps AW, Karttunen TJ. Enterocyte and M-cell transport of native and heat-denatured bovine beta-lactoglobulin: significance of heat denaturation. J Agric Food Chem. 2006;54(4):1500–7.PubMedCrossRef
38.
go back to reference Zlotkowska D, Maddaloni M, Riccardi C, Walters N, Holderness K, Callis G, Rynda-Apple A, Pascual DW. Loss of sialic acid binding domain redirects protein sigma1 to enhance M cell-directed vaccination. PLoS ONE. 2012;7(4):e36182.PubMedPubMedCentralCrossRef Zlotkowska D, Maddaloni M, Riccardi C, Walters N, Holderness K, Callis G, Rynda-Apple A, Pascual DW. Loss of sialic acid binding domain redirects protein sigma1 to enhance M cell-directed vaccination. PLoS ONE. 2012;7(4):e36182.PubMedPubMedCentralCrossRef
39.
go back to reference Brandtzaeg PE. Current understanding of gastrointestinal immunoregulation and its relation to food allergy. Ann N Y Acad Sci. 2002;964:13–45.PubMedCrossRef Brandtzaeg PE. Current understanding of gastrointestinal immunoregulation and its relation to food allergy. Ann N Y Acad Sci. 2002;964:13–45.PubMedCrossRef
40.
go back to reference Sampson HA. Food allergy. Part 1: immunopathogenesis and clinical disorders. J Allergy Clin Immunol. 1999;103(5 Pt 1):717–28.PubMedCrossRef Sampson HA. Food allergy. Part 1: immunopathogenesis and clinical disorders. J Allergy Clin Immunol. 1999;103(5 Pt 1):717–28.PubMedCrossRef
41.
go back to reference Roth-Walter F, Berin MC, Arnaboldi P, Escalante CR, Dahan S, Rauch J, Jensen-Jarolim E, Mayer L. Pasteurization of milk proteins promotes allergic sensitization by enhancing uptake through Peyer’s patches. Allergy. 2008;63(7):882–90.PubMedCrossRef Roth-Walter F, Berin MC, Arnaboldi P, Escalante CR, Dahan S, Rauch J, Jensen-Jarolim E, Mayer L. Pasteurization of milk proteins promotes allergic sensitization by enhancing uptake through Peyer’s patches. Allergy. 2008;63(7):882–90.PubMedCrossRef
42.
go back to reference Chambers SJ, Wickham MS, Regoli M, Bertelli E, Gunning PA, Nicoletti C. Rapid in vivo transport of proteins from digested allergen across pre-sensitized gut. Biochem Biophys Res Commun. 2004;325(4):1258–63.PubMedCrossRef Chambers SJ, Wickham MS, Regoli M, Bertelli E, Gunning PA, Nicoletti C. Rapid in vivo transport of proteins from digested allergen across pre-sensitized gut. Biochem Biophys Res Commun. 2004;325(4):1258–63.PubMedCrossRef
43.
go back to reference Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2(4):361–7.PubMedCrossRef Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2(4):361–7.PubMedCrossRef
44.
go back to reference Rubas W, Grass GM. Gastrointestinal lymphatic absorption of peptides and proteins. Adv Drug Deliv Rev. 1991;7(1):15–69.CrossRef Rubas W, Grass GM. Gastrointestinal lymphatic absorption of peptides and proteins. Adv Drug Deliv Rev. 1991;7(1):15–69.CrossRef
45.
go back to reference McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012;483(7389):345–9.PubMedPubMedCentralCrossRef McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012;483(7389):345–9.PubMedPubMedCentralCrossRef
46.
go back to reference Dunkin D, Berin MC, Mayer L. Allergic sensitization can be induced via multiple physiologic routes in an adjuvant-dependent manner. J Allergy Clin Immunol 2011;128(6):1251–58.e2. Dunkin D, Berin MC, Mayer L. Allergic sensitization can be induced via multiple physiologic routes in an adjuvant-dependent manner. J Allergy Clin Immunol 2011;128(6):1251–58.e2.
47.
go back to reference Birmingham NP, Parvataneni S, Hassan HM, Harkema J, Samineni S, Navuluri L, Kelly CJ, Gangur V. An adjuvant-free mouse model of tree nut allergy using hazelnut as a model tree nut. Int Arch Allergy Immunol. 2007;144(3):203–10.PubMedCrossRef Birmingham NP, Parvataneni S, Hassan HM, Harkema J, Samineni S, Navuluri L, Kelly CJ, Gangur V. An adjuvant-free mouse model of tree nut allergy using hazelnut as a model tree nut. Int Arch Allergy Immunol. 2007;144(3):203–10.PubMedCrossRef
48.
go back to reference Hsieh KY, Tsai CC, Wu CH, Lin RH. Epicutaneous exposure to protein antigen and food allergy. Clin Exp Allergy. 2003;33(8):1067–75.PubMedCrossRef Hsieh KY, Tsai CC, Wu CH, Lin RH. Epicutaneous exposure to protein antigen and food allergy. Clin Exp Allergy. 2003;33(8):1067–75.PubMedCrossRef
49.
go back to reference Fox AT, Sasieni P, du Toit G, Syed H, Lack G. Household peanut consumption as a risk factor for the development of peanut allergy. J Allergy Clin Immunol. 2009;123(2):417–23.PubMedCrossRef Fox AT, Sasieni P, du Toit G, Syed H, Lack G. Household peanut consumption as a risk factor for the development of peanut allergy. J Allergy Clin Immunol. 2009;123(2):417–23.PubMedCrossRef
50.
go back to reference Lack G, Fox D, Northstone K, Golding J. Avon longitudinal study of parents and children study team: factors associated with the development of peanut allergy in childhood. N Engl J Med. 2003;348(11):977–85.PubMedCrossRef Lack G, Fox D, Northstone K, Golding J. Avon longitudinal study of parents and children study team: factors associated with the development of peanut allergy in childhood. N Engl J Med. 2003;348(11):977–85.PubMedCrossRef
51.
go back to reference Agrawal R, Woodfolk JA. Skin barrier defects in atopic dermatitis. Curr Allergy Asthma Rep 2014;14(5):433-014-0433-9. Agrawal R, Woodfolk JA. Skin barrier defects in atopic dermatitis. Curr Allergy Asthma Rep 2014;14(5):433-014-0433-9.
