Skip to main content
Top
Published in: Clinical and Translational Allergy 1/2016

Open Access 01-12-2016 | Review

Current challenges facing the assessment of the allergenic capacity of food allergens in animal models

Authors: Katrine Lindholm Bøgh, Jolanda van Bilsen, Robert Głogowski, Iván López-Expósito, Grégory Bouchaud, Carine Blanchard, Marie Bodinier, Joost Smit, Raymond Pieters, Shanna Bastiaan-Net, Nicole de Wit, Eva Untersmayr, Karine Adel-Patient, Leon Knippels, Michelle M. Epstein, Mario Noti, Unni Cecilie Nygaard, Ian Kimber, Kitty Verhoeckx, Liam O’Mahony

Published in: Clinical and Translational Allergy | Issue 1/2016

Login to get access

Abstract

Food allergy is a major health problem of increasing concern. The insufficiency of protein sources for human nutrition in a world with a growing population is also a significant problem. The introduction of new protein sources into the diet, such as newly developed innovative foods or foods produced using new technologies and production processes, insects, algae, duckweed, or agricultural products from third countries, creates the opportunity for development of new food allergies, and this in turn has driven the need to develop test methods capable of characterizing the allergenic potential of novel food proteins. There is no doubt that robust and reliable animal models for the identification and characterization of food allergens would be valuable tools for safety assessment. However, although various animal models have been proposed for this purpose, to date, none have been formally validated as predictive and none are currently suitable to test the allergenic potential of new foods. Here, the design of various animal models are reviewed, including among others considerations of species and strain, diet, route of administration, dose and formulation of the test protein, relevant controls and endpoints measured.
Literature
1.
go back to reference Muraro A, Werfel T, Hoffmann-Sommergruber K, Roberts G, Beyer K, Bindslev-Jensen C, et al. EAACI food allergy and anaphylaxis guidelines: diagnosis and management of food allergy. Allergy. 2014;69:1008–25.PubMedCrossRef Muraro A, Werfel T, Hoffmann-Sommergruber K, Roberts G, Beyer K, Bindslev-Jensen C, et al. EAACI food allergy and anaphylaxis guidelines: diagnosis and management of food allergy. Allergy. 2014;69:1008–25.PubMedCrossRef
2.
go back to reference Muraro A, Halken S, Arshad SH, Beyer K, Dubois AE, Du TG, et al. EAACI food allergy and anaphylaxis guidelines. Primary prevention of food allergy. Allergy. 2014;69:590–601.PubMedCrossRef Muraro A, Halken S, Arshad SH, Beyer K, Dubois AE, Du TG, et al. EAACI food allergy and anaphylaxis guidelines. Primary prevention of food allergy. Allergy. 2014;69:590–601.PubMedCrossRef
3.
go back to reference McClain S, Bannon GA. Animal models of food allergy: opportunities and barriers. Curr Allergy Asthma Rep. 2006;6:141–4.PubMedCrossRef McClain S, Bannon GA. Animal models of food allergy: opportunities and barriers. Curr Allergy Asthma Rep. 2006;6:141–4.PubMedCrossRef
4.
go back to reference Radauer C, Bublin M, Wagner S, Mari A, Breiteneder H. Allergens are distributed into few protein families and possess a restricted number of biochemical functions. J Allergy Clin Immunol. 2008;121:847–52.PubMedCrossRef Radauer C, Bublin M, Wagner S, Mari A, Breiteneder H. Allergens are distributed into few protein families and possess a restricted number of biochemical functions. J Allergy Clin Immunol. 2008;121:847–52.PubMedCrossRef
5.
go back to reference Lin J, Shewry PR, Archer DB, Beyer K, Niggemann B, Haas H, et al. The potential allergenicity of two 2S albumins from soybean (Glycine max): a protein microarray approach. Int Arch Allergy Immunol. 2006;141:91–102.PubMedCrossRef Lin J, Shewry PR, Archer DB, Beyer K, Niggemann B, Haas H, et al. The potential allergenicity of two 2S albumins from soybean (Glycine max): a protein microarray approach. Int Arch Allergy Immunol. 2006;141:91–102.PubMedCrossRef
6.
go back to reference Kroghsbo S, Bøgh KL, Rigby NM, Mills EN, Rogers A, Madsen CB. Sensitization with 7S globulins from peanut, hazelnut, soy or pea induces IgE with different biological activities which are modified by soy tolerance. Int Arch Allergy Immunol. 2011;155:212–24.PubMedCrossRef Kroghsbo S, Bøgh KL, Rigby NM, Mills EN, Rogers A, Madsen CB. Sensitization with 7S globulins from peanut, hazelnut, soy or pea induces IgE with different biological activities which are modified by soy tolerance. Int Arch Allergy Immunol. 2011;155:212–24.PubMedCrossRef
7.
go back to reference Hazebrouck S, Ah-Leung S, Bidat E, Paty E, Drumare MF, Tilleul S, et al. Goat’s milk allergy without cow’s milk allergy: suppression of non-cross-reactive epitopes on caprine β-casein. Clin Exp Allergy. 2014;44:602–10.PubMedCrossRef Hazebrouck S, Ah-Leung S, Bidat E, Paty E, Drumare MF, Tilleul S, et al. Goat’s milk allergy without cow’s milk allergy: suppression of non-cross-reactive epitopes on caprine β-casein. Clin Exp Allergy. 2014;44:602–10.PubMedCrossRef
8.
go back to reference Astwood JD, Leach JN, Fuchs RL. Stability of food allergens to digestion in vitro. Nat Biotechnol. 1996;14:1269–73.PubMedCrossRef Astwood JD, Leach JN, Fuchs RL. Stability of food allergens to digestion in vitro. Nat Biotechnol. 1996;14:1269–73.PubMedCrossRef
9.
go back to reference Bøgh KL, Madsen CB. Food allergens: is there a correlation between stability to digestion and allergenicity? Crit Rev Food Sci Nutr. 2015. [Epub ahead of print] Bøgh KL, Madsen CB. Food allergens: is there a correlation between stability to digestion and allergenicity? Crit Rev Food Sci Nutr. 2015. [Epub ahead of print]
10.
go back to reference Fu TT, Abbott UR, Hatzos C. Digestibility of food allergens and nonallergenic proteins in simulated gastric fluid and simulated intestinal fluid—a comparative study. J Agric Food Chem. 2002;50:7154–60.PubMedCrossRef Fu TT, Abbott UR, Hatzos C. Digestibility of food allergens and nonallergenic proteins in simulated gastric fluid and simulated intestinal fluid—a comparative study. J Agric Food Chem. 2002;50:7154–60.PubMedCrossRef
11.
go back to reference Dearman RJ, Kimber I. Determination of protein allergenicity: studies in mice. Toxicol Lett. 2001;120:181–6.PubMedCrossRef Dearman RJ, Kimber I. Determination of protein allergenicity: studies in mice. Toxicol Lett. 2001;120:181–6.PubMedCrossRef
12.
go back to reference Ladics GS, Knippels LM, Penninks AH, Bannon GA, Goodman RE, Herouet-Guicheney C. Review of animal models designed to predict the potential allergenicity of novel proteins in genetically modified crops. Regul Toxicol Pharmacol. 2010;56:212–24.PubMedCrossRef Ladics GS, Knippels LM, Penninks AH, Bannon GA, Goodman RE, Herouet-Guicheney C. Review of animal models designed to predict the potential allergenicity of novel proteins in genetically modified crops. Regul Toxicol Pharmacol. 2010;56:212–24.PubMedCrossRef
13.
go back to reference Bowman CC, Selgrade MK. Differences in allergenic potential of food extracts following oral exposure in mice reflect differences in digestibility: potential approaches to safety assessment. Toxicol Sci. 2008;102:100–9.PubMedCrossRef Bowman CC, Selgrade MK. Differences in allergenic potential of food extracts following oral exposure in mice reflect differences in digestibility: potential approaches to safety assessment. Toxicol Sci. 2008;102:100–9.PubMedCrossRef
14.