52.
go back to reference Fukutomi Y, Taniguchi M, Nakamura H, Akiyama K. Epidemiological link between wheat allergy and exposure to hydrolyzed wheat protein in facial soap. Allergy. 2014;69(10):1405–11.PubMedCrossRef Fukutomi Y, Taniguchi M, Nakamura H, Akiyama K. Epidemiological link between wheat allergy and exposure to hydrolyzed wheat protein in facial soap. Allergy. 2014;69(10):1405–11.PubMedCrossRef
53.
go back to reference Beezhold DH, Sussman GL, Liss GM, Chang NS. Latex allergy can induce clinical reactions to specific foods. Clin Exp Allergy. 1996;26(4):416–22.PubMedCrossRef Beezhold DH, Sussman GL, Liss GM, Chang NS. Latex allergy can induce clinical reactions to specific foods. Clin Exp Allergy. 1996;26(4):416–22.PubMedCrossRef
55.
go back to reference Bischoff SC, Mayer JH, Manns MP. Allergy and the gut. Int Arch Allergy Immunol. 2000;121(4):270–83.PubMedCrossRef Bischoff SC, Mayer JH, Manns MP. Allergy and the gut. Int Arch Allergy Immunol. 2000;121(4):270–83.PubMedCrossRef
56.
go back to reference Armentia A, Diaz-Perales A, Castrodeza J, Duenas-Laita A, Palacin A, Fernandez S. Why can patients with baker’s asthma tolerate wheat flour ingestion? Is wheat pollen allergy relevant? Allergol Immunopathol (Madr). 2009;37(4):203–4.CrossRef Armentia A, Diaz-Perales A, Castrodeza J, Duenas-Laita A, Palacin A, Fernandez S. Why can patients with baker’s asthma tolerate wheat flour ingestion? Is wheat pollen allergy relevant? Allergol Immunopathol (Madr). 2009;37(4):203–4.CrossRef
57.
58.
go back to reference van Wijk F, Nierkens S, Hassing I, Feijen M, Koppelman SJ, de Jong GA, Pieters R, Knippels LM. The effect of the food matrix on in vivo immune responses to purified peanut allergens. Toxicol Sci. 2005;86(2):333–41.PubMedCrossRef van Wijk F, Nierkens S, Hassing I, Feijen M, Koppelman SJ, de Jong GA, Pieters R, Knippels LM. The effect of the food matrix on in vivo immune responses to purified peanut allergens. Toxicol Sci. 2005;86(2):333–41.PubMedCrossRef
59.
go back to reference Wavrin S, Bernard H, Wal JM, Adel-Patient K. Influence of the route of exposure and the matrix on the sensitisation potency of a major cows’ milk allergen. Clin Transl Allergy 2015;5(1):3-015-0047-x (eCollection 2015). Wavrin S, Bernard H, Wal JM, Adel-Patient K. Influence of the route of exposure and the matrix on the sensitisation potency of a major cows’ milk allergen. Clin Transl Allergy 2015;5(1):3-015-0047-x (eCollection 2015).
60.
go back to reference Moghaddam AE, Hillson WR, Noti M, Gartlan KH, Johnson S, Thomas B, Artis D, Sattentau QJ. Dry roasting enhances peanut-induced allergic sensitization across mucosal and cutaneous routes in mice. J Allergy Clin Immunol. 2014;134(6):1453–6.PubMedPubMedCentralCrossRef Moghaddam AE, Hillson WR, Noti M, Gartlan KH, Johnson S, Thomas B, Artis D, Sattentau QJ. Dry roasting enhances peanut-induced allergic sensitization across mucosal and cutaneous routes in mice. J Allergy Clin Immunol. 2014;134(6):1453–6.PubMedPubMedCentralCrossRef
61.
go back to reference Hilmenyuk T, Bellinghausen I, Heydenreich B, Ilchmann A, Toda M, Grabbe S, Saloga J. Effects of glycation of the model food allergen ovalbumin on antigen uptake and presentation by human dendritic cells. Immunology. 2010;129(3):437–45.PubMedPubMedCentralCrossRef Hilmenyuk T, Bellinghausen I, Heydenreich B, Ilchmann A, Toda M, Grabbe S, Saloga J. Effects of glycation of the model food allergen ovalbumin on antigen uptake and presentation by human dendritic cells. Immunology. 2010;129(3):437–45.PubMedPubMedCentralCrossRef
62.
go back to reference Salazar F, Sewell HF, Shakib F, Ghaemmaghami AM. The role of lectins in allergic sensitization and allergic disease. J Allergy Clin Immunol. 2013;132(1):27–36.PubMedCrossRef Salazar F, Sewell HF, Shakib F, Ghaemmaghami AM. The role of lectins in allergic sensitization and allergic disease. J Allergy Clin Immunol. 2013;132(1):27–36.PubMedCrossRef
63.
go back to reference Lilly LM, Gessner MA, Dunaway CW, Metz AE, Schwiebert L, Weaver CT, Brown GD, Steele C. The beta-glucan receptor dectin-1 promotes lung immunopathology during fungal allergy via IL-22. J Immunol. 2012;189(7):3653–60.PubMedPubMedCentralCrossRef Lilly LM, Gessner MA, Dunaway CW, Metz AE, Schwiebert L, Weaver CT, Brown GD, Steele C. The beta-glucan receptor dectin-1 promotes lung immunopathology during fungal allergy via IL-22. J Immunol. 2012;189(7):3653–60.PubMedPubMedCentralCrossRef
64.
go back to reference Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, Brown J, Becker CA, Fleshner PR, Dubinsky M, Rotter JI, Wang HL, McGovern DP, Brown GD, Underhill DM. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science. 2012;336(6086):1314–7.PubMedPubMedCentralCrossRef Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, Brown J, Becker CA, Fleshner PR, Dubinsky M, Rotter JI, Wang HL, McGovern DP, Brown GD, Underhill DM. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science. 2012;336(6086):1314–7.PubMedPubMedCentralCrossRef
65.
go back to reference Barrett NA, Rahman OM, Fernandez JM, Parsons MW, Xing W, Austen KF, Kanaoka Y. Dectin-2 mediates Th2 immunity through the generation of cysteinyl leukotrienes. J Exp Med. 2011;208(3):593–604.PubMedPubMedCentralCrossRef Barrett NA, Rahman OM, Fernandez JM, Parsons MW, Xing W, Austen KF, Kanaoka Y. Dectin-2 mediates Th2 immunity through the generation of cysteinyl leukotrienes. J Exp Med. 2011;208(3):593–604.PubMedPubMedCentralCrossRef
66.
go back to reference Clarke DL, Davis NH, Campion CL, Foster ML, Heasman SC, Lewis AR, Anderson IK, Corkill DJ, Sleeman MA, May RD, Robinson MJ. Dectin-2 sensing of house dust mite is critical for the initiation of airway inflammation. Mucosal Immunol. 2014;7(3):558–67.PubMedCrossRef Clarke DL, Davis NH, Campion CL, Foster ML, Heasman SC, Lewis AR, Anderson IK, Corkill DJ, Sleeman MA, May RD, Robinson MJ. Dectin-2 sensing of house dust mite is critical for the initiation of airway inflammation. Mucosal Immunol. 2014;7(3):558–67.PubMedCrossRef
67.
go back to reference Parsons MW, Li L, Wallace AM, Lee MJ, Katz HR, Fernandez JM, Saijo S, Iwakura Y, Austen KF, Kanaoka Y, Barrett NA. Dectin-2 regulates the effector phase of house dust mite-elicited pulmonary inflammation independently from its role in sensitization. J Immunol. 2014;192(4):1361–71.PubMedPubMedCentralCrossRef Parsons MW, Li L, Wallace AM, Lee MJ, Katz HR, Fernandez JM, Saijo S, Iwakura Y, Austen KF, Kanaoka Y, Barrett NA. Dectin-2 regulates the effector phase of house dust mite-elicited pulmonary inflammation independently from its role in sensitization. J Immunol. 2014;192(4):1361–71.PubMedPubMedCentralCrossRef
68.
go back to reference Kamalakannan M, Chang LM, Grishina G, Sampson HA, Masilamani M. Identification and characterization of DC-SIGN-binding glycoproteins in allergenic foods. Allergy. 2016;71(8):1145–55.PubMedCrossRef Kamalakannan M, Chang LM, Grishina G, Sampson HA, Masilamani M. Identification and characterization of DC-SIGN-binding glycoproteins in allergenic foods. Allergy. 2016;71(8):1145–55.PubMedCrossRef
70.
go back to reference Guillon B, Bernard H, Drumare MF, Hazebrouck S, Adel-Patient K. Heat processing of peanut seed enhances the sensitization potential of the major peanut allergen Ara h 6. Mol Nutr Food Res. 2016;60(12):2722–35.PubMedPubMedCentralCrossRef Guillon B, Bernard H, Drumare MF, Hazebrouck S, Adel-Patient K. Heat processing of peanut seed enhances the sensitization potential of the major peanut allergen Ara h 6. Mol Nutr Food Res. 2016;60(12):2722–35.PubMedPubMedCentralCrossRef
71.
go back to reference Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–85.PubMedCrossRef Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–85.PubMedCrossRef
72.
go back to reference Corsini E, Galbiati V, Nikitovic D, Tsatsakis AM. Role of oxidative stress in chemical allergens induced skin cells activation. Food Chem Toxicol. 2013;61:74–81.PubMedCrossRef Corsini E, Galbiati V, Nikitovic D, Tsatsakis AM. Role of oxidative stress in chemical allergens induced skin cells activation. Food Chem Toxicol. 2013;61:74–81.PubMedCrossRef
73.
go back to reference Runswick S, Mitchell T, Davies P, Robinson C, Garrod DR. Pollen proteolytic enzymes degrade tight junctions. Respirology. 2007;12(6):834–42.PubMedCrossRef Runswick S, Mitchell T, Davies P, Robinson C, Garrod DR. Pollen proteolytic enzymes degrade tight junctions. Respirology. 2007;12(6):834–42.PubMedCrossRef
74.
go back to reference Brandt EB, Mingler MK, Stevenson MD, Wang N, Khurana Hershey GK, Whitsett JA, Rothenberg ME. Surfactant protein D alters allergic lung responses in mice and human subjects. J Allergy Clin Immunol 2008;121(5):1140–7.e2. Brandt EB, Mingler MK, Stevenson MD, Wang N, Khurana Hershey GK, Whitsett JA, Rothenberg ME. Surfactant protein D alters allergic lung responses in mice and human subjects. J Allergy Clin Immunol 2008;121(5):1140–7.e2.
75.
go back to reference Boldogh I, Bacsi A, Choudhury BK, Dharajiya N, Alam R, Hazra TK, Mitra S, Goldblum RM, Sur S. ROS generated by pollen NADPH oxidase provide a signal that augments antigen-induced allergic airway inflammation. J Clin Invest. 2005;115(8):2169–79.PubMedPubMedCentralCrossRef Boldogh I, Bacsi A, Choudhury BK, Dharajiya N, Alam R, Hazra TK, Mitra S, Goldblum RM, Sur S. ROS generated by pollen NADPH oxidase provide a signal that augments antigen-induced allergic airway inflammation. J Clin Invest. 2005;115(8):2169–79.PubMedPubMedCentralCrossRef
76.
go back to reference Janssens S, Beyaert R. A universal role for MyD88 in TLR/IL-1R-mediated signaling. Trends Biochem Sci. 2002;27(9):474–82.PubMedCrossRef Janssens S, Beyaert R. A universal role for MyD88 in TLR/IL-1R-mediated signaling. Trends Biochem Sci. 2002;27(9):474–82.PubMedCrossRef
77.
go back to reference Roggen EL. In vitro approaches for detection of chemical sensitization. Basic Clin Pharmacol Toxicol. 2014;115(1):32–40.PubMedCrossRef Roggen EL. In vitro approaches for detection of chemical sensitization. Basic Clin Pharmacol Toxicol. 2014;115(1):32–40.PubMedCrossRef
79.
go back to reference Ather JL, Hodgkins SR, Janssen-Heininger YM, Poynter ME. Airway epithelial NF-kappaB activation promotes allergic sensitization to an innocuous inhaled antigen. Am J Respir Cell Mol Biol. 2011;44(5):631–8.PubMedCrossRef Ather JL, Hodgkins SR, Janssen-Heininger YM, Poynter ME. Airway epithelial NF-kappaB activation promotes allergic sensitization to an innocuous inhaled antigen. Am J Respir Cell Mol Biol. 2011;44(5):631–8.PubMedCrossRef
80.
go back to reference Hartwig C, Tschernig T, Mazzega M, Braun A, Neumann D. Endogenous IL-18 in experimentally induced asthma affects cytokine serum levels but is irrelevant for clinical symptoms. Cytokine. 2008;42(3):298–305.PubMedCrossRef Hartwig C, Tschernig T, Mazzega M, Braun A, Neumann D. Endogenous IL-18 in experimentally induced asthma affects cytokine serum levels but is irrelevant for clinical symptoms. Cytokine. 2008;42(3):298–305.PubMedCrossRef
81.
go back to reference Kool M, Willart MA, van Nimwegen M, Bergen I, Pouliot P, Virchow JC, Rogers N, Osorio F, Reis e Sousa C, Hammad H, Lambrecht BN. An unexpected role for uric acid as an inducer of T helper 2 cell immunity to inhaled antigens and inflammatory mediator of allergic asthma. Immunity 2011;34(4):527–40. Kool M, Willart MA, van Nimwegen M, Bergen I, Pouliot P, Virchow JC, Rogers N, Osorio F, Reis e Sousa C, Hammad H, Lambrecht BN. An unexpected role for uric acid as an inducer of T helper 2 cell immunity to inhaled antigens and inflammatory mediator of allergic asthma. Immunity 2011;34(4):527–40.
82.
go back to reference Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, Moore CL, Seunghyun In T, Waserman S, Coyle AJ, Kolbeck R, Humbles AA, Jordana M. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol 2013;131(1):187-200.e1-8. Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, Moore CL, Seunghyun In T, Waserman S, Coyle AJ, Kolbeck R, Humbles AA, Jordana M. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol 2013;131(1):187-200.e1-8.