go back to reference Eigenmann PA, Antonella MM, Sampson HA, Wahn U. iPAC: an initiative to fight the burden of allergies in children. Pediatr Allergy Immunol. 2008;19:1–3.CrossRef Eigenmann PA, Antonella MM, Sampson HA, Wahn U. iPAC: an initiative to fight the burden of allergies in children. Pediatr Allergy Immunol. 2008;19:1–3.CrossRef
15.
go back to reference Smit JJ, Willemsen K, Hassing I, Fiechter D, Storm G, van Bloois L, et al. Contribution of classic and alternative effector pathways in peanut-induced anaphylactic responses. PLoS One. 2011;6:e28917.PubMedPubMedCentralCrossRef Smit JJ, Willemsen K, Hassing I, Fiechter D, Storm G, van Bloois L, et al. Contribution of classic and alternative effector pathways in peanut-induced anaphylactic responses. PLoS One. 2011;6:e28917.PubMedPubMedCentralCrossRef
16.
go back to reference Berin MC, Zheng Y, Domaradzki M, Li XM, Sampson HA. Role of TLR4 in allergic sensitization to food proteins in mice. Allergy. 2006;61:64–71.PubMedCrossRef Berin MC, Zheng Y, Domaradzki M, Li XM, Sampson HA. Role of TLR4 in allergic sensitization to food proteins in mice. Allergy. 2006;61:64–71.PubMedCrossRef
17.
go back to reference Dearman RJ, Kimber I. Animal models of protein allergenicity: potential benefits, pitfalls and challenges. Clin Exp Allergy. 2009;39:458–68.PubMedCrossRef Dearman RJ, Kimber I. Animal models of protein allergenicity: potential benefits, pitfalls and challenges. Clin Exp Allergy. 2009;39:458–68.PubMedCrossRef
18.
go back to reference Teuber SS, Del VG, Morigasaki S, Jung HR, Eisele PH, Frick OL, Buchanan BB. The atopic dog as a model of peanut and tree nut food allergy. J Allergy Clin Immunol. 2002;110:921–7.PubMedCrossRef Teuber SS, Del VG, Morigasaki S, Jung HR, Eisele PH, Frick OL, Buchanan BB. The atopic dog as a model of peanut and tree nut food allergy. J Allergy Clin Immunol. 2002;110:921–7.PubMedCrossRef
19.
go back to reference Vinuesa M, Bassan N, Cases AI, Krumrik G. Montelukast treatment (cysteinyl leukotriene receptor antagonist) in a model of food allergy: modifications in lymphatic cell population from rectal mucosa. Rev Esp Enferm Dig. 2010;102:421–5.PubMedCrossRef Vinuesa M, Bassan N, Cases AI, Krumrik G. Montelukast treatment (cysteinyl leukotriene receptor antagonist) in a model of food allergy: modifications in lymphatic cell population from rectal mucosa. Rev Esp Enferm Dig. 2010;102:421–5.PubMedCrossRef
20.
go back to reference Dearman RJ, Kimber I. A mouse model for food allergy using intraperitoneal sensitization. Methods. 2007;41:91–8.PubMedCrossRef Dearman RJ, Kimber I. A mouse model for food allergy using intraperitoneal sensitization. Methods. 2007;41:91–8.PubMedCrossRef
21.
go back to reference Gourbeyre P, Dery-Papini S, Larre C, Gaudin JC, Brossard C, Bodinier M. Wheat gliadins modified by deamidation are more efficient than native gliadins in inducing a Th2 response in Balb/c mice experimentally sensitized to wheat allergens. Mol Nutr Food Res. 2012;56:336–44.PubMedCrossRef Gourbeyre P, Dery-Papini S, Larre C, Gaudin JC, Brossard C, Bodinier M. Wheat gliadins modified by deamidation are more efficient than native gliadins in inducing a Th2 response in Balb/c mice experimentally sensitized to wheat allergens. Mol Nutr Food Res. 2012;56:336–44.PubMedCrossRef
22.
go back to reference Dery-Papini S, Bodinier M, Pineau F, Triballeau S, Tranquet O, Adel-Patient K, et al. Immunoglobulin-E-binding epitopes of wheat allergens in patients with food allergy to wheat and in mice experimentally sensitized to wheat proteins. Clin Exp Allergy. 2011;41:1478–92.CrossRef Dery-Papini S, Bodinier M, Pineau F, Triballeau S, Tranquet O, Adel-Patient K, et al. Immunoglobulin-E-binding epitopes of wheat allergens in patients with food allergy to wheat and in mice experimentally sensitized to wheat proteins. Clin Exp Allergy. 2011;41:1478–92.CrossRef
23.
go back to reference Mine Y, Yang M. Epitope characterization of ovalbumin in BALB/c mice using different entry routes. Biochim Biophys Acta. 2007;1774:200–12.PubMedCrossRef Mine Y, Yang M. Epitope characterization of ovalbumin in BALB/c mice using different entry routes. Biochim Biophys Acta. 2007;1774:200–12.PubMedCrossRef
24.
go back to reference Li XM, Schofield BH, Huang CK, Kleiner GI, Sampson HA. A murine model of IgE-mediated cow’s milk hypersensitivity. J Allergy Clin Immunol. 1999;103:206–14.PubMedCrossRef Li XM, Schofield BH, Huang CK, Kleiner GI, Sampson HA. A murine model of IgE-mediated cow’s milk hypersensitivity. J Allergy Clin Immunol. 1999;103:206–14.PubMedCrossRef
25.
go back to reference Knippels LM, Penninks AH, Spanhaak S, Houben GF. Oral sensitization to food proteins: a Brown Norway rat model. Clin Exp Allergy. 1998;28:368–75.PubMedCrossRef Knippels LM, Penninks AH, Spanhaak S, Houben GF. Oral sensitization to food proteins: a Brown Norway rat model. Clin Exp Allergy. 1998;28:368–75.PubMedCrossRef
26.
go back to reference Li XM, Serebrisky D, Lee SY, Huang CK, Bardina L, Schofield BH, et al. A murine model of peanut anaphylaxis: T- and B-cell responses to a major peanut allergen mimic human responses. J Allergy Clin Immunol. 2000;106:150–8.PubMedCrossRef Li XM, Serebrisky D, Lee SY, Huang CK, Bardina L, Schofield BH, et al. A murine model of peanut anaphylaxis: T- and B-cell responses to a major peanut allergen mimic human responses. J Allergy Clin Immunol. 2000;106:150–8.PubMedCrossRef
27.
go back to reference Lack G, Fox D, Northstone K, Golding J. Factors associated with the development of peanut allergy in childhood. N Engl J Med. 2003;348:977–85.PubMedCrossRef Lack G, Fox D, Northstone K, Golding J. Factors associated with the development of peanut allergy in childhood. N Engl J Med. 2003;348:977–85.PubMedCrossRef
28.
go back to reference Fox AT, Sasieni P, Du TG, Syed H, Lack G. Household peanut consumption as a risk factor for the development of peanut allergy. J Allergy Clin Immunol. 2009;123:417–23.PubMedCrossRef Fox AT, Sasieni P, Du TG, Syed H, Lack G. Household peanut consumption as a risk factor for the development of peanut allergy. J Allergy Clin Immunol. 2009;123:417–23.PubMedCrossRef
29.
go back to reference Spergel JM, Mizoguchi E, Brewer JP, Martin TR, Bhan AK, Geha RS. Epicutaneous sensitization with protein antigen induces localized allergic dermatitis and hyperresponsiveness to methacholine after single exposure to aerosolized antigen in mice. J Clin Invest. 1998;101:1614–22.PubMedPubMedCentralCrossRef Spergel JM, Mizoguchi E, Brewer JP, Martin TR, Bhan AK, Geha RS. Epicutaneous sensitization with protein antigen induces localized allergic dermatitis and hyperresponsiveness to methacholine after single exposure to aerosolized antigen in mice. J Clin Invest. 1998;101:1614–22.PubMedPubMedCentralCrossRef
30.