83.
go back to reference Junker Y, Zeissig S, Kim SJ, Barisani D, Wieser H, Leffler DA, Zevallos V, Libermann TA, Dillon S, Freitag TL, Kelly CP, Schuppan D. Wheat amylase trypsin inhibitors drive intestinal inflammation via activation of toll-like receptor 4. J Exp Med. 2012;209(13):2395–408.PubMedPubMedCentralCrossRef Junker Y, Zeissig S, Kim SJ, Barisani D, Wieser H, Leffler DA, Zevallos V, Libermann TA, Dillon S, Freitag TL, Kelly CP, Schuppan D. Wheat amylase trypsin inhibitors drive intestinal inflammation via activation of toll-like receptor 4. J Exp Med. 2012;209(13):2395–408.PubMedPubMedCentralCrossRef
84.
go back to reference Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, Gordon ME, Naji L, Flader K, Larche M, Chu DK, Waserman S, McCarry B, Jordana M. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70(5):495–505.PubMedCrossRef Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, Gordon ME, Naji L, Flader K, Larche M, Chu DK, Waserman S, McCarry B, Jordana M. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70(5):495–505.PubMedCrossRef
85.
go back to reference Tordesillas L, Cuesta-Herranz J, Gonzalez-Munoz M, Pacios LF, Compes E, Garcia-Carrasco B, Sanchez-Monge R, Salcedo G, Diaz-Perales A. T-cell epitopes of the major peach allergen, Pru p 3: identification and differential T-cell response of peach-allergic and non-allergic subjects. Mol Immunol. 2009;46(4):722–8.PubMedCrossRef Tordesillas L, Cuesta-Herranz J, Gonzalez-Munoz M, Pacios LF, Compes E, Garcia-Carrasco B, Sanchez-Monge R, Salcedo G, Diaz-Perales A. T-cell epitopes of the major peach allergen, Pru p 3: identification and differential T-cell response of peach-allergic and non-allergic subjects. Mol Immunol. 2009;46(4):722–8.PubMedCrossRef
86.
go back to reference Tordesillas L, Gomez-Casado C, Garrido-Arandia M, Murua-Garcia A, Palacin A, Varela J, Konieczna P, Cuesta-Herranz J, Akdis CA, O’Mahony L, Diaz-Perales A. Transport of Pru p 3 across gastrointestinal epithelium—an essential step towards the induction of food allergy? Clin Exp Allergy. 2013;43(12):1374–83.PubMedCrossRef Tordesillas L, Gomez-Casado C, Garrido-Arandia M, Murua-Garcia A, Palacin A, Varela J, Konieczna P, Cuesta-Herranz J, Akdis CA, O’Mahony L, Diaz-Perales A. Transport of Pru p 3 across gastrointestinal epithelium—an essential step towards the induction of food allergy? Clin Exp Allergy. 2013;43(12):1374–83.PubMedCrossRef
87.
go back to reference Tan JK, O’Neill HC. Maturation requirements for dendritic cells in T cell stimulation leading to tolerance versus immunity. J Leukoc Biol. 2005;78(2):319–24.PubMedCrossRef Tan JK, O’Neill HC. Maturation requirements for dendritic cells in T cell stimulation leading to tolerance versus immunity. J Leukoc Biol. 2005;78(2):319–24.PubMedCrossRef
88.
go back to reference Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. J Allergy Clin Immunol. 2008;121(6):1484–90.PubMedPubMedCentralCrossRef Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. J Allergy Clin Immunol. 2008;121(6):1484–90.PubMedPubMedCentralCrossRef
89.
go back to reference Chu DK, Jimenez-Saiz R, Verschoor CP, Walker TD, Goncharova S, Llop-Guevara A, Shen P, Gordon ME, Barra NG, Bassett JD, Kong J, Fattouh R, McCoy KD, Bowdish DM, Erjefalt JS, Pabst O, Humbles AA, Kolbeck R, Waserman S, Jordana M. Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo. J Exp Med. 2014;211(8):1657–72.PubMedPubMedCentralCrossRef Chu DK, Jimenez-Saiz R, Verschoor CP, Walker TD, Goncharova S, Llop-Guevara A, Shen P, Gordon ME, Barra NG, Bassett JD, Kong J, Fattouh R, McCoy KD, Bowdish DM, Erjefalt JS, Pabst O, Humbles AA, Kolbeck R, Waserman S, Jordana M. Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo. J Exp Med. 2014;211(8):1657–72.PubMedPubMedCentralCrossRef
90.
go back to reference Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009;31(3):502–12.PubMedCrossRef Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009;31(3):502–12.PubMedCrossRef
91.
go back to reference Scott CL, Bain CC, Wright PB, Sichien D, Kotarsky K, Persson EK, Luda K, Guilliams M, Lambrecht BN, Agace WW, Milling SW, Mowat AM. CCR2(+)CD103(−) intestinal dendritic cells develop from DC-committed precursors and induce interleukin-17 production by T cells. Mucosal Immunol. 2015;8(2):327–39.PubMedCrossRef Scott CL, Bain CC, Wright PB, Sichien D, Kotarsky K, Persson EK, Luda K, Guilliams M, Lambrecht BN, Agace WW, Milling SW, Mowat AM. CCR2(+)CD103(−) intestinal dendritic cells develop from DC-committed precursors and induce interleukin-17 production by T cells. Mucosal Immunol. 2015;8(2):327–39.PubMedCrossRef
92.
go back to reference Smit JJ, Bol-Schoenmakers M, Hassing I, Fiechter D, Boon L, Bleumink R, Pieters RH. The role of intestinal dendritic cells subsets in the establishment of food allergy. Clin Exp Allergy. 2011;41(6):890–8.PubMedCrossRef Smit JJ, Bol-Schoenmakers M, Hassing I, Fiechter D, Boon L, Bleumink R, Pieters RH. The role of intestinal dendritic cells subsets in the establishment of food allergy. Clin Exp Allergy. 2011;41(6):890–8.PubMedCrossRef
93.
go back to reference de Kivit S, van Hoffen E, Korthagen N, Garssen J, Willemsen LE. Apical TLR ligation of intestinal epithelial cells drives a Th1-polarized regulatory or inflammatory type effector response in vitro. Immunobiology. 2011;216(4):518–27.PubMedCrossRef de Kivit S, van Hoffen E, Korthagen N, Garssen J, Willemsen LE. Apical TLR ligation of intestinal epithelial cells drives a Th1-polarized regulatory or inflammatory type effector response in vitro. Immunobiology. 2011;216(4):518–27.PubMedCrossRef
94.
go back to reference Bashir ME, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004;172(11):6978–87.PubMedCrossRef Bashir ME, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004;172(11):6978–87.PubMedCrossRef
95.
go back to reference Bol-Schoenmakers M, Braber S, Akbari P, de Graaff P, van Roest M, Kruijssen L, Smit JJ, van Esch BC, Jeurink PV, Garssen J, Fink-Gremmels J, Pieters RH. The mycotoxin deoxynivalenol facilitates allergic sensitization to whey in mice. Mucosal Immunol. 2016;9(6):1477–86.PubMedCrossRef Bol-Schoenmakers M, Braber S, Akbari P, de Graaff P, van Roest M, Kruijssen L, Smit JJ, van Esch BC, Jeurink PV, Garssen J, Fink-Gremmels J, Pieters RH. The mycotoxin deoxynivalenol facilitates allergic sensitization to whey in mice. Mucosal Immunol. 2016;9(6):1477–86.PubMedCrossRef
97.
go back to reference Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, Bernhardt G, Forster R, Pabst O. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med. 2006;203(3):519–27.PubMedPubMedCentralCrossRef Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, Bernhardt G, Forster R, Pabst O. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med. 2006;203(3):519–27.PubMedPubMedCentralCrossRef
98.
go back to reference Laffont S, Siddiqui KR, Powrie F. Intestinal inflammation abrogates the tolerogenic properties of MLN CD103+ dendritic cells. Eur J Immunol. 2010;40(7):1877–83.PubMedCrossRef Laffont S, Siddiqui KR, Powrie F. Intestinal inflammation abrogates the tolerogenic properties of MLN CD103+ dendritic cells. Eur J Immunol. 2010;40(7):1877–83.PubMedCrossRef
99.
go back to reference Kumar S, Dwivedi PD, Das M, Tripathi A. Macrophages in food allergy: an enigma. Mol Immunol. 2013;56(4):612–8.PubMedCrossRef Kumar S, Dwivedi PD, Das M, Tripathi A. Macrophages in food allergy: an enigma. Mol Immunol. 2013;56(4):612–8.PubMedCrossRef
101.
go back to reference Cerovic V, Bain CC, Mowat AM, Milling SW. Intestinal macrophages and dendritic cells: what’s the difference? Trends Immunol. 2014;35(6):270–7.PubMedCrossRef Cerovic V, Bain CC, Mowat AM, Milling SW. Intestinal macrophages and dendritic cells: what’s the difference? Trends Immunol. 2014;35(6):270–7.PubMedCrossRef
102.
go back to reference Smeets RL, Fleuren WW, He X, Vink PM, Wijnands F, Gorecka M, Klop H, Bauerschmidt S, Garritsen A, Koenen HJ, Joosten I, Boots AM, Alkema W. Molecular pathway profiling of T lymphocyte signal transduction pathways; Th1 and Th2 genomic fingerprints are defined by TCR and CD28-mediated signaling. BMC Immunol 2012;13:12-2172-13-12. Smeets RL, Fleuren WW, He X, Vink PM, Wijnands F, Gorecka M, Klop H, Bauerschmidt S, Garritsen A, Koenen HJ, Joosten I, Boots AM, Alkema W. Molecular pathway profiling of T lymphocyte signal transduction pathways; Th1 and Th2 genomic fingerprints are defined by TCR and CD28-mediated signaling. BMC Immunol 2012;13:12-2172-13-12.
103.
go back to reference Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol. 2003;3(12):939–51.PubMedCrossRef Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol. 2003;3(12):939–51.PubMedCrossRef
104.
go back to reference Kumar S, Verma AK, Das M, Dwivedi PD. A molecular insight of CTLA-4 in food allergy. Immunol Lett. 2013;149(1–2):101–9.PubMedCrossRef Kumar S, Verma AK, Das M, Dwivedi PD. A molecular insight of CTLA-4 in food allergy. Immunol Lett. 2013;149(1–2):101–9.PubMedCrossRef
105.
go back to reference Nasta F, Corinti S, Bonura A, Colombo P, Di Felice G, Pioli C. CTLA-4 regulates allergen response by modulating GATA-3 protein level per cell. Immunology. 2007;121(1):62–70.PubMedPubMedCentralCrossRef Nasta F, Corinti S, Bonura A, Colombo P, Di Felice G, Pioli C. CTLA-4 regulates allergen response by modulating GATA-3 protein level per cell. Immunology. 2007;121(1):62–70.PubMedPubMedCentralCrossRef
106.
go back to reference van Wijk F, Nierkens S, de Jong W, Wehrens EJ, Boon L, van Kooten P, Knippels LM, Pieters R. The CD28/CTLA-4-B7 signaling pathway is involved in both allergic sensitization and tolerance induction to orally administered peanut proteins. J Immunol. 2007;178(11):6894–900.PubMedCrossRef van Wijk F, Nierkens S, de Jong W, Wehrens EJ, Boon L, van Kooten P, Knippels LM, Pieters R. The CD28/CTLA-4-B7 signaling pathway is involved in both allergic sensitization and tolerance induction to orally administered peanut proteins. J Immunol. 2007;178(11):6894–900.PubMedCrossRef
107.
go back to reference Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12(3):180–90.PubMed Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12(3):180–90.PubMed
108.
go back to reference Kuchroo VK, Das MP, Brown JA, Ranger AM, Zamvil SS, Sobel RA, Weiner HL, Nabavi N, Glimcher LH. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy. Cell. 1995;80(5):707–18.PubMedCrossRef Kuchroo VK, Das MP, Brown JA, Ranger AM, Zamvil SS, Sobel RA, Weiner HL, Nabavi N, Glimcher LH. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy. Cell. 1995;80(5):707–18.PubMedCrossRef
109.
go back to reference Zhu J, Jankovic D, Oler AJ, Wei G, Sharma S, Hu G, Guo L, Yagi R, Yamane H, Punkosdy G, Feigenbaum L, Zhao K, Paul WE. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity. 2012;37(4):660–73.PubMedPubMedCentralCrossRef Zhu J, Jankovic D, Oler AJ, Wei G, Sharma S, Hu G, Guo L, Yagi R, Yamane H, Punkosdy G, Feigenbaum L, Zhao K, Paul WE. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity. 2012;37(4):660–73.PubMedPubMedCentralCrossRef
110.
go back to reference Wei G, Abraham BJ, Yagi R, Jothi R, Cui K, Sharma S, Narlikar L, Northrup DL, Tang Q, Paul WE, Zhu J, Zhao K. Genome-wide analyses of transcription factor GATA3-mediated gene regulation in distinct T cell types. Immunity. 2011;35(2):299–311.PubMedPubMedCentralCrossRef Wei G, Abraham BJ, Yagi R, Jothi R, Cui K, Sharma S, Narlikar L, Northrup DL, Tang Q, Paul WE, Zhu J, Zhao K. Genome-wide analyses of transcription factor GATA3-mediated gene regulation in distinct T cell types. Immunity. 2011;35(2):299–311.PubMedPubMedCentralCrossRef
111.
go back to reference Ho IC, Tai TS, Pai SY. GATA3 and the T-cell lineage: essential functions before and after T-helper-2-cell differentiation. Nat Rev Immunol. 2009;9(2):125–35.PubMedPubMedCentralCrossRef Ho IC, Tai TS, Pai SY. GATA3 and the T-cell lineage: essential functions before and after T-helper-2-cell differentiation. Nat Rev Immunol. 2009;9(2):125–35.PubMedPubMedCentralCrossRef
112.