go back to reference Strid J, Hourihane J, Kimber I, Callard R, Strobel S. Disruption of the stratum corneum allows potent epicutaneous immunization with protein antigens resulting in a dominant systemic Th2 response. Eur J Immunol. 2004;34:2100–9.PubMedCrossRef Strid J, Hourihane J, Kimber I, Callard R, Strobel S. Disruption of the stratum corneum allows potent epicutaneous immunization with protein antigens resulting in a dominant systemic Th2 response. Eur J Immunol. 2004;34:2100–9.PubMedCrossRef
31.
go back to reference Morafo V, Srivastava K, Huang CK, Kleiner G, Lee SY, Sampson HA, Li AM. Genetic susceptibility to food allergy is linked to differential TH2-TH1 responses in C3H/HeJ and BALB/c mice. J Allergy Clin Immunol. 2003;111:1122–8.PubMedCrossRef Morafo V, Srivastava K, Huang CK, Kleiner G, Lee SY, Sampson HA, Li AM. Genetic susceptibility to food allergy is linked to differential TH2-TH1 responses in C3H/HeJ and BALB/c mice. J Allergy Clin Immunol. 2003;111:1122–8.PubMedCrossRef
32.
go back to reference Madsen CB, Hattersley S, Buck J, Gendel SM, Houben GF, Hourihane JO, et al. Approaches to risk assessment in food allergy: report from a workshop ‘‘developing a framework for assessing the risk from allergenic foods”. Food Chem Toxicol. 2009;47:480–9.PubMedCrossRef Madsen CB, Hattersley S, Buck J, Gendel SM, Houben GF, Hourihane JO, et al. Approaches to risk assessment in food allergy: report from a workshop ‘‘developing a framework for assessing the risk from allergenic foods”. Food Chem Toxicol. 2009;47:480–9.PubMedCrossRef
33.
go back to reference Spanjersberg MQ, Lucas Luijckx NB, Houben GF. Risk analysis of food allergens. In: Jedrychowski L, Wichers HJ, editors. Chemical and biological properties of food allergens. CRC Press, Florence, KY, 2009. p. 387–97. Spanjersberg MQ, Lucas Luijckx NB, Houben GF. Risk analysis of food allergens. In: Jedrychowski L, Wichers HJ, editors. Chemical and biological properties of food allergens. CRC Press, Florence, KY, 2009. p. 387–97.
34.
go back to reference Crevel RW, Baumert JL, Baka A, Houben GF, Knulst AC, Kruizinga AG, et al. Development and evolution of risk assessment for food allergens. Food Chem Toxicol. 2014;67:262–76.PubMedCrossRef Crevel RW, Baumert JL, Baka A, Houben GF, Knulst AC, Kruizinga AG, et al. Development and evolution of risk assessment for food allergens. Food Chem Toxicol. 2014;67:262–76.PubMedCrossRef
36.
go back to reference Kroghsbo S, Andersen NB, Rasmussen TF, Jacobsen S, Madsen CB. Acid hydrolysis of wheat gluten induces formation of new epitopes but does not enhance sensitizing capacity by the oral route: a study in “gluten free” Brown Norway rats. PLoS One. 2014;9:e107137.PubMedPubMedCentralCrossRef Kroghsbo S, Andersen NB, Rasmussen TF, Jacobsen S, Madsen CB. Acid hydrolysis of wheat gluten induces formation of new epitopes but does not enhance sensitizing capacity by the oral route: a study in “gluten free” Brown Norway rats. PLoS One. 2014;9:e107137.PubMedPubMedCentralCrossRef
37.
go back to reference Kroghsbo S, Rigby NM, Johnson PE, Adel-Patient K, Bøgh KL, Salt LJ, et al. Assessment of the sensitizing potential of processed peanut proteins in Brown Norway rats: roasting does not enhance allergenicity. PLoS One. 2014;9:e96475.PubMedPubMedCentralCrossRef Kroghsbo S, Rigby NM, Johnson PE, Adel-Patient K, Bøgh KL, Salt LJ, et al. Assessment of the sensitizing potential of processed peanut proteins in Brown Norway rats: roasting does not enhance allergenicity. PLoS One. 2014;9:e96475.PubMedPubMedCentralCrossRef
38.
go back to reference Madsen JL, Kroghsbo S, Madsen CB, Pozdnyakova I, Barkholt V, Bøgh KL. The impact of structural integrity and route of administration on the antibody specificity against three cow’s milk allergens—a study in Brown Norway rats. Clin Transl Allergy. 2014;4:25.PubMedPubMedCentralCrossRef Madsen JL, Kroghsbo S, Madsen CB, Pozdnyakova I, Barkholt V, Bøgh KL. The impact of structural integrity and route of administration on the antibody specificity against three cow’s milk allergens—a study in Brown Norway rats. Clin Transl Allergy. 2014;4:25.PubMedPubMedCentralCrossRef
39.
go back to reference Verhoeckx KC, Vissers YM, Baumert JL, Faludi R, Feys M, Flanagan S, et al. Food processing and allergenicity. Food Chem Toxicol. 2015;80:223–40.PubMedCrossRef Verhoeckx KC, Vissers YM, Baumert JL, Faludi R, Feys M, Flanagan S, et al. Food processing and allergenicity. Food Chem Toxicol. 2015;80:223–40.PubMedCrossRef
40.
go back to reference Bernhisel-Broadbent J, Dintzis HM, Dintzis RZ, Sampson HA. Allergenicity and antigenicity of chicken egg ovomucoid (Gal d III) compared with ovalbumin (Gal d I) in children with egg allergy and in mice. J Allergy Clin Immunol. 1994;93:1047–59.PubMedCrossRef Bernhisel-Broadbent J, Dintzis HM, Dintzis RZ, Sampson HA. Allergenicity and antigenicity of chicken egg ovomucoid (Gal d III) compared with ovalbumin (Gal d I) in children with egg allergy and in mice. J Allergy Clin Immunol. 1994;93:1047–59.PubMedCrossRef
41.
go back to reference Brix S, Kjaer TM, Barkholt V, Frokiaer H. Lipopolysaccharide contamination of beta-lactoglobulin affects the immune response against intraperitoneally and orally administered antigen. Int Arch Allergy Immunol. 2004;135:216–20.PubMedCrossRef Brix S, Kjaer TM, Barkholt V, Frokiaer H. Lipopolysaccharide contamination of beta-lactoglobulin affects the immune response against intraperitoneally and orally administered antigen. Int Arch Allergy Immunol. 2004;135:216–20.PubMedCrossRef
42.
go back to reference Thomas K, MacIntosh S, Bannon G, Herouet-Guicheney C, Holsapple M, Ladics G, et al. Scientific advancement of novel protein allergenicity evaluation: an overview of work from the HESI Protein Allergenicity Technical Committee (2000–2008). Food Chem Toxicol. 2009;47:1041–50.PubMedCrossRef Thomas K, MacIntosh S, Bannon G, Herouet-Guicheney C, Holsapple M, Ladics G, et al. Scientific advancement of novel protein allergenicity evaluation: an overview of work from the HESI Protein Allergenicity Technical Committee (2000–2008). Food Chem Toxicol. 2009;47:1041–50.PubMedCrossRef
43.
go back to reference Benede S, Lopez-Exposito I, Molina E, Lopez-Fandino R. Egg proteins as allergens and the effects of the food matrix and processing. Food Funct. 2015;6:694–713.PubMedCrossRef Benede S, Lopez-Exposito I, Molina E, Lopez-Fandino R. Egg proteins as allergens and the effects of the food matrix and processing. Food Funct. 2015;6:694–713.PubMedCrossRef
44.
go back to reference Mouecoucou J, Sanchez C, Villaume C, Marrion O, Fremont S, Laurent F, Mejean L. Effects of different levels of gum arabic, low methylated pectin and xylan on in vitro digestibility of beta-lactoglobulin. J Dairy Sci. 2003;86:3857–65.PubMedCrossRef Mouecoucou J, Sanchez C, Villaume C, Marrion O, Fremont S, Laurent F, Mejean L. Effects of different levels of gum arabic, low methylated pectin and xylan on in vitro digestibility of beta-lactoglobulin. J Dairy Sci. 2003;86:3857–65.PubMedCrossRef
45.