113.
go back to reference Liao W, Lin JX, Leonard WJ. IL-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation. Curr Opin Immunol. 2011;23(5):598–604.PubMedPubMedCentralCrossRef Liao W, Lin JX, Leonard WJ. IL-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation. Curr Opin Immunol. 2011;23(5):598–604.PubMedPubMedCentralCrossRef
114.
go back to reference Cote-Sierra J, Foucras G, Guo L, Chiodetti L, Young HA, Hu-Li J, Zhu J, Paul WE. Interleukin 2 plays a central role in Th2 differentiation. Proc Natl Acad Sci USA. 2004;101(11):3880–5.PubMedPubMedCentralCrossRef Cote-Sierra J, Foucras G, Guo L, Chiodetti L, Young HA, Hu-Li J, Zhu J, Paul WE. Interleukin 2 plays a central role in Th2 differentiation. Proc Natl Acad Sci USA. 2004;101(11):3880–5.PubMedPubMedCentralCrossRef
115.
go back to reference Chu DK, Mohammed-Ali Z, Jimenez-Saiz R, Walker TD, Goncharova S, Llop-Guevara A, Kong J, Gordon ME, Barra NG, Gillgrass AE, Van Seggelen H, Khan WI, Ashkar AA, Bramson JL, Humbles AA, Kolbeck R, Waserman S, Jordana M. T helper cell IL-4 drives intestinal Th2 priming to oral peanut antigen, under the control of OX40L and independent of innate-like lymphocytes. Mucosal Immunol. 2014;7(6):1395–404.PubMedCrossRef Chu DK, Mohammed-Ali Z, Jimenez-Saiz R, Walker TD, Goncharova S, Llop-Guevara A, Kong J, Gordon ME, Barra NG, Gillgrass AE, Van Seggelen H, Khan WI, Ashkar AA, Bramson JL, Humbles AA, Kolbeck R, Waserman S, Jordana M. T helper cell IL-4 drives intestinal Th2 priming to oral peanut antigen, under the control of OX40L and independent of innate-like lymphocytes. Mucosal Immunol. 2014;7(6):1395–404.PubMedCrossRef
116.
117.
go back to reference Angkasekwinai P, Park H, Wang YH, Wang YH, Chang SH, Corry DB, Liu YJ, Zhu Z, Dong C. Interleukin 25 promotes the initiation of proallergic type 2 responses. J Exp Med. 2007;204(7):1509–17.PubMedPubMedCentralCrossRef Angkasekwinai P, Park H, Wang YH, Wang YH, Chang SH, Corry DB, Liu YJ, Zhu Z, Dong C. Interleukin 25 promotes the initiation of proallergic type 2 responses. J Exp Med. 2007;204(7):1509–17.PubMedPubMedCentralCrossRef
118.
go back to reference Zhao Q, Chen G. Role of IL-33 and its receptor in T cell-mediated autoimmune diseases. Biomed Res Int. 2014;2014:587376.PubMedPubMedCentral Zhao Q, Chen G. Role of IL-33 and its receptor in T cell-mediated autoimmune diseases. Biomed Res Int. 2014;2014:587376.PubMedPubMedCentral
119.
go back to reference Maciolek JA, Pasternak JA, Wilson HL. Metabolism of activated T lymphocytes. Curr Opin Immunol. 2014;27:60–74.PubMedCrossRef Maciolek JA, Pasternak JA, Wilson HL. Metabolism of activated T lymphocytes. Curr Opin Immunol. 2014;27:60–74.PubMedCrossRef
120.
go back to reference Melnik BC. The potential mechanistic link between allergy and obesity development and infant formula feeding. Allergy Asthma Clin Immunol 2014;10(1):37-1492-10-37 (eCollection 2014). Melnik BC. The potential mechanistic link between allergy and obesity development and infant formula feeding. Allergy Asthma Clin Immunol 2014;10(1):37-1492-10-37 (eCollection 2014).
121.
go back to reference Xu Z, Zan H, Pone EJ, Mai T, Casali P. Immunoglobulin class-switch DNA recombination: induction, targeting and beyond. Nat Rev Immunol. 2012;12(7):517–31.PubMedPubMedCentralCrossRef Xu Z, Zan H, Pone EJ, Mai T, Casali P. Immunoglobulin class-switch DNA recombination: induction, targeting and beyond. Nat Rev Immunol. 2012;12(7):517–31.PubMedPubMedCentralCrossRef
122.
go back to reference Berkowska MA, Heeringa JJ, Hajdarbegovic E, van der Burg M, Thio HB, van Hagen PM, Boon L, Orfao A, van Dongen JJ, van Zelm MC. Human IgE(+) B cells are derived from T cell-dependent and T cell-independent pathways. J Allergy Clin Immunol 2014;134(3):688–97.e6. Berkowska MA, Heeringa JJ, Hajdarbegovic E, van der Burg M, Thio HB, van Hagen PM, Boon L, Orfao A, van Dongen JJ, van Zelm MC. Human IgE(+) B cells are derived from T cell-dependent and T cell-independent pathways. J Allergy Clin Immunol 2014;134(3):688–97.e6.
123.
go back to reference Xiong H, Dolpady J, Wabl M, Curotto de Lafaille MA, Lafaille JJ. Sequential class switching is required for the generation of high affinity IgE antibodies. J Exp Med. 2012;209(2):353–64.PubMedPubMedCentralCrossRef Xiong H, Dolpady J, Wabl M, Curotto de Lafaille MA, Lafaille JJ. Sequential class switching is required for the generation of high affinity IgE antibodies. J Exp Med. 2012;209(2):353–64.PubMedPubMedCentralCrossRef
124.
go back to reference Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14(3):141–53.PubMedCrossRef Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14(3):141–53.PubMedCrossRef
125.
go back to reference Vascotto F, Lankar D, Faure-Andre G, Vargas P, Diaz J, Le Roux D, Yuseff MI, Sibarita JB, Boes M, Raposo G, Mougneau E, Glaichenhaus N, Bonnerot C, Manoury B, Lennon-Dumenil AM. The actin-based motor protein myosin II regulates MHC class II trafficking and BCR-driven antigen presentation. J Cell Biol. 2007;176(7):1007–19.PubMedPubMedCentralCrossRef Vascotto F, Lankar D, Faure-Andre G, Vargas P, Diaz J, Le Roux D, Yuseff MI, Sibarita JB, Boes M, Raposo G, Mougneau E, Glaichenhaus N, Bonnerot C, Manoury B, Lennon-Dumenil AM. The actin-based motor protein myosin II regulates MHC class II trafficking and BCR-driven antigen presentation. J Cell Biol. 2007;176(7):1007–19.PubMedPubMedCentralCrossRef
126.
go back to reference van Ree R, Hummelshoj L, Plantinga M, Poulsen LK, Swindle E: Allergic sensitization: host-immune factors. Clin Transl Allergy 2014;4(1):12-7022-4-12. van Ree R, Hummelshoj L, Plantinga M, Poulsen LK, Swindle E: Allergic sensitization: host-immune factors. Clin Transl Allergy 2014;4(1):12-7022-4-12.