go back to reference Schulten V, Lauer I, Scheurer S, Thalhammer T, Bohle B. A food matrix reduces digestion and absorption of food allergens in vivo. Mol Nutr Food Res. 2011;55:1484–91.PubMedCrossRef Schulten V, Lauer I, Scheurer S, Thalhammer T, Bohle B. A food matrix reduces digestion and absorption of food allergens in vivo. Mol Nutr Food Res. 2011;55:1484–91.PubMedCrossRef
46.
go back to reference Teuber SS. Hypothesis: The protein body effect and other aspects of food matrix effects. Ann N Y Acad Sci. 2002;964:111–6.PubMedCrossRef Teuber SS. Hypothesis: The protein body effect and other aspects of food matrix effects. Ann N Y Acad Sci. 2002;964:111–6.PubMedCrossRef
47.
go back to reference Mackie A, Knulst A, Le TM, Bures P, Salt L, Mills EN, et al. High fat food increases gastric residence and thus thresholds for objective symptoms in allergic patients. Mol Nutr Food Res. 2012;56:1708–14.PubMedCrossRef Mackie A, Knulst A, Le TM, Bures P, Salt L, Mills EN, et al. High fat food increases gastric residence and thus thresholds for objective symptoms in allergic patients. Mol Nutr Food Res. 2012;56:1708–14.PubMedCrossRef
48.
go back to reference van Wijk F, Nierkens S, Hassing I, Feijen M, Koppelman SJ, de Jong GAH, et al. The effect of the food matrix on in vivo immune responses to purified peanut allergens. Toxicol Sci. 2005;86:333–41.PubMedCrossRef van Wijk F, Nierkens S, Hassing I, Feijen M, Koppelman SJ, de Jong GAH, et al. The effect of the food matrix on in vivo immune responses to purified peanut allergens. Toxicol Sci. 2005;86:333–41.PubMedCrossRef
49.
go back to reference Adel-Patient K, Guimaraes VD, Paris A, Drumare MF, Ah-Leung S, Lamourette P, et al. Immunological and metabolomic impacts of administration of Cry1Ab protein and MON 810 maize in mouse. PLoS One. 2011;6:e16346.PubMedPubMedCentralCrossRef Adel-Patient K, Guimaraes VD, Paris A, Drumare MF, Ah-Leung S, Lamourette P, et al. Immunological and metabolomic impacts of administration of Cry1Ab protein and MON 810 maize in mouse. PLoS One. 2011;6:e16346.PubMedPubMedCentralCrossRef
50.
go back to reference Mirotti L, Florsheim E, Rundqvist L, Larsson G, Spinozzi F, Leite-de-Moraes M, et al. Lipids are required for the development of Brazil nut allergy: the role of mouse and human iNKT cells. Allergy. 2013;68:74–83.PubMedCrossRef Mirotti L, Florsheim E, Rundqvist L, Larsson G, Spinozzi F, Leite-de-Moraes M, et al. Lipids are required for the development of Brazil nut allergy: the role of mouse and human iNKT cells. Allergy. 2013;68:74–83.PubMedCrossRef
51.
go back to reference Kocabas CN, Sekerel BE. Does systemic exposure to aflatoxin B(1) cause allergic sensitization? Allergy. 2003;58:363–5.PubMedCrossRef Kocabas CN, Sekerel BE. Does systemic exposure to aflatoxin B(1) cause allergic sensitization? Allergy. 2003;58:363–5.PubMedCrossRef
52.
go back to reference Kunisawa J, Arita M, Hayasaka T, Harada T, Iwamoto R, Nagasawa R, et al. Dietary omega3 fatty acid exerts anti-allergic effect through the conversion to 17,18-epoxyeicosatetraenoic acid in the gut. Sci Rep. 2015;5:9750.PubMedPubMedCentralCrossRef Kunisawa J, Arita M, Hayasaka T, Harada T, Iwamoto R, Nagasawa R, et al. Dietary omega3 fatty acid exerts anti-allergic effect through the conversion to 17,18-epoxyeicosatetraenoic acid in the gut. Sci Rep. 2015;5:9750.PubMedPubMedCentralCrossRef
53.
go back to reference Schiavi E, Smolinska S, O’Mahony L. Intestinal dendritic cells. Curr Opin Gastroenterol. 2015;31:98–103.PubMedCrossRef Schiavi E, Smolinska S, O’Mahony L. Intestinal dendritic cells. Curr Opin Gastroenterol. 2015;31:98–103.PubMedCrossRef
54.
go back to reference Noti M, Kim BS, Siracusa MC, Rak GD, Kubo M, Moghaddam AE, et al. Exposure to food allergens through inflamed skin promotes intestinal food allergy through the thymic stromal lymphopoietin-basophil axis. J Allergy Clin Immunol. 2014;133:1390–9.PubMedPubMedCentralCrossRef Noti M, Kim BS, Siracusa MC, Rak GD, Kubo M, Moghaddam AE, et al. Exposure to food allergens through inflamed skin promotes intestinal food allergy through the thymic stromal lymphopoietin-basophil axis. J Allergy Clin Immunol. 2014;133:1390–9.PubMedPubMedCentralCrossRef
55.
go back to reference Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, et al. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol. 2013;131:187–200.PubMedCrossRef Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, et al. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol. 2013;131:187–200.PubMedCrossRef
56.
go back to reference Hammad H, Lambrecht BN. Barrier epithelial cells and the control of type 2 immunity. Immunity. 2015;43:29–40.PubMedCrossRef Hammad H, Lambrecht BN. Barrier epithelial cells and the control of type 2 immunity. Immunity. 2015;43:29–40.PubMedCrossRef
57.
go back to reference Bol-Schoenmakers M, Bleumink R, Marcondes RM, Mouser E, Hassing I, Ludwig I, et al. Diclofenac enhances allergic responses in a mouse peanut allergy model. Clin Exp Allergy. 2011;41:424–33.PubMedCrossRef Bol-Schoenmakers M, Bleumink R, Marcondes RM, Mouser E, Hassing I, Ludwig I, et al. Diclofenac enhances allergic responses in a mouse peanut allergy model. Clin Exp Allergy. 2011;41:424–33.PubMedCrossRef
58.
go back to reference Bol-Schoenmakers M, Marcondes RM, Bleumink R, Boon L, Man S, Hassing I, et al. Regulation by intestinal gammadelta T cells during establishment of food allergic sensitization in mice. Allergy. 2011;66:331–40.PubMedCrossRef Bol-Schoenmakers M, Marcondes RM, Bleumink R, Boon L, Man S, Hassing I, et al. Regulation by intestinal gammadelta T cells during establishment of food allergic sensitization in mice. Allergy. 2011;66:331–40.PubMedCrossRef
59.
go back to reference Frossard CP, Asigbetse KE, Burger D, Eigenmann PA. Gut T cell receptor-gammadelta(+) intraepithelial lymphocytes are activated selectively by cholera toxin to break oral tolerance in mice. Clin Exp Immunol. 2015;180:118–30.PubMedPubMedCentralCrossRef Frossard CP, Asigbetse KE, Burger D, Eigenmann PA. Gut T cell receptor-gammadelta(+) intraepithelial lymphocytes are activated selectively by cholera toxin to break oral tolerance in mice. Clin Exp Immunol. 2015;180:118–30.PubMedPubMedCentralCrossRef
61.
go back to reference Oyoshi MK, Oettgen HC, Chatila TA, Geha RS, Bryce PJ. Food allergy: insights into etiology, prevention, and treatment provided by murine models. J Allergy Clin Immunol. 2014;133:309–17.PubMedPubMedCentralCrossRef Oyoshi MK, Oettgen HC, Chatila TA, Geha RS, Bryce PJ. Food allergy: insights into etiology, prevention, and treatment provided by murine models. J Allergy Clin Immunol. 2014;133:309–17.PubMedPubMedCentralCrossRef
62.
go back to reference Heydenreich B, Bellinghausen I, Lund L, Henmar H, Lund G, Adler WP, Saloga J. Adjuvant effects of aluminium hydroxide-adsorbed allergens and allergoids—differences in vivo and in vitro. Clin Exp Immunol. 2014;176:310–9.PubMedPubMedCentralCrossRef Heydenreich B, Bellinghausen I, Lund L, Henmar H, Lund G, Adler WP, Saloga J. Adjuvant effects of aluminium hydroxide-adsorbed allergens and allergoids—differences in vivo and in vitro. Clin Exp Immunol. 2014;176:310–9.PubMedPubMedCentralCrossRef
64.
go back to reference Comoy EE, Capron A, Thyphronitis G. In vivo induction of type 1 and 2 immune responses against protein antigens. Int Immunol. 1997;9:523–31.PubMedCrossRef Comoy EE, Capron A, Thyphronitis G. In vivo induction of type 1 and 2 immune responses against protein antigens. Int Immunol. 1997;9:523–31.PubMedCrossRef
65.
go back to reference Zlatkovic J, Tsouchnikas G, Jarmer J, Koessl C, Stiasny K, Heinz FX. Aluminum hydroxide influences not only the extent but also the fine specificity and functional activity of antibody responses to tick-borne encephalitis virus in mice. J Virol. 2013;87:12187–95.PubMedPubMedCentralCrossRef Zlatkovic J, Tsouchnikas G, Jarmer J, Koessl C, Stiasny K, Heinz FX. Aluminum hydroxide influences not only the extent but also the fine specificity and functional activity of antibody responses to tick-borne encephalitis virus in mice. J Virol. 2013;87:12187–95.PubMedPubMedCentralCrossRef
66.
go back to reference Bøgh KL, Barkholt V, Madsen CB. Characterization of the immunogenicity and allergenicity of two cow’s milk hydrolysates–a study in Brown Norway rats. Scand J Immunol. 2015;81:274–83.PubMedCrossRef Bøgh KL, Barkholt V, Madsen CB. Characterization of the immunogenicity and allergenicity of two cow’s milk hydrolysates–a study in Brown Norway rats. Scand J Immunol. 2015;81:274–83.PubMedCrossRef
67.
go back to reference Jones LS, Peek LJ, Power J, Markham A, Yazzie B, Middaugh CR. Effects of adsorption to aluminum salt adjuvants on the structure and stability of model protein antigens. J Biol Chem. 2005;280:13406–14.PubMedCrossRef Jones LS, Peek LJ, Power J, Markham A, Yazzie B, Middaugh CR. Effects of adsorption to aluminum salt adjuvants on the structure and stability of model protein antigens. J Biol Chem. 2005;280:13406–14.PubMedCrossRef
68.
go back to reference Mattsson J, Schon K, Ekman L, Fahlen-Yrlid L, Yrlid U, Lycke NY. Cholera toxin adjuvant promotes a balanced Th1/Th2/Th17 response independently of IL-12 and IL-17 by acting on Gsalpha in CD11b(+) DCs. Mucosal Immunol. 2015;8:815–27.PubMedCrossRef Mattsson J, Schon K, Ekman L, Fahlen-Yrlid L, Yrlid U, Lycke NY. Cholera toxin adjuvant promotes a balanced Th1/Th2/Th17 response independently of IL-12 and IL-17 by acting on Gsalpha in CD11b(+) DCs. Mucosal Immunol. 2015;8:815–27.PubMedCrossRef
69.
go back to reference Ganeshan K, Neilsen CV, Hadsaitong A, Schleimer RP, Luo X, Bryce PJ. Impairing oral tolerance promotes allergy and anaphylaxis: a new murine food allergy model. J Allergy Clin Immunol. 2009;123:231–8.PubMedPubMedCentralCrossRef Ganeshan K, Neilsen CV, Hadsaitong A, Schleimer RP, Luo X, Bryce PJ. Impairing oral tolerance promotes allergy and anaphylaxis: a new murine food allergy model. J Allergy Clin Immunol. 2009;123:231–8.PubMedPubMedCentralCrossRef
70.
go back to reference Li J, Wang Y, Tang L, de Villiers WJ, Cohen D, Woodward J, et al. Dietary medium-chain triglycerides promote oral allergic sensitization and orally induced anaphylaxis to peanut protein in mice. J Allergy Clin Immunol. 2013;131:442–50.PubMedPubMedCentralCrossRef Li J, Wang Y, Tang L, de Villiers WJ, Cohen D, Woodward J, et al. Dietary medium-chain triglycerides promote oral allergic sensitization and orally induced anaphylaxis to peanut protein in mice. J Allergy Clin Immunol. 2013;131:442–50.PubMedPubMedCentralCrossRef
71.
go back to reference Torii I, Shimizu S, Daimon T, Shinohara Y, Kudo T, Sato A, Tsujimura T. Exposure to high doses of Lipopolysaccharide during ovalbumin sensitization prevents the development of allergic Th2 responses to a dietary antigen. J Toxicol Pathol. 2014;27:205–15.PubMedPubMedCentralCrossRef Torii I, Shimizu S, Daimon T, Shinohara Y, Kudo T, Sato A, Tsujimura T. Exposure to high doses of Lipopolysaccharide during ovalbumin sensitization prevents the development of allergic Th2 responses to a dietary antigen. J Toxicol Pathol. 2014;27:205–15.PubMedPubMedCentralCrossRef
72.
go back to reference Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70:495–505.PubMedCrossRef Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy. 2015;70:495–505.PubMedCrossRef
73.
go back to reference Birmingham NP, Parvataneni S, Hassan HM, Harkema J, Samineni S, Navuluri L, et al. An adjuvant-free mouse model of tree nut allergy using hazelnut as a model tree nut. Int Arch Allergy Immunol. 2007;144:203–10.PubMedCrossRef Birmingham NP, Parvataneni S, Hassan HM, Harkema J, Samineni S, Navuluri L, et al. An adjuvant-free mouse model of tree nut allergy using hazelnut as a model tree nut. Int Arch Allergy Immunol. 2007;144:203–10.PubMedCrossRef
74.
go back to reference Gonipeta B, Parvataneni S, Tempelman RJ, Gangur V. An adjuvant-free mouse model to evaluate the allergenicity of milk whey protein. J Dairy Sci. 2009;92:4738–44.PubMedCrossRef Gonipeta B, Parvataneni S, Tempelman RJ, Gangur V. An adjuvant-free mouse model to evaluate the allergenicity of milk whey protein. J Dairy Sci. 2009;92:4738–44.PubMedCrossRef
75.
go back to reference Chandra SA, Stokes AH, Hailey R, Merrill CL, Melich DH, DeSmet K, et al. Dermal toxicity studies: factors impacting study interpretation and outcome. Toxicol Pathol. 2015;43:474–81.PubMedCrossRef Chandra SA, Stokes AH, Hailey R, Merrill CL, Melich DH, DeSmet K, et al. Dermal toxicity studies: factors impacting study interpretation and outcome. Toxicol Pathol. 2015;43:474–81.PubMedCrossRef
76.
go back to reference Bartnikas LM, Gurish MF, Burton OT, Leisten S, Janssen E, Oettgen HC, et al. Epicutaneous sensitization results in IgE-dependent intestinal mast cell expansion and food-induced anaphylaxis. J Allergy Clin Immunol. 2013;131:451–60.PubMedPubMedCentralCrossRef Bartnikas LM, Gurish MF, Burton OT, Leisten S, Janssen E, Oettgen HC, et al. Epicutaneous sensitization results in IgE-dependent intestinal mast cell expansion and food-induced anaphylaxis. J Allergy Clin Immunol. 2013;131:451–60.PubMedPubMedCentralCrossRef
77.
go back to reference Devey ME, Anderson KJ, Coombs RR, Henschel MJ, Coates ME. The modified anaphylaxis hypothesis for cot death. Anaphylactic sensitization in guinea-pigs fed cow’s milk. Clin Exp Immunol. 1976;26:542–8.PubMedPubMedCentral Devey ME, Anderson KJ, Coombs RR, Henschel MJ, Coates ME. The modified anaphylaxis hypothesis for cot death. Anaphylactic sensitization in guinea-pigs fed cow’s milk. Clin Exp Immunol. 1976;26:542–8.PubMedPubMedCentral
78.