127.
go back to reference Park SR, Zan H, Pal Z, Zhang J, Al-Qahtani A, Pone EJ, Xu Z, Mai T, Casali P. HoxC4 binds to the promoter of the cytidine deaminase AID gene to induce AID expression, class-switch DNA recombination and somatic hypermutation. Nat Immunol. 2009;10(5):540–50.PubMedPubMedCentralCrossRef Park SR, Zan H, Pal Z, Zhang J, Al-Qahtani A, Pone EJ, Xu Z, Mai T, Casali P. HoxC4 binds to the promoter of the cytidine deaminase AID gene to induce AID expression, class-switch DNA recombination and somatic hypermutation. Nat Immunol. 2009;10(5):540–50.PubMedPubMedCentralCrossRef
128.
go back to reference Geha RS, Jabara HH, Brodeur SR. The regulation of immunoglobulin E class-switch recombination. Nat Rev Immunol. 2003;3(9):721–32.PubMedCrossRef Geha RS, Jabara HH, Brodeur SR. The regulation of immunoglobulin E class-switch recombination. Nat Rev Immunol. 2003;3(9):721–32.PubMedCrossRef
129.
go back to reference Wu LC, Scheerens H. Targeting IgE production in mice and humans. Curr Opin Immunol. 2014;31:8–15.PubMedCrossRef Wu LC, Scheerens H. Targeting IgE production in mice and humans. Curr Opin Immunol. 2014;31:8–15.PubMedCrossRef
130.
go back to reference Takai T, Kato T, Ota M, Yasueda H, Kuhara T, Okumura K, Ogawa H. Recombinant Der p 1 and Der f 1 with in vitro enzymatic activity to cleave human CD23, CD25 and alpha1-antitrypsin, and in vivo IgE-eliciting activity in mice. Int Arch Allergy Immunol. 2005;137(3):194–200.PubMedCrossRef Takai T, Kato T, Ota M, Yasueda H, Kuhara T, Okumura K, Ogawa H. Recombinant Der p 1 and Der f 1 with in vitro enzymatic activity to cleave human CD23, CD25 and alpha1-antitrypsin, and in vivo IgE-eliciting activity in mice. Int Arch Allergy Immunol. 2005;137(3):194–200.PubMedCrossRef
131.
go back to reference Gough L, Schulz O, Sewell HF, Shakib F. The cysteine protease activity of the major dust mite allergen Der p 1 selectively enhances the immunoglobulin E antibody response. J Exp Med. 1999;190(12):1897–902.PubMedPubMedCentralCrossRef Gough L, Schulz O, Sewell HF, Shakib F. The cysteine protease activity of the major dust mite allergen Der p 1 selectively enhances the immunoglobulin E antibody response. J Exp Med. 1999;190(12):1897–902.PubMedPubMedCentralCrossRef
132.
go back to reference Mayer RJ, Bolognese BJ, Al-Mahdi N, Cook RM, Flamberg PL, Hansbury MJ, Khandekar S, Appelbaum E, Faller A, Marshall LA. Inhibition of CD23 processing correlates with inhibition of IL-4-stimulated IgE production in human PBL and hu-PBL-reconstituted SCID mice. Clin Exp Allergy. 2000;30(5):719–27.PubMedCrossRef Mayer RJ, Bolognese BJ, Al-Mahdi N, Cook RM, Flamberg PL, Hansbury MJ, Khandekar S, Appelbaum E, Faller A, Marshall LA. Inhibition of CD23 processing correlates with inhibition of IL-4-stimulated IgE production in human PBL and hu-PBL-reconstituted SCID mice. Clin Exp Allergy. 2000;30(5):719–27.PubMedCrossRef
133.
go back to reference Cubells-Baeza N, Verhoeckx KCM, Larre C, Denery-Papini S, Gavrovic-Jankulovic M, Diaz Perales A. Applicability of epithelial models in protein permeability/transport studies and food allergy. Drug Discovery Today: Disease Models. 2015;17–18:13–21. Cubells-Baeza N, Verhoeckx KCM, Larre C, Denery-Papini S, Gavrovic-Jankulovic M, Diaz Perales A. Applicability of epithelial models in protein permeability/transport studies and food allergy. Drug Discovery Today: Disease Models. 2015;17–18:13–21.
134.
go back to reference Vickery BP, Burks AW. Immunotherapy in the treatment of food allergy: focus on oral tolerance. Curr Opin Allergy Clin Immunol. 2009;9(4):364–70.PubMedCrossRef Vickery BP, Burks AW. Immunotherapy in the treatment of food allergy: focus on oral tolerance. Curr Opin Allergy Clin Immunol. 2009;9(4):364–70.PubMedCrossRef
135.
go back to reference Noval Rivas M, Burton OT, Wise P, Charbonnier LM, Georgiev P, Oettgen HC, Rachid R, Chatila TA. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42(3):512–23.PubMedCrossRef Noval Rivas M, Burton OT, Wise P, Charbonnier LM, Georgiev P, Oettgen HC, Rachid R, Chatila TA. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42(3):512–23.PubMedCrossRef
136.
go back to reference Schmitt EG, Haribhai D, Williams JB, Aggarwal P, Jia S, Charbonnier LM, Yan K, Lorier R, Turner A, Ziegelbauer J, Georgiev P, Simpson P, Salzman NH, Hessner MJ, Broeckel U, Chatila TA, Williams CB. IL-10 produced by induced regulatory T cells (iTregs) controls colitis and pathogenic ex-iTregs during immunotherapy. J Immunol. 2012;189(12):5638–48.PubMedPubMedCentralCrossRef Schmitt EG, Haribhai D, Williams JB, Aggarwal P, Jia S, Charbonnier LM, Yan K, Lorier R, Turner A, Ziegelbauer J, Georgiev P, Simpson P, Salzman NH, Hessner MJ, Broeckel U, Chatila TA, Williams CB. IL-10 produced by induced regulatory T cells (iTregs) controls colitis and pathogenic ex-iTregs during immunotherapy. J Immunol. 2012;189(12):5638–48.PubMedPubMedCentralCrossRef
137.
go back to reference Berin MC, Shreffler WG. Mechanisms underlying induction of tolerance to foods. Immunol Allergy Clin North Am. 2016;36(1):87–102.PubMedCrossRef Berin MC, Shreffler WG. Mechanisms underlying induction of tolerance to foods. Immunol Allergy Clin North Am. 2016;36(1):87–102.PubMedCrossRef
138.
go back to reference Buyuktiryaki B, Sahiner UM, Girgin G, Birben E, Soyer OU, Cavkaytar O, Cetin C, Arik Yilmaz E, Yavuz ST, Kalayci O, Baydar T, Sackesen C. Low indoleamine 2,3-dioxygenase activity in persistent food allergy in children. Allergy. 2016;71(2):258–66.PubMedCrossRef Buyuktiryaki B, Sahiner UM, Girgin G, Birben E, Soyer OU, Cavkaytar O, Cetin C, Arik Yilmaz E, Yavuz ST, Kalayci O, Baydar T, Sackesen C. Low indoleamine 2,3-dioxygenase activity in persistent food allergy in children. Allergy. 2016;71(2):258–66.PubMedCrossRef
139.
go back to reference Chinthrajah RS, Hernandez JD, Boyd SD, Galli SJ, Nadeau KC. Molecular and cellular mechanisms of food allergy and food tolerance. J Allergy Clin Immunol. 2016;137(4):984–97.PubMedPubMedCentralCrossRef Chinthrajah RS, Hernandez JD, Boyd SD, Galli SJ, Nadeau KC. Molecular and cellular mechanisms of food allergy and food tolerance. J Allergy Clin Immunol. 2016;137(4):984–97.PubMedPubMedCentralCrossRef
141.