go back to reference Knippels LM, Houben GF, Spanhaak S, Penninks AH. An oral sensitization model in Brown Norway rats to screen for potential allergenicity of food proteins. Methods. 1999;19:78–82.PubMedCrossRef Knippels LM, Houben GF, Spanhaak S, Penninks AH. An oral sensitization model in Brown Norway rats to screen for potential allergenicity of food proteins. Methods. 1999;19:78–82.PubMedCrossRef
79.
go back to reference Knippels LMJ, Penninks AH, Houben GF. Continued expression of anti-soy protein antibodies in rats bred on a soy protein-free diet for one generation: the importance of dietary control in oral sensitization research. J Allergy Clin Immunol. 1998;101:815–20.PubMedCrossRef Knippels LMJ, Penninks AH, Houben GF. Continued expression of anti-soy protein antibodies in rats bred on a soy protein-free diet for one generation: the importance of dietary control in oral sensitization research. J Allergy Clin Immunol. 1998;101:815–20.PubMedCrossRef
80.
go back to reference Bernard H, Ah-Leung S, Drumare MF, Feraudet-Tarisse C, Verhasselt V, Wal JM, et al. Peanut allergens are rapidly transferred in human breast milk and can prevent sensitization in mice. Allergy. 2014;69:888–97.PubMedCrossRef Bernard H, Ah-Leung S, Drumare MF, Feraudet-Tarisse C, Verhasselt V, Wal JM, et al. Peanut allergens are rapidly transferred in human breast milk and can prevent sensitization in mice. Allergy. 2014;69:888–97.PubMedCrossRef
81.
go back to reference Julia V, Macia L, Dombrowicz D. The impact of diet on asthma and allergic diseases. Nat Rev Immunol. 2015;15:308–22.PubMedCrossRef Julia V, Macia L, Dombrowicz D. The impact of diet on asthma and allergic diseases. Nat Rev Immunol. 2015;15:308–22.PubMedCrossRef
82.
go back to reference Merrill AH Jr, Schmelz EM, Wang E, Dillehay DL, Rice LG, Meredith F, et al. Importance of sphingolipids and inhibitors of sphingolipid metabolism as components of animal diets. J Nutr. 1997;127:830S–3S.PubMed Merrill AH Jr, Schmelz EM, Wang E, Dillehay DL, Rice LG, Meredith F, et al. Importance of sphingolipids and inhibitors of sphingolipid metabolism as components of animal diets. J Nutr. 1997;127:830S–3S.PubMed
83.
go back to reference Hogenkamp A, Knippels LM, Garssen J, van Esch BC. Supplementation of mice with specific nondigestible oligosaccharides during pregnancy or lactation leads to diminished sensitization and allergy in the female offspring. J Nutr. 2015;145:996–1002.PubMedCrossRef Hogenkamp A, Knippels LM, Garssen J, van Esch BC. Supplementation of mice with specific nondigestible oligosaccharides during pregnancy or lactation leads to diminished sensitization and allergy in the female offspring. J Nutr. 2015;145:996–1002.PubMedCrossRef
84.
85.
go back to reference Diesner SC, Knittelfelder R, Krishnamurthy D, Pali-Scholl I, Gajdzik L, Jensen-Jarolim E, Untersmayr E. Dose-dependent food allergy induction against ovalbumin under acid-suppression: a murine food allergy model. Immunol Lett. 2008;121:45–51.PubMedPubMedCentralCrossRef Diesner SC, Knittelfelder R, Krishnamurthy D, Pali-Scholl I, Gajdzik L, Jensen-Jarolim E, Untersmayr E. Dose-dependent food allergy induction against ovalbumin under acid-suppression: a murine food allergy model. Immunol Lett. 2008;121:45–51.PubMedPubMedCentralCrossRef
86.
go back to reference Untersmayr E, Diesner SC, Bramswig KH, Knittelfelder R, Bakos N, Gundacker C, et al. Characterization of intrinsic and extrinsic risk factors for celery allergy in immunosenescence. Mech Ageing Dev. 2008;129:120–8.PubMedCrossRef Untersmayr E, Diesner SC, Bramswig KH, Knittelfelder R, Bakos N, Gundacker C, et al. Characterization of intrinsic and extrinsic risk factors for celery allergy in immunosenescence. Mech Ageing Dev. 2008;129:120–8.PubMedCrossRef
87.
go back to reference Hanson DG. Ontogeny of orally induced tolerance to soluble proteins in mice. I. Priming and tolerance in newborns. J Immunol. 1981;127:1518–24.PubMed Hanson DG. Ontogeny of orally induced tolerance to soluble proteins in mice. I. Priming and tolerance in newborns. J Immunol. 1981;127:1518–24.PubMed
88.
go back to reference Frei R, Lauener RP, Crameri R, O’Mahony L. Microbiota and dietary interactions: an update to the hygiene hypothesis? Allergy. 2012;67:451–61.PubMedCrossRef Frei R, Lauener RP, Crameri R, O’Mahony L. Microbiota and dietary interactions: an update to the hygiene hypothesis? Allergy. 2012;67:451–61.PubMedCrossRef
89.
go back to reference Konieczna P, Groeger D, Ziegler M, Frei R, Ferstl R, Shanahan F, et al. Bifidobacterium infantis 35624 administration induces Foxp3 T regulatory cells in human peripheral blood: potential role for myeloid and plasmacytoid dendritic cells. Gut. 2012;61:354–66.PubMedCrossRef Konieczna P, Groeger D, Ziegler M, Frei R, Ferstl R, Shanahan F, et al. Bifidobacterium infantis 35624 administration induces Foxp3 T regulatory cells in human peripheral blood: potential role for myeloid and plasmacytoid dendritic cells. Gut. 2012;61:354–66.PubMedCrossRef
90.
go back to reference Kelly D, King T, Aminov R. Importance of microbial colonization of the gut in early life to the development of immunity. Mutat Res. 2007;622:58–69.PubMedCrossRef Kelly D, King T, Aminov R. Importance of microbial colonization of the gut in early life to the development of immunity. Mutat Res. 2007;622:58–69.PubMedCrossRef
91.
go back to reference Frei R, Akdis M, O’Mahony L. Prebiotics, probiotics, synbiotics, and the immune system: experimental data and clinical evidence. Curr Opin Gastroenterol. 2015;31:153–8.PubMedCrossRef Frei R, Akdis M, O’Mahony L. Prebiotics, probiotics, synbiotics, and the immune system: experimental data and clinical evidence. Curr Opin Gastroenterol. 2015;31:153–8.PubMedCrossRef
92.
go back to reference Lyons A, O’Mahony D, O’Brien F, MacSharry J, Sheil B, Ceddia M, et al. Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy. 2010;40:811–9.PubMed Lyons A, O’Mahony D, O’Brien F, MacSharry J, Sheil B, Ceddia M, et al. Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy. 2010;40:811–9.PubMed
93.
go back to reference Sibartie S, O’Hara AM, Ryan J, Fanning A, O’Mahony J, O’Neill S, et al. Modulation of pathogen-induced CCL20 secretion from HT-29 human intestinal epithelial cells by commensal bacteria. BMC Immunol. 2009;10:54.PubMedPubMedCentralCrossRef Sibartie S, O’Hara AM, Ryan J, Fanning A, O’Mahony J, O’Neill S, et al. Modulation of pathogen-induced CCL20 secretion from HT-29 human intestinal epithelial cells by commensal bacteria. BMC Immunol. 2009;10:54.PubMedPubMedCentralCrossRef
94.
go back to reference O’Mahony C, Scully P, O’Mahony D, Murphy S, O’Brien F, Lyons A, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008;4:e1000112.PubMedPubMedCentralCrossRef O’Mahony C, Scully P, O’Mahony D, Murphy S, O’Brien F, Lyons A, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008;4:e1000112.PubMedPubMedCentralCrossRef
95.
go back to reference Hazebrouck S, Przybylski-Nicaise L, Ah-Leung S, del-Patient K, Corthier G, Wal JM, Rabot S. Allergic sensitization to bovine beta-lactoglobulin: comparison between germ-free and conventional BALB/c mice. Int Arch Allergy Immunol. 2009;148:65–72.PubMedCrossRef Hazebrouck S, Przybylski-Nicaise L, Ah-Leung S, del-Patient K, Corthier G, Wal JM, Rabot S. Allergic sensitization to bovine beta-lactoglobulin: comparison between germ-free and conventional BALB/c mice. Int Arch Allergy Immunol. 2009;148:65–72.PubMedCrossRef
96.
go back to reference Morin S, Bernard H, Przybylski-Nicaise L, Corthier G, Rabot S, Wal JM, Hazebrouck S. Allergenic and immunogenic potential of cow’s milk beta-lactoglobulin and caseins evidenced without adjuvant in germ-free mice. Mol Nutr Food Res. 2011;55:1700–7.PubMedCrossRef Morin S, Bernard H, Przybylski-Nicaise L, Corthier G, Rabot S, Wal JM, Hazebrouck S. Allergenic and immunogenic potential of cow’s milk beta-lactoglobulin and caseins evidenced without adjuvant in germ-free mice. Mol Nutr Food Res. 2011;55:1700–7.PubMedCrossRef
97.
go back to reference Konieczna P, Ferstl R, Ziegler M, Frei R, Nehrbass D, Lauener RP, et al. Immunomodulation by Bifidobacterium infantis 35624 in the murine lamina propria requires retinoic acid-dependent and independent mechanisms. PLoS One. 2013;8:e62617.PubMedPubMedCentralCrossRef Konieczna P, Ferstl R, Ziegler M, Frei R, Nehrbass D, Lauener RP, et al. Immunomodulation by Bifidobacterium infantis 35624 in the murine lamina propria requires retinoic acid-dependent and independent mechanisms. PLoS One. 2013;8:e62617.PubMedPubMedCentralCrossRef
98.
go back to reference McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMedCrossRef McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMedCrossRef
99.
100.
go back to reference Knippels LMJ, van Wijk F, Penninks AH. Food allergy: what do we learn from animal models? Curr Opin Allergy Clin Immunol. 2004;4:205–9.PubMedCrossRef Knippels LMJ, van Wijk F, Penninks AH. Food allergy: what do we learn from animal models? Curr Opin Allergy Clin Immunol. 2004;4:205–9.PubMedCrossRef
101.
go back to reference Kimber I, Dearman RJ, Penninks AH, Knippels LMJ, Buchanan RB, Hammerberg B, et al. Assessment of protein allergenicity on the basis of immune reactivity: animal models. Environ Health Perspect. 2003;111:1125–30.PubMedPubMedCentralCrossRef Kimber I, Dearman RJ, Penninks AH, Knippels LMJ, Buchanan RB, Hammerberg B, et al. Assessment of protein allergenicity on the basis of immune reactivity: animal models. Environ Health Perspect. 2003;111:1125–30.PubMedPubMedCentralCrossRef
102.
go back to reference Strait RT, Morris SC, Yang M, Qu XW, Finkelman FD. Pathways of anaphylaxis in the mouse. J Allergy Clin Immunol. 2002;109:658–68.PubMedCrossRef Strait RT, Morris SC, Yang M, Qu XW, Finkelman FD. Pathways of anaphylaxis in the mouse. J Allergy Clin Immunol. 2002;109:658–68.PubMedCrossRef
103.
go back to reference Evans H, Killoran KE, Mitre E. Measuring local anaphylaxis in mice. J Vis Exp. 2014;92:e52005.PubMed Evans H, Killoran KE, Mitre E. Measuring local anaphylaxis in mice. J Vis Exp. 2014;92:e52005.PubMed
104.
105.
go back to reference Cockcroft DW, Davis BE. Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol. 2006;118:551–9.PubMedCrossRef Cockcroft DW, Davis BE. Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol. 2006;118:551–9.PubMedCrossRef
106.
go back to reference Schouten B, van Esch BC, Hofman GA, van den Elsen LW, Willemsen LE, Garssen J. Acute allergic skin reactions and intestinal contractility changes in mice orally sensitized against casein or whey. Int Arch Allergy Immunol. 2008;147:125–34.PubMedCrossRef Schouten B, van Esch BC, Hofman GA, van den Elsen LW, Willemsen LE, Garssen J. Acute allergic skin reactions and intestinal contractility changes in mice orally sensitized against casein or whey. Int Arch Allergy Immunol. 2008;147:125–34.PubMedCrossRef
107.
go back to reference van Esch BC, Schouten B, Hofman GA, van Baalen T, Nijkamp FP, Knippels LM, et al. Acute allergic skin response as a new tool to evaluate the allergenicity of whey hydrolysates in a mouse model of orally induced cow’s milk allergy. Pediatr Allergy Immunol. 2010;21:e780–6.PubMedCrossRef van Esch BC, Schouten B, Hofman GA, van Baalen T, Nijkamp FP, Knippels LM, et al. Acute allergic skin response as a new tool to evaluate the allergenicity of whey hydrolysates in a mouse model of orally induced cow’s milk allergy. Pediatr Allergy Immunol. 2010;21:e780–6.PubMedCrossRef
108.
go back to reference Prescott VE, Forbes E, Foster PS, Matthaei K, Hogan SP. Mechanistic analysis of experimental food allergen-induced cutaneous reactions. J Leukoc Biol. 2006;80:258–66.PubMedCrossRef Prescott VE, Forbes E, Foster PS, Matthaei K, Hogan SP. Mechanistic analysis of experimental food allergen-induced cutaneous reactions. J Leukoc Biol. 2006;80:258–66.PubMedCrossRef
109.
go back to reference Mathias CB, Hobson SA, Garcia-Lloret M, Lawson G, Poddighe D, Freyschmidt EJ, et al. IgE-mediated systemic anaphylaxis and impaired tolerance to food antigens in mice with enhanced IL-4 receptor signaling. J Allergy Clin Immunol. 2011;127:795–805.PubMedPubMedCentralCrossRef Mathias CB, Hobson SA, Garcia-Lloret M, Lawson G, Poddighe D, Freyschmidt EJ, et al. IgE-mediated systemic anaphylaxis and impaired tolerance to food antigens in mice with enhanced IL-4 receptor signaling. J Allergy Clin Immunol. 2011;127:795–805.PubMedPubMedCentralCrossRef
110.
go back to reference Han H, Thelen TD, Comeau MR, Ziegler SF. Thymic stromal lymphopoietin-mediated epicutaneous inflammation promotes acute diarrhea and anaphylaxis. J Clin Invest. 2014;124:5442–52.PubMedPubMedCentralCrossRef Han H, Thelen TD, Comeau MR, Ziegler SF. Thymic stromal lymphopoietin-mediated epicutaneous inflammation promotes acute diarrhea and anaphylaxis. J Clin Invest. 2014;124:5442–52.PubMedPubMedCentralCrossRef
111.
go back to reference Zabel BA, Nakae S, Zúñiga L, Kim JY, Ohyama T, Alt C, et al. Mast cell-expressed orphan receptor CCRL2 binds chemerin and is required for optimal induction of IgE-mediated passive cutaneous anaphylaxis. J Exp Med. 2008;205:2207–20.PubMedPubMedCentralCrossRef Zabel BA, Nakae S, Zúñiga L, Kim JY, Ohyama T, Alt C, et al. Mast cell-expressed orphan receptor CCRL2 binds chemerin and is required for optimal induction of IgE-mediated passive cutaneous anaphylaxis. J Exp Med. 2008;205:2207–20.PubMedPubMedCentralCrossRef
112.