go back to reference Scott CL, Aumeunier AM, Mowat AM. Intestinal CD103+ dendritic cells: master regulators of tolerance? Trends Immunol. 2011;32(9):412–9.PubMedCrossRef Scott CL, Aumeunier AM, Mowat AM. Intestinal CD103+ dendritic cells: master regulators of tolerance? Trends Immunol. 2011;32(9):412–9.PubMedCrossRef
142.
go back to reference Hadis U, Wahl B, Schulz O, Hardtke-Wolenski M, Schippers A, Wagner N, Muller W, Sparwasser T, Forster R, Pabst O. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 2011;34(2):237–46.PubMedCrossRef Hadis U, Wahl B, Schulz O, Hardtke-Wolenski M, Schippers A, Wagner N, Muller W, Sparwasser T, Forster R, Pabst O. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 2011;34(2):237–46.PubMedCrossRef
143.
go back to reference Shiokawa A, Tanabe K, Tsuji NM, Sato R, Hachimura S. IL-10 and IL-27 producing dendritic cells capable of enhancing IL-10 production of T cells are induced in oral tolerance. Immunol Lett. 2009;125(1):7–14.PubMedCrossRef Shiokawa A, Tanabe K, Tsuji NM, Sato R, Hachimura S. IL-10 and IL-27 producing dendritic cells capable of enhancing IL-10 production of T cells are induced in oral tolerance. Immunol Lett. 2009;125(1):7–14.PubMedCrossRef
145.
go back to reference MaruYama T. TGF-beta-induced IκB-zeta controls Foxp3 gene expression. Biochem Biophys Res Commun. 2015;464(2):586–9.PubMedCrossRef MaruYama T. TGF-beta-induced IκB-zeta controls Foxp3 gene expression. Biochem Biophys Res Commun. 2015;464(2):586–9.PubMedCrossRef
146.
go back to reference Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, Hall JA, Yagi R, Naik S, Bhairavabhotla R, Paul WE, Bosselut R, Wei G, Zhao K, Oukka M, Zhu J, Belkaid Y. GATA3 controls Foxp3(+) regulatory T cell fate during inflammation in mice. J Clin Investig. 2011;121(11):4503–15.PubMedPubMedCentralCrossRef Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, Hall JA, Yagi R, Naik S, Bhairavabhotla R, Paul WE, Bosselut R, Wei G, Zhao K, Oukka M, Zhu J, Belkaid Y. GATA3 controls Foxp3(+) regulatory T cell fate during inflammation in mice. J Clin Investig. 2011;121(11):4503–15.PubMedPubMedCentralCrossRef
148.
go back to reference van de Veen W, Stanic B, Wirz OF, Jansen K, Globinska A, Akdis M. Role of regulatory B cells in immune tolerance to allergens and beyond. J Allergy Clin Immunol. 2016;138(3):654–65.PubMedCrossRef van de Veen W, Stanic B, Wirz OF, Jansen K, Globinska A, Akdis M. Role of regulatory B cells in immune tolerance to allergens and beyond. J Allergy Clin Immunol. 2016;138(3):654–65.PubMedCrossRef
149.
go back to reference Noh J, Lee JH, Noh G, Bang SY, Kim HS, Choi WS, Cho S, Lee SS. Characterisation of allergen-specific responses of IL-10-producing regulatory B cells (Br 1) in Cow Milk Allergy. Cell Immunol. 2010;264(2):143–9.PubMedCrossRef Noh J, Lee JH, Noh G, Bang SY, Kim HS, Choi WS, Cho S, Lee SS. Characterisation of allergen-specific responses of IL-10-producing regulatory B cells (Br 1) in Cow Milk Allergy. Cell Immunol. 2010;264(2):143–9.PubMedCrossRef
151.
go back to reference Liu ZQ, Wu Y, Song JP, Liu X, Liu Z, Zheng PY, Yang PC. Tolerogenic CX3CR1+ B cells suppress food allergy-induced intestinal inflammation in mice. Allergy. 2013;68(10):1241–8.PubMedCrossRef Liu ZQ, Wu Y, Song JP, Liu X, Liu Z, Zheng PY, Yang PC. Tolerogenic CX3CR1+ B cells suppress food allergy-induced intestinal inflammation in mice. Allergy. 2013;68(10):1241–8.PubMedCrossRef
152.
go back to reference Saluja R, Khan M, Church MK, Maurer M. The role of IL-33 and mast cells in allergy and inflammation. Clin Transl Allergy 2015;5:33-015-0076-5 (eCollection 2015). Saluja R, Khan M, Church MK, Maurer M. The role of IL-33 and mast cells in allergy and inflammation. Clin Transl Allergy 2015;5:33-015-0076-5 (eCollection 2015).
153.
go back to reference Asokananthan N, Graham PT, Stewart DJ, Bakker AJ, Eidne KA, Thompson PJ, Stewart GA. House dust mite allergens induce proinflammatory cytokines from respiratory epithelial cells: the cysteine protease allergen, Der p 1, activates protease-activated receptor (PAR)-2 and inactivates PAR-1. J Immunol. 2002;169(8):4572–8.PubMedCrossRef Asokananthan N, Graham PT, Stewart DJ, Bakker AJ, Eidne KA, Thompson PJ, Stewart GA. House dust mite allergens induce proinflammatory cytokines from respiratory epithelial cells: the cysteine protease allergen, Der p 1, activates protease-activated receptor (PAR)-2 and inactivates PAR-1. J Immunol. 2002;169(8):4572–8.PubMedCrossRef
154.
go back to reference Palomares O. The role of regulatory T cells in IgE-mediated food allergy. J Investig Allergol Clin Immunol 2013;23(6):371–82 (quiz 2 p preceding 382). Palomares O. The role of regulatory T cells in IgE-mediated food allergy. J Investig Allergol Clin Immunol 2013;23(6):371–82 (quiz 2 p preceding 382).
Metadata
Title
Application of the adverse outcome pathway (AOP) concept to structure the available in vivo and in vitro mechanistic data for allergic sensitization to food proteins
Authors
Jolanda H. M. van Bilsen
Edyta Sienkiewicz-Szłapka
Daniel Lozano-Ojalvo
Linette E. M. Willemsen
Celia M. Antunes
Elena Molina
Joost J. Smit
Barbara Wróblewska
Harry J. Wichers
Edward F. Knol
Gregory S. Ladics
Raymond H. H. Pieters
Sandra Denery-Papini
Yvonne M. Vissers
Simona L. Bavaro
Colette Larré
Kitty C. M. Verhoeckx
Erwin L. Roggen
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Clinical and Translational Allergy / Issue 1/2017
Electronic ISSN: 2045-7022
DOI
https://doi.org/10.1186/s13601-017-0152-0

Other articles of this Issue 1/2017

Clinical and Translational Allergy 1/2017 Go to the issue