go back to reference Birmingham N, Payankaulam S, Thanesvorakul S, Stefura B, HayGlass K, Gangur V. An ELISA-based method for measurement of food-specific IgE antibody in mouse serum: an alternative to the passive cutaneous anaphylaxis assay. J Immunol Methods. 2003;275:89–98.PubMedCrossRef Birmingham N, Payankaulam S, Thanesvorakul S, Stefura B, HayGlass K, Gangur V. An ELISA-based method for measurement of food-specific IgE antibody in mouse serum: an alternative to the passive cutaneous anaphylaxis assay. J Immunol Methods. 2003;275:89–98.PubMedCrossRef
113.
go back to reference Adel-Patient K, Bernard H, Ah-Leung S, Creminon C, Wal JM. Peanut- and cow’s milk-specific IgE, Th2 cells and local anaphylactic reaction are induced in Balb/c mice orally sensitized with cholera toxin. Allergy. 2005;60:658–64.PubMedCrossRef Adel-Patient K, Bernard H, Ah-Leung S, Creminon C, Wal JM. Peanut- and cow’s milk-specific IgE, Th2 cells and local anaphylactic reaction are induced in Balb/c mice orally sensitized with cholera toxin. Allergy. 2005;60:658–64.PubMedCrossRef
114.
go back to reference Lovik M, Hogseth AK, Gaarder PI, Hagemann R, Eide I. Diesel exhaust particles and carbon black have adjuvant activity on the local lymph node response and systemic IgE production to ovalbumin. Toxicology. 1997;121:165–78.PubMedCrossRef Lovik M, Hogseth AK, Gaarder PI, Hagemann R, Eide I. Diesel exhaust particles and carbon black have adjuvant activity on the local lymph node response and systemic IgE production to ovalbumin. Toxicology. 1997;121:165–78.PubMedCrossRef
115.
go back to reference Olzhausen J, Schawaller M, Wiki M, Akdis C, Jutel M, Crameri R, Rhyner C. Evanescent field-based fast measurements of allergen specific antibodies during SIT. Allergy. 2014;69(s99):72. Olzhausen J, Schawaller M, Wiki M, Akdis C, Jutel M, Crameri R, Rhyner C. Evanescent field-based fast measurements of allergen specific antibodies during SIT. Allergy. 2014;69(s99):72.
116.
go back to reference Granum B, Gaarder PI, Groeng E, Leikvold R, Namork E, Lovik M. Fine particles of widely different composition have an adjuvant effect on the production of allergen-specific antibodies. Toxicol Lett. 2001;118:171–81.PubMedCrossRef Granum B, Gaarder PI, Groeng E, Leikvold R, Namork E, Lovik M. Fine particles of widely different composition have an adjuvant effect on the production of allergen-specific antibodies. Toxicol Lett. 2001;118:171–81.PubMedCrossRef
117.
go back to reference Bøgh KL, Kroghsbo S, Dahl L, Rigby NM, Barkholt V, Mills EN, Madsen CB. Digested Ara h 1 has sensitizing capacity in Brown Norway rats. Clin Exp Allergy. 2009;39:1611–21.PubMedCrossRef Bøgh KL, Kroghsbo S, Dahl L, Rigby NM, Barkholt V, Mills EN, Madsen CB. Digested Ara h 1 has sensitizing capacity in Brown Norway rats. Clin Exp Allergy. 2009;39:1611–21.PubMedCrossRef
118.
go back to reference Vercelli D, Jabara HH, Arai K, Geha RS. Induction of human IgE synthesis requires interleukin 4 and T/B cell interactions involving the T cell receptor/CD3 complex and MHC class II antigens. J Exp Med. 1989;169:1295–307.PubMedCrossRef Vercelli D, Jabara HH, Arai K, Geha RS. Induction of human IgE synthesis requires interleukin 4 and T/B cell interactions involving the T cell receptor/CD3 complex and MHC class II antigens. J Exp Med. 1989;169:1295–307.PubMedCrossRef
119.
go back to reference Finkelman FD, Holmes J, Katona IM, Urban JF Jr, Beckmann MP, Park LS, et al. Lymphokine control of in vivo immunoglobulin isotype selection. Annu Rev Immunol. 1990;8:303–33.PubMedCrossRef Finkelman FD, Holmes J, Katona IM, Urban JF Jr, Beckmann MP, Park LS, et al. Lymphokine control of in vivo immunoglobulin isotype selection. Annu Rev Immunol. 1990;8:303–33.PubMedCrossRef
120.
go back to reference Nakajima-Adachi H, Kikuchi A, Fujimura Y, Shibahara K, Makino T, Goseki-Sone M, et al. Peyer’s patches and mesenteric lymph nodes cooperatively promote enteropathy in a mouse model of food allergy. PLoS One. 2014;9:e107492.PubMedPubMedCentralCrossRef Nakajima-Adachi H, Kikuchi A, Fujimura Y, Shibahara K, Makino T, Goseki-Sone M, et al. Peyer’s patches and mesenteric lymph nodes cooperatively promote enteropathy in a mouse model of food allergy. PLoS One. 2014;9:e107492.PubMedPubMedCentralCrossRef
121.
go back to reference Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, et al. Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases. J Allergy Clin Immunol. 2011;127:701–21.PubMedCrossRef Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, et al. Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases. J Allergy Clin Immunol. 2011;127:701–21.PubMedCrossRef
122.
go back to reference van Zijverden M, van der Pijl A, Bol M, van Pinxteren FA, de Haar C, Penninks AH, et al. Diesel exhaust, carbon black, and silica particles display distinct Th1/Th2 modulating activity. Toxicol Appl Pharmacol. 2000;168:131–9.PubMedCrossRef van Zijverden M, van der Pijl A, Bol M, van Pinxteren FA, de Haar C, Penninks AH, et al. Diesel exhaust, carbon black, and silica particles display distinct Th1/Th2 modulating activity. Toxicol Appl Pharmacol. 2000;168:131–9.PubMedCrossRef
123.
go back to reference Nygaard UC, Ormstad H, Aase A, Lovik M. The IgE adjuvant effect of particles: characterisation of the primary cellular response in the draining lymph node. Toxicology. 2005;206:181–93.PubMedCrossRef Nygaard UC, Ormstad H, Aase A, Lovik M. The IgE adjuvant effect of particles: characterisation of the primary cellular response in the draining lymph node. Toxicology. 2005;206:181–93.PubMedCrossRef
124.
go back to reference Gerberick GF, Ryan CA, Dearman RJ, Kimber I. Local lymph node assay (LLNA) for detection of sensitization capacity of chemicals. Methods. 2007;41:54–60.PubMedCrossRef Gerberick GF, Ryan CA, Dearman RJ, Kimber I. Local lymph node assay (LLNA) for detection of sensitization capacity of chemicals. Methods. 2007;41:54–60.PubMedCrossRef
125.
go back to reference Vaali K, Puumalainen TJ, Lehto M, Wolff H, Rita H, Alenius H, Palosuo T. Murine model of food allergy after epicutaneous sensitization: role of mucosal mast cell protease-1. Scand J Gastroenterol. 2006;41:1405–13.PubMedCrossRef Vaali K, Puumalainen TJ, Lehto M, Wolff H, Rita H, Alenius H, Palosuo T. Murine model of food allergy after epicutaneous sensitization: role of mucosal mast cell protease-1. Scand J Gastroenterol. 2006;41:1405–13.PubMedCrossRef
Metadata
Title
Current challenges facing the assessment of the allergenic capacity of food allergens in animal models
Authors
Katrine Lindholm Bøgh
Jolanda van Bilsen
Robert Głogowski
Iván López-Expósito
Grégory Bouchaud
Carine Blanchard
Marie Bodinier
Joost Smit
Raymond Pieters
Shanna Bastiaan-Net
Nicole de Wit
Eva Untersmayr
Karine Adel-Patient
Leon Knippels
Michelle M. Epstein
Mario Noti
Unni Cecilie Nygaard
Ian Kimber
Kitty Verhoeckx
Liam O’Mahony
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Clinical and Translational Allergy / Issue 1/2016
Electronic ISSN: 2045-7022
DOI
https://doi.org/10.1186/s13601-016-0110-2

Other articles of this Issue 1/2016

Clinical and Translational Allergy 1/2016 Go to the issue